1
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [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: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|
2
|
Fu E, Huang KF, Chang HH, Tseng HH, Yang SSD. Periodontitis Increases Gingival, Serum and Hippocampus β-Amyloid Expressions but Reduces Neurovascular Coupling in Basilar Artery of Rats. J Clin Periodontol 2025; 52:762-772. [PMID: 39780369 DOI: 10.1111/jcpe.14110] [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: 08/13/2024] [Revised: 11/19/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
AIM Neurodegenerative diseases are characterized by early increased beta-amyloid (Aβ) and decreased cerebrovascular reactivity. We investigated Aβ in gingiva, serum or hippocampus and neurovascular reactivity in basilar artery (BA) of periodontitis rats, to test the impact of Aβ on BA vasoreactivity ex vivo. MATERIALS AND METHODS Periodontitis was induced in 32 rats using silk-ligation. Rats were sacrificed at weeks 0, 1, 2 and 4. Gingival TNF-α, IL-1β and Aβ were quantified via immunoblotting. Alveolar bone destruction was examined by micro-computed tomography. Serum and hippocampus Aβ values were measured by enzyme-linked immunosorbent assay and fluorescence staining, respectively. Vasoreactivity was measured by myography on isolated BA. RESULTS From Week 1, gingival TNF-α and IL-1β and bone destruction increased. Gingiva, serum and hippocampus Aβ values increased from Week 2. Nicotine-induced BA relaxation declined from Week 2, while acetylcholine-induced relaxation decreased by Week 4. Bone loss correlated with Aβ and nicotine-induced relaxation. Correlations were observed between Aβs in tissues, between two induced BA relaxations and between Aβ expressions and the induced relaxations. Ex vivo, Aβ reduced nicotine- and isoproterenol-induced relaxations but not electrical stimulated relaxation. CONCLUSIONS Periodontitis may increase Aβ expressions and reduce BA neurovascular reactivity, with Aβ contributing to this abnormal neurovascular coupling.
Collapse
Affiliation(s)
- Earl Fu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Kuo-Feng Huang
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Hsi-Hsien Chang
- Division of Urology, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Medical Foundation, New Taipei, Taiwan, ROC
| | - Hui-Hwa Tseng
- Department of Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Stephen Shei-Dei Yang
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
- Division of Urology, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Medical Foundation, New Taipei, Taiwan, ROC
| |
Collapse
|
3
|
Badea A, Mahzarnia A, Reddy D, Dong Z, Anderson RJ, Moon HS, Stout JA, Williams J, Hirschler L, Barbier EL, Williams CL. Neuroimaging Biomarkers of Neuroprotection: Impact of Voluntary versus Enforced Exercise in Alzheimer's Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646015. [PMID: 40236037 PMCID: PMC11996325 DOI: 10.1101/2025.03.28.646015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Exercise is a promising strategy for preventing or delaying Alzheimer's disease (AD), yet its mechanisms remain unclear. We investigated how exercise influences brain structure, function, and behavior in a familial AD model. Mice underwent voluntary, voluntary plus enforced exercise, or remained sedentary. Neuroimaging included in vivo manganese-enhanced MRI (MEMRI). perfusion, and ex vivo diffusion MRI to assess morphometry, activity, cerebral blood flow (CBF), microstructural integrity and connectivity. Both exercise regimens induced structural and functional brain adaptations while reducing anhedonia. Voluntary exercise increased cortical and limbic volumes, particularly in the hippocampus, cingulate, and entorhinal cortex, supporting cognitive and emotional regulation. Adding enforced exercise influenced subcortical and sensory regions, including visual, motor and associative areas, supporting sensory-motor integration. MEMRI revealed increased activity in sensorimotor, limbic, and associative cortices, with voluntary exercise enhancing limbic and associative regions, and enforced exercise strengthening sensorimotor and subcortical circuits. White matter integrity improved in memory-associate pathways such as the corpus callosum, cingulum, and hippocampal commissure. Synaptic remodeling was observed in the cingulate cortex, anterior thalamic nuclei, and amygdala. Voluntary exercise enhanced CBF in the motor cortex and hippocampus, while enforced exercise limited these increases. Connectivity analyses revealed exercise-responsive networks spanning the cingulate cortex, entorhinal cortex, anterior thalamic nuclei, and basolateral amygdala, and associated tracts. Graph analyses linked running distance with increased thalamic, brainstem, and cerebellar connectivity, associating exercise intensity with plasticity. These findings highlight the ability of chronic exercise to modulate neuroimaging biomarkers through distinct but complementary pathways, reinforcing its potential as a neuroprotective intervention for AD. Highlights Exercise alters MRI biomarkers via distinct and partially overlapping mechanisms.Voluntary exercise boosts cortical and limbic regions for emotion and cognition.Enforced exercise strengthens subcortical and sensory areas for motor control.FA increases suggest memory tract reinforcement and grey matter remodelling.Graph analysis reveals plasticity in memory, emotion, and reward circuits.
Collapse
|
4
|
Jahng GH, Lee MB, Kwon OI. Gadolinium based contrast agent induced electrical conductivity heterogeneity analysis in the brain of Alzheimer's disease. Sci Rep 2025; 15:10832. [PMID: 40155644 PMCID: PMC11953297 DOI: 10.1038/s41598-025-92966-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Magnetic resonance imaging (MRI) often uses gadolinium-based contrast agents (GBCAs) to improve the characterization of imaging contrast, owing to their strong paramagnetic properties. Magnetic resonance electrical properties tomography (MREPT) visualizes the conductivity distribution of biological tissues at the Larmor frequency using the [Formula: see text] field phase signal. In this paper, we investigate the effect of GBCA on brain conductivity. To compare the differences of reconstructed noisy conductivity maps before and after the GBCA injection, we propose a method to remove the background low-frequency noise artifact based on an elliptic partial differential equation. By analyzing the relationship between electrical conductivity and magnetic permeability, the objective of this study is to develop a cost-effective and accessible initial screening imaging tool for diagnosing and monitoring the treatment of Alzheimer's disease (AD) pathophysiology. To investigate vascular damage in AD, we define a conductivity heterogeneity volume fraction (CHVF) caused by GBCA leakage. Using CHVF, we develop three indices to characterize mild cognitive impairment (MCI) and AD. To verify the proposed method, we studied a total of 42 participants, including 14 individuals diagnosed with AD, 18 participants with MCI, and 10 cognitively normal (CN) participants. Finally, we designed a radar chart informed by the CHVF analysis, to exhibit the pertinent parameters for MCI and AD patients, facilitating the evaluation and ongoing monitoring of each patient's diagnosis and treatment regimen.
Collapse
Affiliation(s)
- Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, 05278, Korea
| | - Mun Bae Lee
- Department of Mathematics, College of Basic Science, Konkuk University, Seoul, 05029, Korea
| | - Oh-In Kwon
- Department of Mathematics, College of Basic Science, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
5
|
Vandal M, Institoris A, Reveret L, Korin B, Gunn C, Hirai S, Jiang Y, Lee S, Lee J, Bourassa P, Mishra RC, Peringod G, Arellano F, Belzil C, Tremblay C, Hashem M, Gorzo K, Elias E, Yao J, Meilandt B, Foreman O, Roose-Girma M, Shin S, Muruve D, Nicola W, Körbelin J, Dunn JF, Chen W, Park SK, Braun AP, Bennett DA, Gordon GRJ, Calon F, Shaw AS, Nguyen MD. Loss of endothelial CD2AP causes sex-dependent cerebrovascular dysfunction. Neuron 2025; 113:876-895.e11. [PMID: 39892386 DOI: 10.1016/j.neuron.2025.01.006] [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: 09/06/2022] [Revised: 08/27/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025]
Abstract
Polymorphisms in CD2-associated protein (CD2AP) predispose to Alzheimer's disease (AD), but the underlying mechanisms remain unknown. Here, we show that loss of CD2AP in cerebral blood vessels is associated with cognitive decline in AD subjects and that genetic downregulation of CD2AP in brain vascular endothelial cells impairs memory function in male mice. Animals with reduced brain endothelial CD2AP display altered blood flow regulation at rest and during neurovascular coupling, defects in mural cell activity, and an abnormal vascular sex-dependent response to Aβ. Antagonizing endothelin-1 receptor A signaling partly rescues the vascular impairments, but only in male mice. Treatment of CD2AP mutant mice with reelin glycoprotein that mitigates the effects of CD2AP loss function via ApoER2 increases resting cerebral blood flow and even protects male mice against the noxious effect of Aβ. Thus, endothelial CD2AP plays critical roles in cerebrovascular functions and represents a novel target for sex-specific treatment in AD.
Collapse
Affiliation(s)
- Milène Vandal
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Adam Institoris
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Louise Reveret
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Ben Korin
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Colin Gunn
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Sotaro Hirai
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Sukyoung Lee
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Jiyeon Lee
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Philippe Bourassa
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Ramesh C Mishra
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Govind Peringod
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Faye Arellano
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Camille Belzil
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Cyntia Tremblay
- Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Mada Hashem
- Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Kelsea Gorzo
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Esteban Elias
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Bill Meilandt
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Oded Foreman
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Meron Roose-Girma
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Steven Shin
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Daniel Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Wilten Nicola
- Departments of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Jeff F Dunn
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada; Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Wayne Chen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Sang-Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Andrew P Braun
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - David A Bennett
- Rush Alzheimer's disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Grant R J Gordon
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada.
| | - Andrey S Shaw
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA.
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada.
| |
Collapse
|
6
|
Fang X, Border JJ, Zhang H, Challagundla L, Kaur J, Hwang SH, Hammock BD, Fan F, Roman RJ. A Soluble Epoxide Hydrolase Inhibitor Improves Cerebrovascular Dysfunction, Neuroinflammation, Amyloid Burden, and Cognitive Impairments in the hAPP/PS1 TgF344-AD Rat Model of Alzheimer's Disease. Int J Mol Sci 2025; 26:2433. [PMID: 40141075 PMCID: PMC11942141 DOI: 10.3390/ijms26062433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is an increasing global healthcare crisis with few effective treatments. The accumulation of amyloid plaques and hyper-phosphorylated tau are thought to underlie the pathogenesis of AD. However, current studies have recognized a prominent role of cerebrovascular dysfunction in AD. We recently reported that SNPs in soluble epoxide hydrolase (sEH) are linked to AD in human genetic studies and that long-term administration of an sEH inhibitor attenuated cerebral vascular and cognitive dysfunction in a rat model of AD. However, the mechanisms linking changes in cerebral vascular function and neuroprotective actions of sEH inhibitors in AD remain to be determined. This study investigated the effects of administration of an sEH inhibitor, 1-(1-Propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea (TPPU), on neurovascular coupling, blood-brain barrier (BBB) function, neuroinflammation, and cognitive dysfunction in an hAPP/PS1 TgF344-AD rat model of AD. We observed predominant β-amyloid accumulation in the brains of 9-10-month-old AD rats and that TPPU treatment for three months reduced amyloid burden. The functional hyperemic response to whisker stimulation was attenuated in AD rats, and TPPU normalized the response. The sEH inhibitor, TPPU, mitigated capillary rarefaction, BBB leakage, and activation of astrocytes and microglia in AD rats. TPPU increased the expression of pre- and post-synaptic proteins and reduced loss of hippocampal neurons and cognitive impairments in the AD rats, which was confirmed in a transcriptome and GO analysis. These results suggest that sEH inhibitors could be a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Lavanya Challagundla
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Jasleen Kaur
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Sung Hee Hwang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Bruce D. Hammock
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Fan Fan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| |
Collapse
|
7
|
Bohannon DG, Long D, Okhravi HR, Lee SC, De Jesus CL, Neubert TA, Rostagno AA, Ghiso JA, Kim W. Functionally distinct pericyte subsets differently regulate amyloid-β deposition in patients with Alzheimer's disease. Brain Pathol 2025; 35:e13282. [PMID: 38932696 PMCID: PMC11835444 DOI: 10.1111/bpa.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Although the concept that the blood-brain barrier (BBB) plays an important role in the etiology and pathogenesis of Alzheimer's disease (AD) has become increasingly accepted, little is known yet about how it actually contributes. We and others have recently identified a novel functionally distinct subset of BBB pericytes (PCs). In the present study, we sought to determine whether these PC subsets differentially contribute to AD-associated pathologies by immunohistochemistry and amyloid beta (Aβ) peptidomics. We demonstrated that a disease-associated PC subset (PC2) expanded in AD patients compared to age-matched, cognitively unimpaired controls. Surprisingly, we found that this increase in the percentage of PC2 (%PC2) was correlated negatively with BBB breakdown in AD patients, unlike in natural aging or other reported disease conditions. The higher %PC2 in AD patients was also correlated with a lower Aβ42 plaque load and a lower Aβ42:Aβ40 ratio in the brain as determined by immunohistochemistry. Colocalization analysis of multicolor confocal immunofluorescence microscopy images suggests that AD patient with low %PC2 have higher BBB breakdown due to internalization of Aβ42 by the physiologically normal PC subset (PC1) and their concomitant cell death leading to more vessels without PCs and increased plaque load. On the contrary, it appears that PC2 can secrete cathepsin D to cleave and degrade Aβ built up outside of PC2 into more soluble forms, ultimately contributing to less BBB breakdown and reducing Aβ plaque load. Collectively our data shows functionally distinct mechanisms for PC1 and PC2 in high Aβ conditions, demonstrating the importance of correctly identifying these populations when investigating the contribution of neurovascular dysfunction to AD pathogenesis.
Collapse
Affiliation(s)
- Diana G. Bohannon
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Danielle Long
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Hamid R. Okhravi
- Glennan Center for Geriatrics and GerontologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Integrated Neurodegenerative Disorders CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | | | | | - Thomas A. Neubert
- Department of Neuroscience and PhysiologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Agueda A. Rostagno
- Department of PathologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Jorge A. Ghiso
- Department of PathologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
- Department of PsychiatryNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Woong‐Ki Kim
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Integrated Neurodegenerative Disorders CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Division of MicrobiologyTulane National Primate Research CenterCovingtonLouisianaUSA
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
8
|
Tang C, Border JJ, Zhang H, Gregory A, Bai S, Fang X, Liu Y, Wang S, Hwang SH, Gao W, Morgan GC, Smith J, Bunn D, Cantwell C, Wagner KM, Morisseau C, Yang J, Shin SM, O'Herron P, Bagi Z, Filosa JA, Dong Y, Yu H, Hammock BD, Roman RJ, Fan F. Inhibition of soluble epoxide hydrolase ameliorates cerebral blood flow autoregulation and cognition in alzheimer's disease and diabetes-related dementia rat models. GeroScience 2025:10.1007/s11357-025-01550-8. [PMID: 39903369 DOI: 10.1007/s11357-025-01550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
Alzheimer's Disease and Alzheimer's Disease-related dementias (AD/ADRD) pose major global healthcare challenges, with diabetes mellitus (DM) being a key risk factor. Both AD and DM-related ADRD are characterized by reduced cerebral blood flow, although the exact mechanisms remain unclear. We previously identified compromised cerebral hemodynamics as early signs in TgF344-AD and type 2 DM-ADRD (T2DN) rat models. Genome-wide studies have linked AD/ADRD to SNPs in soluble epoxide hydrolase (sEH). This study explored the effects of sEH inhibition with TPPU on cerebral vascular function and cognition in AD and DM-ADRD models. Chronic TPPU treatment improved cognition in both AD and DM-ADRD rats without affecting body weight. In DM-ADRD rats, TPPU reduced plasma glucose and HbA1c levels. Transcriptomic analysis of primary cerebral vascular smooth muscle cells from AD rats treated with TPPU revealed enhanced pathways related to cell contraction, alongside decreased oxidative stress and inflammation. Both AD and DM-ADRD rats exhibited impaired myogenic responses and autoregulation in the cerebral circulation, which were normalized with chronic sEH inhibition. Additionally, TPPU improved acetylcholine-induced vasodilation in the middle cerebral arteries (MCA) of DM-ADRD rats. Acute TPPU administration unexpectedly caused vasoconstriction in the MCA of DM-ADRD rats at lower doses. In contrast, higher doses or longer durations were required to induce effective vasodilation at physiological perfusion pressure in both control and ADRD rats. Additionally, TPPU decreased reactive oxygen species production in cerebral vessels of AD and DM-ADRD rats. These findings provide novel evidence that chronic sEH inhibition can reverse cerebrovascular dysfunction and cognitive impairments in AD/ADRD, offering a promising avenue for therapeutic development.
Collapse
Affiliation(s)
- Chengyun Tang
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jane J Border
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Huawei Zhang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Andrew Gregory
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Xing Fang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yedan Liu
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sung Hee Hwang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Wenjun Gao
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gilbert C Morgan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jhania Smith
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David Bunn
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Cameron Cantwell
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Karen M Wagner
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Christophe Morisseau
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Jun Yang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Seung Min Shin
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Philip O'Herron
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zsolt Bagi
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jessica A Filosa
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yanbin Dong
- Georgia Prevention Center, Augusta University, Augusta, GA, USA
| | - Hongwei Yu
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bruce D Hammock
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Richard J Roman
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Fan Fan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
9
|
Wang J, Meng X, Yang J, Tang Y, Zeng F, Wang Y, Chen Z, Chen D, Zou R, Liu W. Improvements in Exercise for Alzheimer's Disease: Highlighting FGF21-Induced Cerebrovascular Protection. Neurochem Res 2025; 50:95. [PMID: 39903342 DOI: 10.1007/s11064-025-04350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Currently, it has shown a trend of earlier onset, with most patients experiencing a progressive decline in cognitive function following the disease's onset, which places a heavy burden on society and family. Since no drug cure for AD exists, exploring new ways for its treatment and prevention has become critical. Early vascular damage is an initial trigger for neuronal injury in AD, underscoring the importance of vascular health in the early stages of the disease. Patients with early AD experience abnormal blood-brain barrier transport of amyloid-β (Aβ) peptides, with excess Aβ being deposited in the cerebral vasculature. The toxic effects of Aβ lead to abnormalities in cerebrovascular structure and function. Fibroblast growth factor21 (FGF21) is an endocrine factor that positively regulates energy homeostasis and glucose-lipid metabolism. Notably, it is one of the effective targets for metabolic disease prevention and treatment. Recent studies have found that FGF21 has anti-aging and vasoprotective effects, with receptors for FGF21 present in the brain. Exercise stimulates the liver to produce large amounts of FGF21, which enters the blood-brain barrier with the blood to exert neurovascular protection. Therefore, we review the biological properties of FGF21, its role in the cerebrovascular structure and function in AD, and the mechanism of exercise-regulated FGF21 action on AD-related cerebrovascular changes, aiming to provide a new theoretical basis for using exercise to ameliorate degenerative neurological diseases.
Collapse
Affiliation(s)
- Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Jialun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Yingzhe Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Dandan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Hunan Normal University, Changsha, Hunan Province, 410081, China.
- Physical Education College, Yuelu District, Hunan Normal University, Changsha, Hunan Province, 410081, China.
| |
Collapse
|
10
|
Kim TA, Cruz G, Syty MD, Wang F, Wang X, Duan A, Halterman M, Xiong Q, Palop JJ, Ge S. Neural circuit mechanisms underlying aberrantly prolonged functional hyperemia in young Alzheimer's disease mice. Mol Psychiatry 2025; 30:367-378. [PMID: 39043843 PMCID: PMC11750623 DOI: 10.1038/s41380-024-02680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
Neurovascular defects are one of the most common alterations in Alzheimer's disease (AD) pathogenesis, but whether these deficits develop before the onset of amyloid beta (Aβ) accumulation remains to be determined. Using in vivo optical imaging in freely moving mice, we explored activity-induced hippocampal microvascular blood flow dynamics in AppSAA knock-in and J20 mouse models of AD at early stages of disease progression. We found that prior to the onset of Aβ accumulation, there was a pathologically elevated blood flow response to context exploration, termed functional hyperemia. After the onset of Aβ accumulation, this context exploration-induced hyperemia declined rapidly relative to that in control mice. Using in vivo electrophysiology recordings to explore the neural circuit mechanism underlying this blood flow alteration, we found that hippocampal interneurons before the onset of Aβ accumulation were hyperactive during context exploration. Chemogenetic tests suggest that hyperactive activation of inhibitory neurons accounted for the elevated functional hyperemia. The suppression of nitric oxide (NO) produced from hippocampal interneurons in young AD mice decreased the accumulation of Aβ. Together, these findings reveal that neurovascular coupling is aberrantly elevated before Aβ deposition, and this hyperactive functional hyperemia declines rapidly upon Aβ accumulation.
Collapse
Affiliation(s)
- Thomas A Kim
- Medical Scientist Training Program (MSTP), Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794, USA
| | - George Cruz
- Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michelle D Syty
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Faye Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Xinxing Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Alexandra Duan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Marc Halterman
- Department of Neurology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
11
|
Bai X, Song L, Liu X, Yang W, Li M, Xu B, Yang Z, Wang Z, Wang H. Cognitive Impairment in Chronic Kidney Disease Across Different Stages: The Role of Structural and Perfusion-Driven Functional Connectivity Changes. Brain Behav 2025; 15:e70330. [PMID: 39957426 PMCID: PMC11830999 DOI: 10.1002/brb3.70330] [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: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
INTRODUCTION Chronic kidney disease (CKD) is associated with cognitive impairment (CI), yet the exact pathophysiological mechanisms remain unclear. This study aims to investigate the alterations in gray matter volume (GMV) and cerebral blood flow (CBF) across CKD stages, identify co-changed brain regions, explore abnormal seed-based functional connectivity (FC) in patients with CKD, and investigate the correlation between the abnormal brain regions and neuropsychological test scores. METHODS Two hundred and eight participants (66 healthy controls, 70 CKD Stages 1-3a, and 72 CKD Stages 3b-5) were consecutively recruited and underwent high-resolution T1-weighted imaging, arterial spin labeling, and functional MR imaging. The imaging parameters were compared among three groups, and correlations with MoCA scores were analyzed. RESULTS Compared to CKD 1-3a group, the bilateral fusiform gyrus (FFG.L and FFG.R) exhibited reduced GMV, increased CBF, and decreased FFG.L-FC with bilateral inferior frontal gyrus, triangular part (IFGtriang.L and IFGtriang.R), left middle occipital gyrus (MOG.L), and left hippocampus (HIP.L), as well as decreased FFG.R-FC with bilateral median cingulate and paracingulate gyri (DCG.L and DCG.R), left superior frontal gyrus, medial (SFGmed.L), IFGtriang.L, and right middle temporal gyrus (MTG.R) in CKD 3b-5 group. A negative correlation was observed between the MoCA scores and FFG.L-FC with right middle frontal gyrus (MFG.R), IFGtriang.L, IFGtriang.R, HIP.L, and left putamen in patients with CKD 1-3a. CONCLUSION Brain structural and perfusion alterations may underlie the reduced FC between fusiform gyrus and cognitive-related regions, providing potential neuroimaging evidence for the neuropathological mechanisms of CI in patients with different stages CKD.
Collapse
Affiliation(s)
- Xiaoyan Bai
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Lijun Song
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Xu Liu
- Department of NephrologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Wenbo Yang
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Mingan Li
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Boyan Xu
- MR ResearchGE HealthcareBeijingChina
| | - Zhenghan Yang
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Zhen‐Chang Wang
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Hao Wang
- Department of RadiologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
12
|
Zhang T, Yang J, Xu H, Cao Y, Du X, Guo L, Liang B, Su L, Chai L, Yuan Q, Hu L. Acorus tatarinowii alleviates D-galactose-induced Alzheimer's-like disease cognitive impairment and Aβ-induced pericytes dysfunction in mice. Brain Res 2025; 1847:149312. [PMID: 39515743 DOI: 10.1016/j.brainres.2024.149312] [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: 06/27/2024] [Revised: 09/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Pericytes regulate cerebral blood flow (CBF) and excess amyloid in the brain. Pericyte dysfunction may contribute to the pathology of Alzheimer's disease (AD). Acorus tatarinowii (AT), a Chinese medicine commonly used to treat AD, protects the central nervous system. However, whether AT can regulate pericyte function and ameliorate cognitive dysfunction remains unclear. We employed a novel target recognition assay, quantitative measurement of CBF, hematoxylin and eosin staining, immunofluorescence staining, and Western blot to investigate the role of AT in improving cognitive function in patients with AD. Additionally, we investigated the therapeutic potential of β-Asarone, the primary active compound in AT, for treating AD by modulating pericyte function using transmission electron microscopy, silver staining, electrical impedance, and other methodologies. The results revealed that administration of AT effectively alleviated the cognitive impairments induced by D-galactose in mice, as evidenced by enhanced CBF, improved histological characteristics of damaged brain tissue cells, increased expression of platelet-derived growth factor-β (PDGF-β), decreased Aβ accumulation via enhanced lipoprotein receptor-related protein 1 (LRP1), and reduced beta-site APP-cleaving enzyme 1 (BACE1). β-Asarone treatment mitigated ROS release and BACE1 expression while elevating the cell index in Aβ1-40 injured mouse brain vascular pericytes (MBVP). These findings suggest that AT has the potential to enhance CBF and mitigate pericellular dysfunction, thereby ameliorating Aβ deposition in the brain and improving cognitive impairment in patients with AD.
Collapse
Affiliation(s)
- Tong Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Juan Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haiying Xu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yushuang Cao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyuan Du
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lichen Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bing Liang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Linlin Su
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lijuan Chai
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qing Yuan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Limin Hu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
13
|
Jones RM, DeRuiter RM, Deshmukh M, Dayton PA, Pinton GF. Non-invasive volumetric ultrasound localization microscopy detects vascular changes in mice with Alzheimer's disease. Theranostics 2025; 15:1110-1121. [PMID: 39776806 PMCID: PMC11700853 DOI: 10.7150/thno.99097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's Disease (AD) is the most common form of dementia and one of the leading causes of death. AD is known to be correlated to tortuosity in the microvasculature as well as decreases in blood flow throughout the brain. However, the mechanisms behind these changes and their causal relation to AD are poorly understood. Methods: Here, we use volumetric ultrasound localization microscopy (ULM) to non-invasively and quantitatively compare the microvascular morphology and flow dynamics of five wildtype (WT) and five APPNL-G-F Knock-in mice, a mouse model of AD, across a 1cmx1cmx1cm brain volume and in four specific brain regions: the hippocampal formation, thalamus, hypothalamus, and cerebral cortex. Results: Comparisons between groups showed a significant increase in tortuosity, as measured by the Sum of Angles Metric (SOAM), throughout the brain (p < 0.01) and the hypothalamus (p = 0.01), in mice with AD. While differences in mean velocity (p < 0.01) and blood flow (p=0.04) were detected across the whole brain, their effect size was small and no differences were detected in the four selected regions. There was a significant decrease in the linear log relationship between vessel diameter and blood flow, with AD mice experiencing a lower slope than WT mice across the whole brain volume (p = 0.02) and in the hippocampal formation (p = 0.05), a region affected by Amyloid Beta plaques in this mouse model. The AD mice had higher blood flows in smaller vessels and smaller blood flows in larger vessels than the WT mice. Conclusions: This preliminary demonstrates that the imaging technique can be used for non-invasive, longitudinal, volumetric assessment of AD, which may allow for investigation into the poorly understood microvascular degeneration associated with AD through time as well as the development of early diagnostic techniques.
Collapse
Affiliation(s)
- Rebecca M. Jones
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27599, USA
| | - Ryan M. DeRuiter
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27599, USA
| | - Mohanish Deshmukh
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Paul A. Dayton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27599, USA
| | - Gianmarco F. Pinton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27599, USA
| |
Collapse
|
14
|
Evans LE, Gray AL, Walsh KR, Danby TGE, Pritchard HAT, Allan SM, Gurney AM, Greenstein AS, Schiessl I. Combining In Vivo Two-Photon and Laser Speckle Microscopy With the Ex Vivo Capillary-Parenchymal Arteriole Preparation as a Novel Approach to Study Neurovascular Coupling. Microcirculation 2025; 32:e70001. [PMID: 39777814 PMCID: PMC11706670 DOI: 10.1111/micc.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE Cerebral blood flow (CBF) decline is increasingly recognized as an area of importance for targeting neurodegenerative disorders, yet full understanding of the mechanisms that underlie CBF changes are lacking. Animal models are crucial for expanding our knowledge as methods for studying global CBF and neurovascular coupling in humans are limited and require expensive specialized scanners. METHODS Use of appropriate animal models can increase our understanding of cerebrovascular function, so we have combined chronic cranial windows with in vivo two-photon and laser speckle microscopy and ex vivo capillary-parenchymal arteriole (CaPA) preparations. Chronic cranial windows allow for longitudinal direct observation of the cerebral microvasculature and surrounding parenchyma while the CaPA preparation can assess capillary and arteriole function in isolation of the neuronal tissue. RESULTS Here, we found that extra-dural cranial windows and related imaging protocols do not affect vascular function in the CaPA preparation. Cortical vessels from animals that have undergone imaging can therefore be taken to discover physiological alterations in the cerebral vasculature that contribute to any observed in vivo changes. CONCLUSION This approach will enhance neurodegenerative research with the benefit of limiting animal usage.
Collapse
Affiliation(s)
- Lowri E. Evans
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Anna L. Gray
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Wellcome Centre for Cell‐Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Katy R. Walsh
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Thea G. E. Danby
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Harry A. T. Pritchard
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Alison M. Gurney
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Adam S. Greenstein
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
15
|
Osborne OM, Daftari M, Naranjo O, Johar AN, Brooks S, Colbert BM, Torices S, Lewis E, Sendaydiego J, Drexler G, Bashti M, Margetts AV, Tuesta LM, Mason C, Bilbao D, Vontell R, Griswold AJ, Dykxhoorn DM, Toborek M. Post-stroke hippocampal neurogenesis is impaired by microvascular dysfunction and PI3K signaling in cerebral amyloid angiopathy. Cell Rep 2024; 43:114848. [PMID: 39392753 PMCID: PMC11562893 DOI: 10.1016/j.celrep.2024.114848] [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: 04/30/2024] [Revised: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
Ischemic stroke and cerebral amyloid angiopathy (CAA) pose significant challenges in an aging population, particularly in post-stroke recovery. Using the 5xFAD mouse model, we explore the relationship between CAA, ischemic stroke, and tissue recovery. We hypothesize that amyloid-beta accumulation worsens stroke outcomes by inducing blood-brain barrier (BBB) dysfunction, leading to impaired neurogenesis. Our findings show that CAA exacerbates stroke outcomes, with mice exhibiting constricted BBB microvessels, reduced cerebral blood flow, and impaired tissue recovery. Transcriptional analysis shows that endothelial cells and neural progenitor cells (NPCs) in the hippocampus exhibit differential gene expression in response to CAA and stroke, specifically targeting the phosphatidylinositol 3-kinase (PI3K) pathway. In vitro experiments with human NPCs validate these findings, showing that disruption of the CXCL12-PIK3C2A-CREB3L2 axis impairs neurogenesis. Notably, PI3K pathway activation restores neurogenesis, highlighting a potential therapeutic approach. These results suggest that CAA combined with stroke induces microvascular dysfunction and aberrant neurogenesis through this specific pathway.
Collapse
Affiliation(s)
- Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Manav Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adarsh N Johar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Samantha Brooks
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brett M Colbert
- Medical Scientist Training Program, 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
| | - Elizabeth Lewis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jet Sendaydiego
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gillian Drexler
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Malek Bashti
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander V Margetts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Luis M Tuesta
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurology and Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Regina Vontell
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anthony J Griswold
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek M Dykxhoorn
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, 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.
| |
Collapse
|
16
|
Rodriguez Moore G, Melo-Escobar I, Stegner D, Bracko O. One immune cell to bind them all: platelet contribution to neurodegenerative disease. Mol Neurodegener 2024; 19:65. [PMID: 39334369 PMCID: PMC11438031 DOI: 10.1186/s13024-024-00754-4] [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: 03/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease (AD) and related dementias (ADRD) collectively affect a significant portion of the aging population worldwide. The pathological progression of AD involves not only the classical hallmarks of amyloid beta (Aβ) plaque buildup and neurofibrillary tangle development but also the effects of vasculature and chronic inflammatory processes. Recently, platelets have emerged as central players in systemic and neuroinflammation. Studies have shown that patients with altered platelet receptor expression exhibit accelerated cognitive decline independent of traditional risk factors. Additionally, platelets from AD patients exhibit heightened unstimulated activation compared to control groups. Platelet granules contain crucial AD-related proteins like tau and amyloid precursor protein (APP). Dysregulation of platelet exocytosis contributes to disease phenotypes characterized by increased bleeding, stroke, and cognitive decline risk. Recent studies have indicated that these effects are not associated with the quantity of platelets present in circulation. This underscores the hypothesis that disruptions in platelet-mediated inflammation and healing processes may play a crucial role in the development of ADRD. A thorough look at platelets, encompassing their receptors, secreted molecules, and diverse roles in inflammatory interactions with other cells in the circulatory system in AD and ADRD, holds promising prospects for disease management and intervention. This review discusses the pivotal roles of platelets in ADRD.
Collapse
Affiliation(s)
| | - Isabel Melo-Escobar
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
17
|
Magrì A, Tomasello B, Naletova I, Tabbì G, Cairns WRL, Greco V, Sciuto S, La Mendola D, Rizzarelli E. New BDNF and NT-3 Cyclic Mimetics Concur with Copper to Activate Trophic Signaling Pathways as Potential Molecular Entities to Protect Old Brains from Neurodegeneration. Biomolecules 2024; 14:1104. [PMID: 39334869 PMCID: PMC11430436 DOI: 10.3390/biom14091104] [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: 08/05/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
A low level of Neurotrophins (NTs), their Tyrosine Kinase Receptors (Trks), Vascular Endothelial Growth Factors (VEGFs) and their receptors, mainly VEGFR1 and VEGFR2, characterizes AD brains. The use of NTs and VEGFs as drugs presents different issues due to their low permeability of the blood-brain barrier, the poor pharmacokinetic profile, and the relevant side effects. To overcome these issues, different functional and structural NT mimics have been employed. Being aware that the N-terminus domain as the key domain of NTs for the binding selectivity and activation of Trks and the need to avoid or delay proteolysis, we herein report on the mimicking ability of two cyclic peptide encompassing the N-terminus of Brain Derived Growth Factor (BDNF), (c-[HSDPARRGELSV-]), cBDNF(1-12) and of Neurotrophin3 (NT3), (c-[YAEHKSHRGEYSV-]), cNT3(1-13). The two cyclic peptide features were characterized by a combined thermodynamic and spectroscopic approach (potentiometry, NMR, UV-vis and CD) that was extended to their copper(II) ion complexes. SH-SY5Y cell assays show that the Cu2+ present at the sub-micromolar level in the complete culture media affects the treatments with the two peptides. cBDNF(1-12) and cNT3(1-13) act as ionophores, induce neuronal differentiation and promote Trks and CREB phosphorylation in a copper dependent manner. Consistently, both peptide and Cu2+ stimulate BDNF and VEGF expression as well as VEGF release; cBDNF(1-12) and cNT3(1-13) induce the expression of Trks and VEGFRs.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Giovanni Tabbì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Warren R. L. Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), 155 Via Torino, 30172 Venice, Italy;
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
18
|
Aamand R, Rasmussen PM, Andersen KS, de Paoli S, Weitzberg E, Christiansen M, Lund TE, Østergaard L. Cerebral microvascular changes in healthy carriers of the APOE-ɛ4 Alzheimer's disease risk gene. PNAS NEXUS 2024; 3:pgae369. [PMID: 39253395 PMCID: PMC11382292 DOI: 10.1093/pnasnexus/pgae369] [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: 05/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
APOE-ɛ4 is a genetic risk factor for Alzheimer's disease (AD). AD is associated with reduced cerebral blood flow (CBF) and with microvascular changes that limit the transport of oxygen from blood into brain tissue: reduced microvascular cerebral blood volume and high relative transit time heterogeneity (RTH). Healthy APOE-ɛ4 carriers reveal brain regions with elevated CBF compared with carriers of the common ɛ3 allele. Such asymptomatic hyperemia may reflect microvascular dysfunction: a vascular disease entity characterized by suboptimal tissue oxygen uptake, rather than limited blood flow per se. Here, we used perfusion MRI to show that elevated regional CBF is accompanied by reduced capillary blood volume in healthy APOE-ɛ4 carriers (carriers) aged 30-70 years compared with similarly aged APOE-ɛ3 carriers (noncarriers). Younger carriers have elevated hippocampal RTH and more extreme RTH values throughout both white matter (WM) and cortical gray matter (GM) compared with noncarriers. Older carriers have reduced WM CBF and more extreme GM RTH values than noncarriers. Across all groups, lower WM and hippocampal RTH correlate with higher educational attainment, which is associated with lower AD risk. Three days of dietary nitrate supplementation increased carriers' WM CBF but caused older carriers to score worse on two of six aggregate neuropsychological scores. The intervention improved late recall in younger carriers and in noncarriers. The APOE-ɛ4 gene is associated with microvascular changes that may impair tissue oxygen extraction. We speculate that vascular risk factor control is particularly important for APOE-ɛ4 carriers' healthy aging.
Collapse
Affiliation(s)
- Rasmus Aamand
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Peter M Rasmussen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Katrine Schilling Andersen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Stine de Paoli
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Torben E Lund
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
- Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
19
|
Tang C, Border JJ, Zhang H, Gregory A, Bai S, Fang X, Liu Y, Wang S, Hwang SH, Gao W, Morgan GC, Smith J, Bunn D, Cantwell C, Wagner KM, Morisseau C, Yang J, Shin SM, O’Herron P, Bagi Z, Filosa JA, Dong Y, Yu H, Hammock BD, Roman RJ, Fan F. Inhibition of Soluble Epoxide Hydrolase Ameliorates Cerebral Blood Flow Autoregulation and Cognition in Alzheimer's Disease and Diabetes-Related Dementia Rat Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610540. [PMID: 39257786 PMCID: PMC11383657 DOI: 10.1101/2024.08.30.610540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Alzheimer's Disease and Alzheimer's Disease-related dementias (AD/ADRD) pose major global healthcare challenges, with diabetes mellitus (DM) being a key risk factor. Both AD and DM-related ADRD are characterized by reduced cerebral blood flow, although the exact mechanisms remain unclear. We previously identified compromised cerebral hemodynamics as early signs in TgF344-AD and type 2 DM-ADRD (T2DN) rat models. Genome-wide studies have linked AD/ADRD to SNPs in soluble epoxide hydrolase (sEH). This study explored the effects of sEH inhibition with TPPU on cerebral vascular function and cognition in AD and DM-ADRD models. Chronic TPPU treatment improved cognition in both AD and DM-ADRD rats without affecting body weight. In DM-ADRD rats, TPPU reduced plasma glucose and HbA1C levels. Transcriptomic analysis of primary cerebral vascular smooth muscle cells from AD rats treated with TPPU revealed enhanced pathways related to cell contraction, alongside decreased oxidative stress and inflammation. Both AD and DM-ADRD rats exhibited impaired myogenic responses and autoregulation in the cerebral circulation, which were normalized with chronic sEH inhibition. Additionally, TPPU improved acetylcholine-induced vasodilation in the middle cerebral arteries (MCA) of DM-ADRD rats. Acute TPPU administration unexpectedly caused vasoconstriction in the MCA of DM-ADRD rats at lower doses. In contrast, higher doses or longer durations were required to induce effective vasodilation at physiological perfusion pressure in both control and ADRD rats. Additionally, TPPU decreased reactive oxygen species production in cerebral vessels of AD and DM-ADRD rats. These findings provide novel evidence that chronic sEH inhibition can reverse cerebrovascular dysfunction and cognitive impairments in AD/ADRD, offering a promising avenue for therapeutic development.
Collapse
Affiliation(s)
- Chengyun Tang
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Jane J. Border
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Huawei Zhang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Andrew Gregory
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Xing Fang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Yedan Liu
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Shaoxun Wang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Sung Hee Hwang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA
| | - Wenjun Gao
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Gilbert C. Morgan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jhania Smith
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - David Bunn
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Cameron Cantwell
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Karen M. Wagner
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA
| | - Christophe Morisseau
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA
| | - Jun Yang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA
| | - Seung Min Shin
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Philip O’Herron
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Zsolt Bagi
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jessica A. Filosa
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Yanbin Dong
- Georgia Prevention Center, Augusta University, Augusta, GA
| | - Hongwei Yu
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Bruce D. Hammock
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA
| | - Richard J. Roman
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Fan Fan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
20
|
Bagheri M, Habibzadeh S, Moeini M. Transient Changes in Cerebral Tissue Oxygen, Glucose, and Temperature by Microstrokes: A Computational Study. Microcirculation 2024; 31:e12872. [PMID: 38944839 DOI: 10.1111/micc.12872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/09/2024] [Accepted: 06/08/2024] [Indexed: 07/02/2024]
Abstract
OBJECTIVE This study focuses on evaluating the disruptions in key physiological parameters during microstroke events to assess their severity. METHODS A mathematical model was developed to simulate the changes in cerebral tissue pO2, glucose concentration, and temperature due to blood flow interruptions. The model considers variations in baseline cerebral blood flow (CBF), capillary density, and blood oxygen/glucose levels, as well as ambient temperature changes. RESULTS Simulations indicate that complete blood flow obstruction still allows for limited glucose availability, supporting nonoxidative metabolism and potentially exacerbating lactate buildup and acidosis. Partial obstructions decrease tissue pO2, with minimal impact on glucose level, which can remain almost unchanged or even slightly increase. Reduced CBF, capillary density, or blood oxygen due to aging or disease enhances hypoxia risk at lower obstruction levels, with capillary density having a significant effect on stroke severity by influencing both pO2 and glucose levels. Conditions could lead to co-occurrence of hypoxia/hypoglycemia or hypoxia/hyperglycemia, each worsening outcomes. Temperature effects were minimal in deep brain regions but varied near the skull by 0.2-0.8°C depending on ambient temperature. CONCLUSIONS The model provides insights into the conditions driving severe stroke outcomes based on estimated levels of hypoxia, hypoglycemia, hyperglycemia, and temperature changes.
Collapse
Affiliation(s)
- Marzieh Bagheri
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Sajjad Habibzadeh
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mohammad Moeini
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
21
|
Shirzadi Z, Boyle R, Yau WYW, Coughlan G, Fu JF, Properzi MJ, Buckley RF, Yang HS, Scanlon CE, Hsieh S, Amariglio RE, Papp K, Rentz D, Price JC, Johnson KA, Sperling RA, Chhatwal JP, Schultz AP. Vascular contributions to cognitive decline: Beyond amyloid and tau in the Harvard Aging Brain Study. J Cereb Blood Flow Metab 2024; 44:1319-1328. [PMID: 38452039 PMCID: PMC11342726 DOI: 10.1177/0271678x241237624] [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: 08/02/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Abstract
In addition to amyloid and tau pathology, elevated systemic vascular risk, white matter injury, and reduced cerebral blood flow contribute to late-life cognitive decline. Given the strong collinearity among these parameters, we proposed a framework to extract the independent latent features underlying cognitive decline using the Harvard Aging Brain Study (N = 166 cognitively unimpaired older adults at baseline). We used the following measures from the baseline visit: cortical amyloid, inferior temporal cortex tau, relative cerebral blood flow, white matter hyperintensities, peak width of skeletonized mean diffusivity, and Framingham Heart Study cardiovascular disease risk. We used exploratory factor analysis to extract orthogonal factors from these variables and their interactions. These factors were used in a regression model to explain longitudinal Preclinical Alzheimer Cognitive Composite-5 (PACC) decline (follow-up = 8.5 ±2.7 years). We next examined whether gray matter volume atrophy acts as a mediator of factors and PACC decline. Latent factors of systemic vascular risk, white matter injury, and relative cerebral blood flow independently explain cognitive decline beyond amyloid and tau. Gray matter volume atrophy mediates these associations with the strongest effect on white matter injury. These results suggest that systemic vascular risk contributes to cognitive decline beyond current markers of cerebrovascular injury, amyloid, and tau.
Collapse
Affiliation(s)
- Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wai-Ying W Yau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessie Fanglu Fu
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Catherine E Scanlon
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie Hsieh
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca E Amariglio
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Papp
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorene Rentz
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Pike NA, Roy B, Cabrera-Mino C, Halnon NJ, Lewis AB, Shao X, Wang DJJ, Kumar R. Compromised Cerebral Arterial Perfusion, Altered Brain Tissue Integrity, and Cognitive Impairment in Adolescents with Complex Congenital Heart Disease. J Cardiovasc Dev Dis 2024; 11:236. [PMID: 39195144 PMCID: PMC11354402 DOI: 10.3390/jcdd11080236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
(1) Introduction: Adolescents with complex congenital heart disease (CCHD) show brain tissue injuries in regions associated with cognitive deficits. Alteration in cerebral arterial perfusion (CAP), as measured by arterial transit time (ATT), may lead to perfusion deficits and potential injury. Our study aims to compare ATT values between CCHD patients and controls and assess the associations between ATT values, MD values, and cognitive scores in adolescents with CCHD. (2) Methods: 37 CCHD subjects, 14-18 years of age, who had undergone surgical palliation and 30 healthy controls completed cognitive testing and brain MRI assessments using a 3.0-Tesla scanner. ATT values and regional brain mean diffusivity [MD] were assessed for the whole brain using diffusion tensor imaging. (3) Results: The mean MoCA values [23.1 ± 4.1 vs. 28.1 ± 2.3; p < 0.001] and General Memory Index, with a subscore of WRAML2 [86.8 ± 15.4 vs. 110.3 ± 14.5; p < 0.001], showed significant cognitive deficits in CCHD patients compared to controls. The mean global ATT was significantly higher in CCHD patients versus controls (mean ± SD, s, 1.26 ± 0.11 vs. 1.19 ± 0.11, p = 0.03), respectively. The partial correlations between ATT values, MD values, and cognitive scores (p < 0.005) showed significant associations in areas including the hippocampus, prefrontal cortices, cerebellum, caudate, anterior and mid cingulate, insula, thalamus, and lingual gyrus. (4) Conclusions: Adolescents with CCHD had prolonged ATTs and showed correlation with clinical measurements of cognitive impairment and MRI measurements of brain tissue integrity. This suggests that altered CAP may play a role in brain tissue injury and cognitive impairment after surgical palliation.
Collapse
Affiliation(s)
- Nancy A. Pike
- Sue & Bill Gross School of Nursing, University of California Irvine, Irvine, CA 92697, USA
- The Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Bhaswati Roy
- Departments of Anesthesiology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Cristina Cabrera-Mino
- School of Nursing, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Nancy J. Halnon
- Division of Pediatric Cardiology, Mattel Children’s Hospital UCLA, Los Angeles, CA 90095, USA;
| | - Alan B. Lewis
- Division of Pediatric Cardiology, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (X.S.); (D.J.J.W.)
| | - Danny J. J. Wang
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (X.S.); (D.J.J.W.)
| | - Rajesh Kumar
- Departments of Anesthesiology, Radiological Sciences & Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA;
| |
Collapse
|
23
|
Liu C, Cárdenas-Rivera A, Teitelbaum S, Birmingham A, Alfadhel M, Yaseen MA. Neuroinflammation increases oxygen extraction in a mouse model of Alzheimer's disease. Alzheimers Res Ther 2024; 16:78. [PMID: 38600598 PMCID: PMC11005245 DOI: 10.1186/s13195-024-01444-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Neuroinflammation, impaired metabolism, and hypoperfusion are fundamental pathological hallmarks of early Alzheimer's disease (AD). Numerous studies have asserted a close association between neuroinflammation and disrupted cerebral energetics. During AD progression and other neurodegenerative disorders, a persistent state of chronic neuroinflammation reportedly exacerbates cytotoxicity and potentiates neuronal death. Here, we assessed the impact of a neuroinflammatory challenge on metabolic demand and microvascular hemodynamics in the somatosensory cortex of an AD mouse model. METHODS We utilized in vivo 2-photon microscopy and the phosphorescent oxygen sensor Oxyphor 2P to measure partial pressure of oxygen (pO2) and capillary red blood cell flux in cortical microvessels of awake mice. Intravascular pO2 and capillary RBC flux measurements were performed in 8-month-old APPswe/PS1dE9 mice and wildtype littermates on days 0, 7, and 14 of a 14-day period of lipopolysaccharide-induced neuroinflammation. RESULTS Before the induced inflammatory challenge, AD mice demonstrated reduced metabolic demand but similar capillary red blood cell flux as their wild type counterparts. Neuroinflammation provoked significant reductions in cerebral intravascular oxygen levels and elevated oxygen extraction in both animal groups, without significantly altering red blood cell flux in capillaries. CONCLUSIONS This study provides evidence that neuroinflammation alters cerebral oxygen demand at the early stages of AD without substantially altering vascular oxygen supply. The results will guide our understanding of neuroinflammation's influence on neuroimaging biomarkers for early AD diagnosis.
Collapse
Affiliation(s)
- Chang Liu
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Shayna Teitelbaum
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Austin Birmingham
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Mohammed Alfadhel
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Mohammad A Yaseen
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Beinlich FR, Asiminas A, Untiet V, Bojarowska Z, Plá V, Sigurdsson B, Timmel V, Gehrig L, Graber MH, Hirase H, Nedergaard M. Oxygen imaging of hypoxic pockets in the mouse cerebral cortex. Science 2024; 383:1471-1478. [PMID: 38547288 PMCID: PMC11251491 DOI: 10.1126/science.adn1011] [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: 11/29/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024]
Abstract
Consciousness is lost within seconds upon cessation of cerebral blood flow. The brain cannot store oxygen, and interruption of oxidative phosphorylation is fatal within minutes. Yet only rudimentary knowledge exists regarding cortical partial oxygen tension (Po2) dynamics under physiological conditions. Here we introduce Green enhanced Nano-lantern (GeNL), a genetically encoded bioluminescent oxygen indicator for Po2 imaging. In awake behaving mice, we uncover the existence of spontaneous, spatially defined "hypoxic pockets" and demonstrate their linkage to the abrogation of local capillary flow. Exercise reduced the burden of hypoxic pockets by 52% compared with rest. The study provides insight into cortical oxygen dynamics in awake behaving animals and concurrently establishes a tool to delineate the importance of oxygen tension in physiological processes and neurological diseases.
Collapse
Affiliation(s)
- Felix R.M. Beinlich
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Antonios Asiminas
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Zuzanna Bojarowska
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Virginia Plá
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Björn Sigurdsson
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Vincenzo Timmel
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Lukas Gehrig
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Michael H. Graber
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Hajime Hirase
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical Center; Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical Center; Rochester, NY 14642, USA
| |
Collapse
|
25
|
Ceci C, Lacal PM, Barbaccia ML, Mercuri NB, Graziani G, Ledonne A. The VEGFs/VEGFRs system in Alzheimer's and Parkinson's diseases: Pathophysiological roles and therapeutic implications. Pharmacol Res 2024; 201:107101. [PMID: 38336311 DOI: 10.1016/j.phrs.2024.107101] [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] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The vascular endothelial growth factors (VEGFs) and their cognate receptors (VEGFRs), besides their well-known involvement in physiological angiogenesis/lymphangiogenesis and in diseases associated to pathological vessel formation, play multifaceted functions in the central nervous system (CNS). In addition to shaping brain development, by controlling cerebral vasculogenesis and regulating neurogenesis as well as astrocyte differentiation, the VEGFs/VEGFRs axis exerts essential functions in the adult brain both in physiological and pathological contexts. In this article, after describing the physiological VEGFs/VEGFRs functions in the CNS, we focus on the VEGFs/VEGFRs involvement in neurodegenerative diseases by reviewing the current literature on the rather complex VEGFs/VEGFRs contribution to the pathogenic mechanisms of Alzheimer's (AD) and Parkinson's (PD) diseases. Thereafter, based on the outcome of VEGFs/VEGFRs targeting in animal models of AD and PD, we discuss the factual relevance of pharmacological VEGFs/VEGFRs modulation as a novel and potential disease-modifying approach for these neurodegenerative pathologies. Specific VEGFRs targeting, aimed at selective VEGFR-1 inhibition, while preserving VEGFR-2 signal transduction, appears as a promising strategy to hit the molecular mechanisms underlying AD pathology. Moreover, therapeutic VEGFs-based approaches can be proposed for PD treatment, with the aim of fine-tuning their brain levels to amplify neurotrophic/neuroprotective effects while limiting an excessive impact on vascular permeability.
Collapse
Affiliation(s)
- Claudia Ceci
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Maria Luisa Barbaccia
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Ada Ledonne
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
26
|
Zhang M, Zhang Z, Li H, Xia Y, Xing M, Xiao C, Cai W, Bu L, Li Y, Park TE, Tang Y, Ye X, Lin WJ. Blockage of VEGF function by bevacizumab alleviates early-stage cerebrovascular dysfunction and improves cognitive function in a mouse model of Alzheimer's disease. Transl Neurodegener 2024; 13:1. [PMID: 38173017 PMCID: PMC10763201 DOI: 10.1186/s40035-023-00388-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the predominant type of dementia worldwide. It is characterized by the progressive and irreversible decline of cognitive functions. In addition to the pathological beta-amyloid (Aβ) deposition, glial activation, and neuronal injury in the postmortem brains of AD patients, increasing evidence suggests that the often overlooked vascular dysfunction is an important early event in AD pathophysiology. Vascular endothelial growth factor (VEGF) plays a critical role in regulating physiological functions and pathological changes in blood vessels, but whether VEGF is involved in the early stage of vascular pathology in AD remains unclear. METHODS We used an antiangiogenic agent for clinical cancer treatment, the humanized monoclonal anti-VEGF antibody bevacizumab, to block VEGF binding to its receptors in the 5×FAD mouse model at an early age. After treatment, memory performance was evaluated by a novel object recognition test, and cerebral vascular permeability and perfusion were examined by an Evans blue assay and blood flow scanning imaging analysis. Immunofluorescence staining was used to measure glial activation and Aβ deposits. VEGF and its receptors were analyzed by enzyme-linked immunosorbent assay and immunoblotting. RNA sequencing was performed to elucidate bevacizumab-associated transcriptional signatures in the hippocampus of 5×FAD mice. RESULTS Bevacizumab treatment administered from 4 months of age dramatically improved cerebrovascular functions, reduced glial activation, and restored long-term memory in both sexes of 5×FAD mice. Notably, a sex-specific change in different VEGF receptors was identified in the cortex and hippocampus of 5×FAD mice. Soluble VEGFR1 was decreased in female mice, while full-length VEGFR2 was increased in male mice. Bevacizumab treatment reversed the altered expression of receptors to be comparable to the level in the wild-type mice. Gene Set Enrichment Analysis of transcriptomic changes revealed that bevacizumab effectively reversed the changes in the gene sets associated with blood-brain barrier integrity and vascular smooth muscle contraction in 5×FAD mice. CONCLUSIONS Our study demonstrated the mechanistic roles of VEGF at the early stage of amyloidopathy and the protective effects of bevacizumab on cerebrovascular function and memory performance in 5×FAD mice. These findings also suggest the therapeutic potential of bevacizumab for the early intervention of AD.
Collapse
Affiliation(s)
- Min Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhan Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Honghong Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Mengdan Xing
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Chuan Xiao
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Wenbao Cai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Lulu Bu
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yi Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yamei Tang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
27
|
Wang W, Yang Y, Sang F, Chen Y, Li X, Chen K, Wang J, Zhang Z. Vascular Risk Factors and Brain Health in Aging: Insights from a Community-Based Cohort Study. J Alzheimers Dis 2024; 99:1361-1374. [PMID: 38788079 DOI: 10.3233/jad-240240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Background The aging population and high rates of Alzheimer's disease (AD) create significant medical burdens, prompting a need for early prevention. Targeting modifiable risk factors like vascular risk factors (VRFs), closely linked to AD, may provide a promising strategy for intervention. Objective This study investigates how VRFs influence cognitive performance and brain structures in a community-based cohort. Methods In this cross-sectional study, 4,667 participants over 50 years old, drawn from the Beijing Ageing Brain Rejuvenation Initiative project, were meticulously examined. Cognitive function and VRFs (diabetes mellitus, hypertension, hyperlipidemia, obesity, and smoking), were comprehensively assessed through one-to-one interviews. Additionally, a subset of participants (n = 719) underwent MRI, encompassing T1-weighted and diffusion-weighted scans, to elucidate gray matter volume and white matter structural network organization. Results The findings unveil diabetes as a potent detriment to memory, manifesting in atrophy within the right supramarginal gyrus and diminished nodal efficiency and degree centrality in the right inferior parietal lobe. Hypertension solely impaired memory without significant structural changes. Intriguingly, individuals with comorbid diabetes and hypertension exhibited the most pronounced deficits in both brain structure and cognitive performance. Remarkably, hyperlipidemia emerged as a factor associated with enhanced cognition, and preservation of brain structure. Conclusions This study illuminates the intricate associations between VRFs and the varied patterns of cognitive and brain structural damage. Notably, the synergistic effect of diabetes and hypertension emerges as particularly deleterious. These findings underscore the imperative to tailor interventions for patients with distinct VRF comorbidities, especially when addressing cognitive decline and structural brain changes.
Collapse
Affiliation(s)
- Wenxiao Wang
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Yiru Yang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Sang
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
| | - Yaojing Chen
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
| | - Xin Li
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
| | - Kewei Chen
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Jun Wang
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
| | - Zhanjun Zhang
- Faculty of Psychology, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI Centre), Beijing Normal University, Beijing, China
| |
Collapse
|
28
|
Adams JA, Uryash A, Lopez JR. Harnessing Passive Pulsatile Shear Stress for Alzheimer's Disease Prevention and Intervention. J Alzheimers Dis 2024; 98:387-401. [PMID: 38393906 PMCID: PMC10977433 DOI: 10.3233/jad-231010] [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] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
Alzheimer's disease (AD) affects more than 40 million people worldwide and is the leading cause of dementia. This disease is a challenge for both patients and caregivers and puts a significant strain on the global healthcare system. To address this issue, the Lancet Commission recommends focusing on reducing modifiable lifestyle risk factors such as hypertension, diabetes, and physical inactivity. Passive pulsatile shear stress (PPSS) interventions, which use devices like whole-body periodic acceleration, periodic acceleration along the Z-axis (pGz), and the Jogging Device, have shown significant systemic and cellular effects in preclinical and clinical models which address these modifiable risks factors. Based on this, we propose that PPSS could be a potential non-pharmacological and non-invasive preventive or therapeutic strategy for AD. We perform a comprehensive review of the biological basis based on all publications of PPSS using these devices and demonstrate their effects on the various aspects of AD. We draw from this comprehensive analysis to support our hypothesis. We then delve into the possible application of PPSS as an innovative intervention. We discuss how PPSS holds promise in ameliorating hypertension and diabetes while mitigating physical inactivity, potentially offering a holistic approach to AD prevention and management.
Collapse
Affiliation(s)
- Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
29
|
Fang X, Fan F, Border JJ, Roman RJ. Cerebrovascular Dysfunction in Alzheimer's Disease and Transgenic Rodent Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2024; 5:42-64. [PMID: 38434588 PMCID: PMC10906803 DOI: 10.33696/neurol.5.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Alzheimer's Disease (AD) and Alzheimer's Disease-Related Dementia (ADRD) are the primary causes of dementia that has a devastating effect on the quality of life and is a tremendous economic burden on the healthcare system. The accumulation of extracellular beta-amyloid (Aβ) plaques and intracellular hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain are the hallmarks of AD. They are also thought to be the underlying cause of inflammation, neurodegeneration, brain atrophy, and cognitive impairments that accompany AD. The discovery of APP, PS1, and PS2 mutations that increase Aβ production in families with early onset familial AD led to the development of numerous transgenic rodent models of AD. These models have provided new insight into the role of Aβ in AD; however, they do not fully replicate AD pathology in patients. Familial AD patients with mutations that elevate the production of Aβ represent only a small fraction of dementia patients. In contrast, those with late-onset sporadic AD constitute the majority of cases. This observation, along with the failure of previous clinical trials targeting Aβ or Tau and the modest success of recent trials using Aβ monoclonal antibodies, has led to a reappraisal of the view that Aβ accumulation is the sole factor in the pathogenesis of AD. More recent studies have established that cerebral vascular dysfunction is one of the earliest changes seen in AD, and 67% of the candidate genes linked to AD are expressed in the cerebral vasculature. Thus, there is an increasing appreciation of the vascular contribution to AD, and the National Institute on Aging (NIA) and the Alzheimer's Disease Foundation recently prioritized it as a focused research area. This review summarizes the strengths and limitations of the most commonly used transgenic AD animal models and current views about the contribution of Aβ accumulation versus cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Physiology, Augusta University, Augusta, GA 30912, USA
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
30
|
Harding IH, Ryan J, Heritier S, Spark S, Flanagan Z, McIntyre R, Anderson CS, Naismith SL, Chong TTJ, O'Sullivan M, Egan G, Law M, Zoungas S. STAREE-Mind Imaging Study: a randomised placebo-controlled trial of atorvastatin for prevention of cerebrovascular decline and neurodegeneration in older individuals. BMJ Neurol Open 2023; 5:e000541. [PMID: 37920607 PMCID: PMC10619122 DOI: 10.1136/bmjno-2023-000541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/08/2023] [Indexed: 11/04/2023] Open
Abstract
Introduction Cerebrovascular disease and neurodegeneration are causes of cognitive decline and dementia, for which primary prevention options are currently lacking. Statins are well-tolerated and widely available medications that potentially have neuroprotective effects. The STAREE-Mind Imaging Study is a randomised, double-blind, placebo-controlled clinical trial that will investigate the impact of atorvastatin on markers of neurovascular health and brain atrophy in a healthy, older population using MRI. This is a nested substudy of the 'Statins for Reducing Events in the Elderly' (STAREE) primary prevention trial. Methods Participants aged 70 years or older (n=340) will be randomised to atorvastatin or placebo. Comprehensive brain MRI assessment will be undertaken at baseline and up to 4 years follow-up, including structural, diffusion, perfusion and susceptibility imaging. The primary outcome measures will be change in brain free water fraction (a composite marker of vascular leakage, neuroinflammation and neurodegeneration) and white matter hyperintensity volume (small vessel disease). Secondary outcomes will include change in perivascular space volume (glymphatic drainage), cortical thickness, hippocampal volume, microbleeds and lacunae, prefrontal cerebral perfusion and white matter microstructure. Ethics and dissemination Academic publications from this work will address the current uncertainty regarding the impact of statins on brain structure and vascular integrity. This study will inform the utility of repurposing these well-tolerated, inexpensive and widely available drugs for primary prevention of neurological outcomes in older individuals. Ethics approval was given by Monash University Human Research Ethics Committee, Protocol 12206. Trial registration number ClinicalTrials.gov Identifier: NCT05586750.
Collapse
Affiliation(s)
- Ian H Harding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Stephane Heritier
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Simone Spark
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Zachary Flanagan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Richard McIntyre
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Craig S Anderson
- Global Brain Health Program, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Sharon L Naismith
- School of Psychology, University of Sydney, Sydney, New South Wales, Australia
| | - Trevor T-J Chong
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Michael O'Sullivan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Gary Egan
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Sophia Zoungas
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Liu C, Cardenas-Rivera A, Teitelbaum S, Birmingham A, Alfadhel M, Yaseen MA. Neuroinflammation increases oxygen extraction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562353. [PMID: 37905082 PMCID: PMC10614808 DOI: 10.1101/2023.10.16.562353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Neuroinflammation, impaired metabolism, and hypoperfusion are fundamental pathological hallmarks of early Alzheimer's disease (AD). Numerous studies have asserted a close association between neuroinflammation and disrupted cerebral energetics. During AD progression and other neurodegenerative disorders, a persistent state of chronic neuroinflammation reportedly exacerbates cytotoxicity and potentiates neuronal death. Here, we assessed the impact of a neuroinflammatory challenge on metabolic demand and microvascular hemodynamics in the somatosensory cortex of an AD mouse model. We utilized in vivo 2-photon microscopy and the phosphorescent oxygen sensor Oxyphor 2P to measure partial pressure of oxygen (pO2) and capillary red blood cell flux in cortical microvessels of awake mice. Intravascular pO2 and capillary RBC flux measurements were performed in 8-month-old APPswe/PS1dE9 mice and wildtype littermates on days 0, 7, and 14 of a 14-day period of lipopolysaccaride-induced neuroinflammation. Before the induced inflammatory challenge, AD mice demonstrated reduced metabolic demand but similar capillary red blood cell flux as their wild type counterparts. Neuroinflammation provoked significant reductions in cerebral intravascular oxygen levels and elevated oxygen extraction in both animal groups, without significantly altering red blood cell flux in capillaries. This study provides evidence that neuroinflammation alters cerebral oxygen demand at the early stages of AD without substantially altering vascular oxygen supply. The results will guide our understanding of neuroinflammation's influence on neuroimaging biomarkers for early AD diagnosis.
Collapse
|
32
|
Qi F, Zuo Z, Hu K, Wang R, Wu T, Liu H, Tang J, Wang Q, Xie Y, Tan L, Yang Y, Zhang X, Zheng J, Xu J, Yao Z, Wang S, Wu LJ, Guo K. VEGF-A in serum protects against memory impairment in APP/PS1 transgenic mice by blocking neutrophil infiltration. Mol Psychiatry 2023; 28:4374-4389. [PMID: 37280283 PMCID: PMC10827659 DOI: 10.1038/s41380-023-02097-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 04/17/2023] [Accepted: 04/27/2023] [Indexed: 06/08/2023]
Abstract
Activation of innate immunity in the brain is a prominent feature of Alzheimer's disease (AD). The present study investigated the regulation of innate immunity by wild-type serum injection in a transgenic AD mouse model. We found that treatment with wild-type mouse serum significantly reduced the number of neutrophils and microglial reactivity in the brains of APP/PS1 mice. Mimicking this effect, neutrophil depletion via Ly6G neutralizing antibodies resulted in improvements in AD brain functions. Serum proteomic analysis identified vascular endothelial growth factor-A (VEGF-A) and chemokine (C-X-C motif) ligand 1 (CXCL1) as factors enriched in serum samples, which are crucial for neutrophil migration and chemotaxis, leukocyte migration, and cell chemotaxis. Exogenous VEGF-A reversed amyloid β (Aβ)-induced decreases in cyclin-dependent kinase 5 (Cdk5) and increases in CXCL1 in vitro and blocked neutrophil infiltration into the AD brain. Endothelial Cdk5 overexpression conferred an inhibitory effect on CXCL1 and neutrophil infiltration, thereby restoring memory abilities in APP/PS1 mice. Our findings uncover a previously unknown link between blood-derived VEGF signaling and neutrophil infiltration and support targeting endothelial Cdk5 signaling as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Fangfang Qi
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Editorial Department of Journal of Sun Yat-sen University, Guangzhou, 510080, China
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zejie Zuo
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Rui Wang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tong Wu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Liu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaoling Tang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qingbo Wang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yufeng Xie
- Five-year Programs of Clinical Medicine in the 2017 grade, School of Medicine, Sun Yat-sen University, Shenzhen, 528406, China
| | - Liren Tan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yunjie Yang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jie Xu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhibin Yao
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shengwen Wang
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Kaihua Guo
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Fang X, Border JJ, Rivers PL, Zhang H, Williams JM, Fan F, Roman RJ. Amyloid beta accumulation in TgF344-AD rats is associated with reduced cerebral capillary endothelial Kir2.1 expression and neurovascular uncoupling. GeroScience 2023; 45:2909-2926. [PMID: 37326915 PMCID: PMC10643802 DOI: 10.1007/s11357-023-00841-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023] Open
Abstract
Alzheimer's disease (AD) exerts a tremendous socio-economic burden worldwide. Although reduced cerebral blood flow is an early and persistent symptom that precedes the loss of cognitive function in AD, the underlying molecular and cellular mechanisms remain unclear. The present study investigated whether capillary endothelial inward rectifier potassium 2 (Kir2.1) expression is reduced in TgF344-AD (AD) rats and contributes to neurovascular uncoupling and cognitive deficits in AD. Three- to fourteen-month-old AD rats expressing mutant human APP and PS1 and age-matched wild-type (WT) F344 rats were studied. AD rats exhibited higher amyloid beta (Aβ) expression in the brain as early as 3 months of age and amyloid plaques by 4 months of age. Functional hyperemic responses induced by whisker stimulation were impaired at 4 months of age, which were exacerbated in 6-month- and 14-month-old AD rats. The expression of Kir2.1 protein was significantly lower in the brains of 6-month-old AD versus WT rats, and Kir2.1 coverage was lower in the cerebral microvasculature of AD than in WT rats. Aβ1-42 reduced the Kir2.1 expression in cultured capillary endothelial cells. Cerebral parenchymal arterioles with attached capillaries exhibited a reduced vasodilator in response to 10 mM K+ applied to capillaries, and constricted less following administration of a Kir2.1 channel blocker, compared to WT vessels. These results indicate that capillary endothelial Kir2.1 expression is reduced and contributes to impaired functional hyperemia in AD rats at early ages, perhaps secondary to elevated Aβ expression.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Patrice L Rivers
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jan Michael Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
34
|
Wang ZG, Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Huang H, Chen L, Manzhulo I, Wiklund L, Sharma HS. Co-administration of dl-3-n-butylphthalide and neprilysin is neuroprotective in Alzheimer disease associated with mild traumatic brain injury. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 172:145-185. [PMID: 37833011 DOI: 10.1016/bs.irn.2023.06.006] [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] [Indexed: 10/15/2023]
Abstract
dl-3-n-Butylphthalide is a potent synthetic Chinese celery extract that is highly efficient in inducing neuroprotection in concussive head injury (CHI), Parkinson's disease, Alzheimer's disease, stroke as well as depression, dementia, anxiety and other neurological diseases. Thus, there are reasons to believe that dl-3-n-butylphthalide could effectively prevent Alzheimer's disease brain pathology. Military personnel during combat operation or veterans are often the victims of brain injury that is a major risk factor for developing Alzheimer's disease in their later lives. In our laboratory we have shown that CHI exacerbates Alzheimer's disease brain pathology and reduces the amyloid beta peptide (AβP) inactivating enzyme neprilysin. We have used TiO2 nanowired-dl-3-n-butylphthalide in attenuating Parkinson's disease brain pathology exacerbated by CHI. Nanodelivery of dl-3-n-butylphthalide appears to be more potent as compared to the conventional delivery of the compound. Thus, it would be interesting to examine the effects of nanowired dl-3-n-butylphthalide together with nanowired delivery of neprilysin in Alzheimer's disease model on brain pathology. In this investigation we found that nanowired delivery of dl-3-n-butylphthalide together with nanowired neprilysin significantly attenuated brain pathology in Alzheimer's disease model with CHI, not reported earlier. The possible mechanism and clinical significance is discussed based on the current literature.
Collapse
Affiliation(s)
- Zhenguo G Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, Hebei Province, P.R. China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston, MA, USA
| | - Hongyun Huang
- Beijing Hongtianji Neuroscience Academy, Beijing, P.R. China
| | - Lin Chen
- Department of Neurosurgery, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, P.R. China
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.
| |
Collapse
|
35
|
Wang N, Yang X, Zhao Z, Liu D, Wang X, Tang H, Zhong C, Chen X, Chen W, Meng Q. Cooperation between neurovascular dysfunction and Aβ in Alzheimer's disease. Front Mol Neurosci 2023; 16:1227493. [PMID: 37654789 PMCID: PMC10466809 DOI: 10.3389/fnmol.2023.1227493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/02/2023] [Indexed: 09/02/2023] Open
Abstract
The amyloid-β (Aβ) hypothesis was once believed to represent the pathogenic process of Alzheimer's disease (AD). However, with the failure of clinical drug development and the increasing understanding of the disease, the Aβ hypothesis has been challenged. Numerous recent investigations have demonstrated that the vascular system plays a significant role in the course of AD, with vascular damage occurring prior to the deposition of Aβ and neurofibrillary tangles (NFTs). The question of how Aβ relates to neurovascular function and which is the trigger for AD has recently come into sharp focus. In this review, we outline the various vascular dysfunctions associated with AD, including changes in vascular hemodynamics, vascular cell function, vascular coverage, and blood-brain barrier (BBB) permeability. We reviewed the most recent findings about the complicated Aβ-neurovascular unit (NVU) interaction and highlighted its vital importance to understanding disease pathophysiology. Vascular defects may lead to Aβ deposition, neurotoxicity, glial cell activation, and metabolic dysfunction; In contrast, Aβ and oxidative stress can aggravate vascular damage, forming a vicious cycle loop.
Collapse
Affiliation(s)
- Niya Wang
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Xiang Yang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong Zhao
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Da Liu
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Xiaoyan Wang
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Hao Tang
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Chuyu Zhong
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Xinzhang Chen
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Wenli Chen
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Qiang Meng
- Department of Neurology, The First People’s Hospital of Yunnan Province, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
36
|
Weijs RWJ, Oudegeest-Sander MH, Vloet JIA, Hopman MTE, Claassen JAHR, Thijssen DHJ. A decade of aging in healthy older adults: longitudinal findings on cerebrovascular and cognitive health. GeroScience 2023; 45:2629-2641. [PMID: 37052769 PMCID: PMC10651595 DOI: 10.1007/s11357-023-00790-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Research suggests an association between cerebrovascular health and cognitive decline, but previous work is limited by its cross-sectional nature or short (< 1-2 years) follow-up. Our aim was to examine, across 10 years of follow-up in healthy older adults, changes in cerebrovascular health and their relationship with subjective memory complaints as an early marker of cognitive decline. Between 2008 and 2010, twenty-eight healthy older adults (69 ± 4 years) underwent baseline blood pressure and transcranial Doppler measurements to assess middle cerebral artery blood velocity (MCAv), cerebrovascular resistance index (CVRi), and measures of cerebral autoregulation (CA). After 9-12 years of follow-up, these measurements were repeated, and presence of memory complaints was evaluated. Linear mixed-model analyses explored effects of aging on cerebrovascular parameters and whether memory complaints were associated with cerebrovascular changes. Across a median follow-up of 10.9 years, no changes in MCAv, CVRi, or CA were found. At baseline, these parameters were not different between subjects with (n = 15) versus without (n = 13) memory complaints. During follow-up, subjects with memory complaints showed larger decreases in MCAv (- 10% versus + 9%, P = 0.041) and increases in CVRi (+ 26% versus - 9%, P = 0.029) compared to other peers without memory complaints, but no distinct changes in CA parameters (P > 0.05). Although a decade of aging does not lead to deterioration in cerebral blood flow or autoregulation, our findings suggest that reductions in cerebral blood flow and increases in cerebrovascular resistance are associated with early subjective cognitive decline.
Collapse
Affiliation(s)
- Ralf W J Weijs
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Madelijn H Oudegeest-Sander
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Department of Geriatrics, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janneke I A Vloet
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Maria T E Hopman
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatrics, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Dick H J Thijssen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
37
|
Sadanandan J, Sathyanesan M, Liu Y, Tiwari NK, Newton SS. Carbamoylated Erythropoietin-Induced Cerebral Blood Perfusion and Vascular Gene Regulation. Int J Mol Sci 2023; 24:11507. [PMID: 37511274 PMCID: PMC10380798 DOI: 10.3390/ijms241411507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Cerebral hypoperfusion is associated with enhanced cognitive decline and increased risk of neuropsychiatric disorders. Erythropoietin (EPO) is a neurotrophic factor known to improve cognitive function in preclinical and clinical studies of neurodegenerative and psychiatric disorders. However, the clinical application of EPO is limited due to its erythropoietic activity that can adversely elevate hematocrit in non-anemic populations. Carbamoylated erythropoietin (CEPO), a chemically engineered non-erythropoietic derivative of EPO, does not alter hematocrit and maintains neurotrophic and behavioral effects comparable to EPO. Our study aimed to investigate the role of CEPO in cerebral hemodynamics. Magnetic resonance imaging (MRI) analysis indicated increased blood perfusion in the hippocampal and striatal region without altering tight junction integrity. In vitro and in vivo analyses indicated that hippocampal neurotransmission was unaltered and increased cerebral perfusion was likely due to EDRF, CGRP, and NOS-mediated vasodilation. In vitro analysis using human umbilical vein endothelial cells (HUVEC) and hippocampal vascular gene expression analysis showed CEPO to be a non-angiogenic agent which regulates the MEOX2 gene expression. The results from our study demonstrate a novel role of CEPO in modulating cerebral vasodilation and blood perfusion.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Yutong Liu
- Radiology Research Division, Department of Radiology, Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| |
Collapse
|
38
|
Carter KJ, Ward AT, Kellawan JM, Harrell JW, Peltonen GL, Roberts GS, Al-Subu A, Hagen SA, Serlin RC, Eldridge MW, Wieben O, Schrage WG. Reduced basal macrovascular and microvascular cerebral blood flow in young adults with metabolic syndrome: potential mechanisms. J Appl Physiol (1985) 2023; 135:94-108. [PMID: 37199780 PMCID: PMC10292973 DOI: 10.1152/japplphysiol.00688.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/26/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023] Open
Abstract
Ninety-million Americans suffer metabolic syndrome (MetSyn), increasing the risk of diabetes and poor brain outcomes, including neuropathology linked to lower cerebral blood flow (CBF), predominantly in anterior regions. We tested the hypothesis that total and regional CBF is lower in MetSyn more so in the anterior brain and explored three potential mechanisms. Thirty-four controls (25 ± 5 yr) and 19 MetSyn (30 ± 9 yr), with no history of cardiovascular disease/medications, underwent four-dimensional flow magnetic resonance imaging (MRI) to quantify macrovascular CBF, whereas arterial spin labeling quantified brain perfusion in a subset (n = 38/53). Contributions of cyclooxygenase (COX; n = 14), nitric oxide synthase (NOS, n = 17), or endothelin receptor A signaling (n = 13) were tested with indomethacin, NG-monomethyl-L-arginine (L-NMMA), and Ambrisentan, respectively. Total CBF was 20 ± 16% lower in MetSyn (725 ± 116 vs. 582 ± 119 mL/min, P < 0.001). Anterior and posterior brain regions were 17 ± 18% and 30 ± 24% lower in MetSyn; reductions were not different between regions (P = 0.112). Global perfusion was 16 ± 14% lower in MetSyn (44 ± 7 vs. 36 ± 5 mL/100 g/min, P = 0.002) and regionally in frontal, occipital, parietal, and temporal lobes (range 15-22%). The decrease in CBF with L-NMMA (P = 0.004) was not different between groups (P = 0.244, n = 14, 3), and Ambrisentan had no effect on either group (P = 0.165, n = 9, 4). Interestingly, indomethacin reduced CBF more in Controls in the anterior brain (P = 0.041), but CBF decrease in posterior was not different between groups (P = 0.151, n = 8, 6). These data indicate that adults with MetSyn exhibit substantially reduced brain perfusion without regional differences. Moreover, this reduction is not due to loss of NOS or gain of ET-1 signaling but rather a loss of COX vasodilation.NEW & NOTEWORTHY We tested the impact of insulin resistance (IR) on resting cerebral blood flow (CBF) in adults with metabolic syndrome (MetSyn). Using MRI and research pharmaceuticals to study the role of NOS, ET-1, or COX signaling, we found that adults with MetSyn exhibit substantially lower CBF that is not explained by changes in NOS or ET-1 signaling. Interestingly, adults with MetSyn show a loss of COX-mediated vasodilation in the anterior but not posterior circulation.
Collapse
Affiliation(s)
- Katrina J Carter
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - Aaron T Ward
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - J Mikhail Kellawan
- Department of Health and Exercise Science, University of Oklahoma, Norman, Oklahoma, United States
| | - John W Harrell
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, United States
| | - Garrett L Peltonen
- School of Nursing and Kinesiology, Western New Mexico University, Silver City, New Mexico, United States
| | - Grant S Roberts
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
| | - Awni Al-Subu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Scott A Hagen
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Ronald C Serlin
- Department of Educational Psychology, University of Wisconsin, Madison, Wisconsin, United States
| | - Marlowe W Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Oliver Wieben
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, United States
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| |
Collapse
|
39
|
Giblin J, Kura S, Nunuez JLU, Zhang J, Kureli G, Jiang J, Boas DA, Chen IA. High throughput detection of capillary stalling events with Bessel beam two-photon microscopy. NEUROPHOTONICS 2023; 10:035009. [PMID: 37705938 PMCID: PMC10495839 DOI: 10.1117/1.nph.10.3.035009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 09/15/2023]
Abstract
Significance Brief disruptions in capillary flow, commonly referred to as capillary "stalling," have gained interest recently for their potential role in disrupting cerebral blood flow and oxygen delivery. Approaches to studying this phenomenon have been hindered by limited volumetric imaging rates and cumbersome manual analysis. The ability to precisely and efficiently quantify the dynamics of these events will be key in understanding their potential role in stroke and neurodegenerative diseases, such as Alzheimer's disease. Aim Our study aimed to demonstrate that the fast volumetric imaging rates offered by Bessel beam two-photon microscopy combined with improved data analysis throughput allows for faster and more precise measurement of capillary stall dynamics. Results We found that while our analysis approach was unable to achieve full automation, we were able to cut analysis time in half while also finding stalling events that were missed in traditional blind manual analysis. The resulting data showed that our Bessel beam system was captured more stalling events compared to optical coherence tomography, particularly shorter stalling events. We then compare differences in stall dynamics between a young and old group of mice as well as a demonstrate changes in stalling before and after photothrombotic model of stroke. Finally, we also demonstrate the ability to monitor arteriole dynamics alongside stall dynamics. Conclusions Bessel beam two-photon microscopy combined with high throughput analysis is a powerful tool for studying capillary stalling due to its ability to monitor hundreds of capillaries simultaneously at high frame rates.
Collapse
Affiliation(s)
- John Giblin
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Sreekanth Kura
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Juan Luis Ugarte Nunuez
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Juncheng Zhang
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Gulce Kureli
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - John Jiang
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Ichun A. Chen
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| |
Collapse
|
40
|
Fang X, Tang C, Zhang H, Border JJ, Liu Y, Shin SM, Yu H, Roman RJ, Fan F. Longitudinal characterization of cerebral hemodynamics in the TgF344-AD rat model of Alzheimer's disease. GeroScience 2023; 45:1471-1490. [PMID: 36933144 PMCID: PMC10400494 DOI: 10.1007/s11357-023-00773-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023] Open
Abstract
Alzheimer's disease (AD) is a global healthcare crisis. The TgF344-AD rat is an AD model exhibiting age-dependent AD pathological hallmarks. We confirmed that AD rats developed cognitive deficits at 6 months without alteration of any other major biophysical parameters. We longitudinally characterized cerebral hemodynamics in AD rats at 3, 4, 6, and 14 months. The myogenic responses of the cerebral arteries and arterioles were impaired at 4 months of age in the AD rats. Consistent with the ex vivo results, the AD rat exhibited poor autoregulation of surface and deep cortical cerebral blood flow 2 months preceding cognitive decline. The dysfunction of cerebral hemodynamics in AD is exacerbated with age associated with reduced cerebral perfusion. Further, abolished cell contractility contributes to cerebral hemodynamics imbalance in AD. This may be attributed to enhanced ROS production, reduced mitochondrial respiration and ATP production, and disrupted actin cytoskeleton in cerebral vascular contractile cells.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Chengyun Tang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
41
|
Tarawneh R. Microvascular Contributions to Alzheimer Disease Pathogenesis: Is Alzheimer Disease Primarily an Endotheliopathy? Biomolecules 2023; 13:830. [PMID: 37238700 PMCID: PMC10216678 DOI: 10.3390/biom13050830] [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: 03/13/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer disease (AD) models are based on the notion that abnormal protein aggregation is the primary event in AD, which begins a decade or longer prior to symptom onset, and culminates in neurodegeneration; however, emerging evidence from animal and clinical studies suggests that reduced blood flow due to capillary loss and endothelial dysfunction are early and primary events in AD pathogenesis, which may precede amyloid and tau aggregation, and contribute to neuronal and synaptic injury via direct and indirect mechanisms. Recent data from clinical studies suggests that endothelial dysfunction is closely associated with cognitive outcomes in AD and that therapeutic strategies which promote endothelial repair in early AD may offer a potential opportunity to prevent or slow disease progression. This review examines evidence from clinical, imaging, neuropathological, and animal studies supporting vascular contributions to the onset and progression of AD pathology. Together, these observations support the notion that the onset of AD may be primarily influenced by vascular, rather than neurodegenerative, mechanisms and emphasize the importance of further investigations into the vascular hypothesis of AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, University of New Mexico, Albuquerque, NM 87106, USA
| |
Collapse
|
42
|
Zhang H, Border JJ, Fang X, Liu Y, Tang C, Gao W, Wang S, Shin SM, Guo Y, Zhang C, Gonzalez-Fernandez E, Yu H, Sun P, Roman RJ, Fan F. Enhanced Cerebral Hemodynamics and Cognitive Function Via Knockout of Dual-Specificity Protein Phosphatase 5. JOURNAL OF PHARMACY AND PHARMACOLOGY RESEARCH 2023; 7:49-61. [PMID: 37588944 PMCID: PMC10430881 DOI: 10.26502/fjppr.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Alzheimer's Disease (AD) and Alzheimer's Disease-Related Dementias (ADRD) are neurodegenerative disorders. Recent studies suggest that cerebral hypoperfusion is an early symptom of AD/ADRD. Dual-specificity protein phosphatase 5 (DUSP5) has been implicated in several pathological conditions, including pulmonary hypertension and cancer, but its role in AD/ADRD remains unclear. The present study builds on our previous findings, demonstrating that inhibition of ERK and PKC leads to a dose-dependent dilation of the middle cerebral artery and penetrating arteriole, with a more pronounced effect in Dusp5 KO rats. Both ERK and PKC inhibitors resulted in a significant reduction of myogenic tone in vessels from Dusp5 KO rats. Dusp5 KO rats exhibited stronger autoregulation of the surface but not deep cortical cerebral blood flow. Inhibition of ERK and PKC significantly enhanced the contractile capacity of vascular smooth muscle cells from both strains. Finally, a significant improvement in learning and memory was observed in Dusp5 KO rats 24 hours after initial training. Our results suggest that altered vascular reactivity in Dusp5 KO rats may involve distinct mechanisms for different vascular beds, and DUSP5 deletion could be a potential therapeutic target for AD/ADRD. Further investigations are necessary to determine the effects of DUSP5 inhibition on capillary stalling, blood-brain barrier permeability, and neurodegeneration in aging and disease models.
Collapse
Affiliation(s)
- Huawei Zhang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
- Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jane J Border
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xing Fang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yedan Liu
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Chengyun Tang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wenjun Gao
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Seung Min Shin
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ya Guo
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Chao Zhang
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Hongwei Yu
- Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Peng Sun
- Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Richard J Roman
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Fan
- Pharmacology &Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
43
|
Zhukov O, He C, Soylu-Kucharz R, Cai C, Lauritzen AD, Aldana BI, Björkqvist M, Lauritzen M, Kucharz K. Preserved blood-brain barrier and neurovascular coupling in female 5xFAD model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1089005. [PMID: 37261266 PMCID: PMC10228387 DOI: 10.3389/fnagi.2023.1089005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/17/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Dysfunction of the cerebral vasculature is considered one of the key components of Alzheimer's disease (AD), but the mechanisms affecting individual brain vessels are poorly understood. Methods Here, using in vivo two-photon microscopy in superficial cortical layers and ex vivo imaging across brain regions, we characterized blood-brain barrier (BBB) function and neurovascular coupling (NVC) at the level of individual brain vessels in adult female 5xFAD mice, an aggressive amyloid-β (Aβ) model of AD. Results We report a lack of abnormal increase in adsorptive-mediated transcytosis of albumin and preserved paracellular barrier for fibrinogen and small molecules despite an extensive load of Aβ. Likewise, the NVC responses to somatosensory stimulation were preserved at all regulatory segments of the microvasculature: penetrating arterioles, precapillary sphincters, and capillaries. Lastly, the Aβ plaques did not affect the density of capillary pericytes. Conclusion Our findings provide direct evidence of preserved microvascular function in the 5xFAD mice and highlight the critical dependence of the experimental outcomes on the choice of preclinical models of AD. We propose that the presence of parenchymal Aβ does not warrant BBB and NVC dysfunction and that the generalized view that microvascular impairment is inherent to Aβ aggregation may need to be revised.
Collapse
Affiliation(s)
- Oleg Zhukov
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chen He
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rana Soylu-Kucharz
- Biomarkers in Brain Disease, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Changsi Cai
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Martin Lauritzen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark
| | - Krzysztof Kucharz
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
45
|
Badimon A, Torrente D, Norris EH. Vascular Dysfunction in Alzheimer's Disease: Alterations in the Plasma Contact and Fibrinolytic Systems. Int J Mol Sci 2023; 24:7046. [PMID: 37108211 PMCID: PMC10138543 DOI: 10.3390/ijms24087046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, affecting millions of people worldwide. The classical hallmarks of AD include extracellular beta-amyloid (Aβ) plaques and neurofibrillary tau tangles, although they are often accompanied by various vascular defects. These changes include damage to the vasculature, a decrease in cerebral blood flow, and accumulation of Aβ along vessels, among others. Vascular dysfunction begins early in disease pathogenesis and may contribute to disease progression and cognitive dysfunction. In addition, patients with AD exhibit alterations in the plasma contact system and the fibrinolytic system, two pathways in the blood that regulate clotting and inflammation. Here, we explain the clinical manifestations of vascular deficits in AD. Further, we describe how changes in plasma contact activation and the fibrinolytic system may contribute to vascular dysfunction, inflammation, coagulation, and cognitive impairment in AD. Given this evidence, we propose novel therapies that may, alone or in combination, ameliorate AD progression in patients.
Collapse
Affiliation(s)
| | | | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
46
|
Skapetze L, Owino S, Lo EH, Arai K, Merrow M, Harrington M. Rhythms in barriers and fluids: Circadian clock regulation in the aging neurovascular unit. Neurobiol Dis 2023; 181:106120. [PMID: 37044366 DOI: 10.1016/j.nbd.2023.106120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
The neurovascular unit is where two very distinct physiological systems meet: The central nervous system (CNS) and the blood. The permeability of the barriers separating these systems is regulated by time, including both the 24 h circadian clock and the longer processes of aging. An endogenous circadian rhythm regulates the transport of molecules across the blood-brain barrier and the circulation of the cerebrospinal fluid and the glymphatic system. These fluid dynamics change with time of day, and with age, and especially in the context of neurodegeneration. Factors may differ depending on brain region, as can be highlighted by consideration of circadian regulation of the neurovascular niche in white matter. As an example of a potential target for clinical applications, we highlight chaperone-mediated autophagy as one mechanism at the intersection of circadian dysregulation, aging and neurodegenerative disease. In this review we emphasize key areas for future research.
Collapse
Affiliation(s)
- Lea Skapetze
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sharon Owino
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martha Merrow
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mary Harrington
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America.
| |
Collapse
|
47
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
48
|
Ruiz-Uribe NE, Bracko O, Swallow M, Omurzakov A, Dash S, Uchida H, Xiang D, Haft-Javaherian M, Falkenhain K, Lamont ME, Ali M, Njiru BN, Chang HY, Tan AY, Xiang JZ, Iadecola C, Park L, Sanchez T, Nishimura N, Schaffer CB. Vascular oxidative stress causes neutrophil arrest in brain capillaries, leading to decreased cerebral blood flow and contributing to memory impairment in a mouse model of Alzheimer’s disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528710. [PMID: 36824768 PMCID: PMC9949082 DOI: 10.1101/2023.02.15.528710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION In this study, we explore the role of oxidative stress produced by NOX2-containing NADPH oxidase as a molecular mechanism causing capillary stalling and cerebral blood flow deficits in the APP/PS1 mouse model of AD. METHODS We inhibited NOX2 in APP/PS1 mice by administering a 10 mg/kg dose of the peptide inhibitor gp91-ds-tat i.p., for two weeks. We used in vivo two-photon imaging to measure capillary stalling, penetrating arteriole flow, and vascular inflammation. We also characterized short-term memory function and gene expression changes in cerebral microvessels. RESULTS We found that after NOX2 inhibition capillary stalling, as well as parenchymal and vascular inflammation, were significantly reduced. In addition, we found a significant increase in penetrating arteriole flow, followed by an improvement in short-term memory, and downregulation of inflammatory gene expression pathways. DISCUSSION Oxidative stress is a major mechanism leading to microvascular dysfunction in AD, and represents an important therapeutic target.
Collapse
|
49
|
Cheng ZZ, Gao F, Lv XY, Wang Q, Wu Y, Sun BL, Shen Y. Features of Cerebral Small Vessel Disease Contributes to the Differential Diagnosis of Alzheimer's Disease. J Alzheimers Dis 2023; 91:795-804. [PMID: 36502328 DOI: 10.3233/jad-220872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD), which comprises the typical features of white matter hyperintensity (WMH) and Vichor-Robin spaces (VRSs) in the brain, is one of the leading causes of aging-related cognitive decline and, ultimately, contributes to the occurrence of dementia, including Alzheimer's disease (AD). OBJECTIVE To investigate whether CSVD imaging markers modify the pathological processes of AD and whether these markers improve AD diagnosis. METHODS 208 participants were enrolled in the China Aging and Neurodegenerative Initiative (CANDI). Fluid AD biomarkers were detected using a single-molecule array, and cerebral small vessel dysfunction was determined using magnetic resonance imaging. RESULTS WMH contributed to AD pathology only within the NC and MCI groups (CDR ≤0.5), whereas VRSs had no effect on AD pathology. The associations between AD biomarkers and cognitive mental status were consistent with the presence of CSVD pathology. That is, within individuals without CSVD pathology, the MMSE scores were correlated with AD fluid biomarkers, except for plasma Aβ42 and Aβ40. Increased plasma p-Tau levels were associated with worse cognitive performance in individuals with WMH (β= -0.465, p = 0.0016) or VRSs (β= -0.352, p = 0.0257) pathology. Plasma AD biomarkers combined with CSVD markers showed high accuracy in diagnosing dementia. CONCLUSION Findings from this cross-sectional cohort study support the notion that CSVD is a risk factor for dementia and highlights that vascular pathology can promote AD biomarker levels, especially in the early course of the disease. Moreover, our results suggest that adding a vascular category to the ATN framework improves the diagnostic accuracy of AD.
Collapse
Affiliation(s)
- Zhao-Zhao Cheng
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin-Yi Lv
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiong Wang
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Wu
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bao-Liang Sun
- The Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Yong Shen
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
50
|
Toth D, Reglodi D, Schwieters L, Tamas A. Role of endocrine PACAP in age-related diseases. Front Endocrinol (Lausanne) 2023; 14:1118927. [PMID: 36967746 PMCID: PMC10033946 DOI: 10.3389/fendo.2023.1118927] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a conserved neuropeptide, which confers diverse anti-aging endocrine and paracrine/autocrine effects, including anti-apoptotic, anti-inflammatory and antioxidant action. The results of the in vivo and in vitro experiments show that increasing emphasis is being placed on the diagnostic/prognostic biomarker potential of this neuropeptide in a wide array of age-related diseases. After the initial findings regarding the presence and alteration of PACAP in different body fluids in physiological processes, an increasing number of studies have focused on the changes of its levels in various pathological conditions associated with advanced aging. Until 2016 - when the results of previous human studies were reviewed - a vast majority of the studies had dealt with age-related neurological diseases, like cerebrovascular and neurodegenerative diseases, multiple sclerosis, as well as some other common diseases in elderly such as migraine, traumatic brain injury and post-traumatic stress disorder, chronic hepatitis and nephrotic syndrome. The aim of this review is to summarize the old and the new results and highlight those 'classical' and emerging clinical fields in which PACAP may become subject to further investigation as a diagnostic and/or prognostic biomarker in age-related diseases.
Collapse
Affiliation(s)
- Denes Toth
- Department of Forensic Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Dora Reglodi
- Department of Anatomy, ELKH-PTE PACAP Research Team, Centre for Neuroscience, University of Pécs Medical School, Pécs, Hungary
| | - Lili Schwieters
- Department of Anatomy, ELKH-PTE PACAP Research Team, Centre for Neuroscience, University of Pécs Medical School, Pécs, Hungary
| | - Andrea Tamas
- Department of Anatomy, ELKH-PTE PACAP Research Team, Centre for Neuroscience, University of Pécs Medical School, Pécs, Hungary
- *Correspondence: Andrea Tamas,
| |
Collapse
|