1
|
Zhang Y, Li T, Miao J, Zhang Z, Yang M, Wang Z, Yang B, Zhang J, Li H, Su Q, Guo J. Gamma-glutamyl transferase 5 overexpression in cerebrovascular endothelial cells improves brain pathology, cognition, and behavior in APP/PS1 mice. Neural Regen Res 2025; 20:533-547. [PMID: 38819065 PMCID: PMC11317949 DOI: 10.4103/nrr.nrr-d-23-01525] [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: 09/10/2023] [Revised: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00030/figure1/v/2024-05-28T214302Z/r/image-tiff In patients with Alzheimer's disease, gamma-glutamyl transferase 5 (GGT5) expression has been observed to be downregulated in cerebrovascular endothelial cells. However, the functional role of GGT5 in the development of Alzheimer's disease remains unclear. This study aimed to explore the effect of GGT5 on cognitive function and brain pathology in an APP/PS1 mouse model of Alzheimer's disease, as well as the underlying mechanism. We observed a significant reduction in GGT5 expression in two in vitro models of Alzheimer's disease (Aβ1-42-treated hCMEC/D3 and bEnd.3 cells), as well as in the APP/PS1 mouse model. Additionally, injection of APP/PS1 mice with an adeno-associated virus encoding GGT5 enhanced hippocampal synaptic plasticity and mitigated cognitive deficits. Interestingly, increasing GGT5 expression in cerebrovascular endothelial cells reduced levels of both soluble and insoluble amyloid-β in the brains of APP/PS1 mice. This effect may be attributable to inhibition of the expression of β-site APP cleaving enzyme 1, which is mediated by nuclear factor-kappa B. Our findings demonstrate that GGT5 expression in cerebrovascular endothelial cells is inversely associated with Alzheimer's disease pathogenesis, and that GGT5 upregulation mitigates cognitive deficits in APP/PS1 mice. These findings suggest that GGT5 expression in cerebrovascular endothelial cells is a potential therapeutic target and biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi Province, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhina Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mingxuan Yang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhuoran Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Yang
- Department of Hernia and Abdominal Wall Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jiawei Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Haiting Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi Province, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
2
|
Jeong H, Pan Y, Akhter F, Volkow ND, Zhu D, Du C. Evidence of cortical vascular impairments in early stage of Alzheimer's transgenic mice: Optical imaging. J Cereb Blood Flow Metab 2024:271678X241304893. [PMID: 39696904 DOI: 10.1177/0271678x241304893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder with progressive cognitive decline, remains clinically challenging with limited understanding of etiology and interventions. Clinical studies have reported vascular defects prior to other pathological manifestations of AD, leading to the "Vascular Hypothesis" for the disorder. However, in vivo assessments of cerebral vasculature in AD rodent models have been constrained by limited spatiotemporal resolution or field of view of conventional imaging. We herein employed two in vivo imaging technologies, Dual-Wavelength Imaging and Optical Coherence Doppler Tomography, to evaluate cerebrovascular reactivity (CVR) to vasoconstrictive cocaine and vasodilatory hypercapnia challenges and to detect resting 3D cerebral blood flow (CBF) in living transgenic AD mice at capillary resolution. Results showed that CVR to cocaine and hypercapnia was significantly attenuated in 7-10 months old AD mice vs controls, indicating reduced vascular flexibility and reactivity. Additionally, in the AD mice, arterial CBF velocities were slower and the microvascular density in cortex was decreased compared to controls. These results reveal significant vascular impairments including reduced CVR and resting CBF in early-staged AD mice. Hence, this cutting-edge in vivo optical imaging offers an innovative venue for detecting early neurovascular dysfunction in AD brain with translational potential.
Collapse
Affiliation(s)
- Hyomin Jeong
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Firoz Akhter
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Congwu Du
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| |
Collapse
|
3
|
Charlton BT, Goulding RP, Jaspers RT, Appelman B, van Vugt M, Wüst RCI. Skeletal muscle adaptations and post-exertional malaise in long COVID. Trends Endocrinol Metab 2024:S1043-2760(24)00298-4. [PMID: 39694730 DOI: 10.1016/j.tem.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024]
Abstract
When acute SARS-CoV-2 infections cause symptoms that persist longer than 3 months, this condition is termed long COVID. Symptoms experienced by patients often include myalgia, fatigue, brain fog, cognitive impairments, and post-exertional malaise (PEM), which is the worsening of symptoms following mental or physical exertion. There is little consensus on the pathophysiology of exercise-induced PEM and skeletal-muscle-related symptoms. In this opinion article we highlight intrinsic mitochondrial dysfunction, endothelial abnormalities, and a muscle fiber type shift towards a more glycolytic phenotype as main contributors to the reduced exercise capacity in long COVID. The mechanistic trigger for physical exercise to induce PEM is unknown, but rapid skeletal muscle tissue damage and intramuscular infiltration of immune cells contribute to PEM-related symptoms.
Collapse
Affiliation(s)
- Braeden T Charlton
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands; Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands
| | - Richie P Goulding
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands; Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands
| | - Richard T Jaspers
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands; Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands
| | - Brent Appelman
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Michèle van Vugt
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands; Division of Infectious Diseases, Tropical Medicine, Department of Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Rob C I Wüst
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands; Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Li H, Zheng C, Zheng Y, Wen K, Zhang Y. Distinct functional diversity of branched oligosaccharides as chaperones and inhibitory-binding partners of amyloid beta-protein and its aggregates. Neuropharmacology 2024; 261:110141. [PMID: 39251087 DOI: 10.1016/j.neuropharm.2024.110141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/14/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
Aggregation and deposition of amyloid beta-protein 1-42 (Aβ42) in the brain, primarily owing to hydrophobic interactions between Aβ42 chains, is a common pathology in all forms of Alzheimer's disease (AD). Hydrophilic oligosaccharides are widely present in the extracellular matrix and on the cytoplasmic membrane. To determine if oligosaccharides bind to Aβ42 or its aggregates and consequently affect their aggregation and cellular function, this study examined the interaction of typical functional oligosaccharides with Aβ42 or its aggregates. Isomaltooligosaccharides (IMOs), particularly isomaltotriose, panose, and isomaltotetraose, functioned as molecular chaperones for Aβ42 by binding directly to Aβ42, preserving Aβ42's active conformation and cytotrophic activity. Oral IMOs reduced total plasma Aβ level and indirectly caused a slight reduction in the load of Aβ42 spots/plaques in the brain of AD model mice (male). Another branched oligosaccharide, bianntennary core pentasaccharide (BCP), had a relatively high binding specificity for Aβ42 oligomers (Aβ42O) and acted as an antagonistic binding partner for Aβ42O. Free BCP effectively blocked/prevented further assembly of Aβ42O and their toxicity to neural and vascular endothelial cell lines. Since BCP is also a signaling component of membrane targets (glycolipids, glycoproteins or receptors), it seemed that BCP had two opposing effects on the binding of Aβ42O to target cells. This study's findings suggest that these branched oligosaccharides may be potential candidates for blocking or preventing Aβ42 aggregation and Aβ42O cytotoxicity/neurotoxicity, respectively, and that IMO-like or free BCP-like oligosaccharide deficiencies in the brain may be one of the underlying mechanisms for Aβ42 aggregation and Aβ42O cytotoxicity.
Collapse
Affiliation(s)
- He Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Changxin Zheng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yanru Zheng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Kai Wen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yingjiu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|
5
|
Thomsen MS, Kostrikov S, Routhe LG, Johnsen KB, Helgudóttir SS, Gudbergsson JM, Andresen TL, Moos T. Remodeling of the brain angioarchitecture in experimental chronic neurodegeneration. Neurobiol Dis 2024; 204:106761. [PMID: 39662534 DOI: 10.1016/j.nbd.2024.106761] [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: 08/28/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Chronic neurodegenerative diseases are characterized by substantial inflammation with putative impairment of the brain vasculature also commonly observed. To address effects of chronic neurodegeneration on the regional vasculature under experimentally controlled circumstances, the glutamate receptor agonist ibotenic acid was injected into striatum of adult rats, which causes excitotoxicity in the substantia nigra pars reticulata (SNpr) due to imbalance between inhibitory inputs from the striatum and excitatory signals from the subthalamic nucleus. Brains were examined at 28 days (short-term neurodegeneration) and 91 days (long-term neurodegeneration) and analyzed for vascular remodeling taking both 2D and 3D approaches, the latter involving confocal microscopy of optically cleared samples combined with machine learning-based image analysis. Crysectioned and microdissected samples were analyzed for protein and gene expression respectively. The resulting neurodegeneration was accompanied by regional tissue loss and inflammation. The 3D analysis of the degenerating SNpr revealed substantial changes of the vasculature with higher density, increased diameter, and number of tortuous vessels already after 28 days, evidently continuing at 91 days. Interestingly, the vascular remodeling changes occurred without changes in the expression of endothelial tight junction proteins, vascular basement membrane proteins, or markers of angiogenesis. We propose that remodeling of the vasculature in neurodegeneration occurs due to regional tissue atrophy, which leaves the vasculature operating but prone to additional pathologies.
Collapse
Affiliation(s)
- Maj Schneider Thomsen
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Serhii Kostrikov
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lisa Greve Routhe
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Kasper Bendix Johnsen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Steinunn Sara Helgudóttir
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Johann Mar Gudbergsson
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Torben Moos
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
6
|
Wang X, Yang J, Zhang X, Cai J, Zhang J, Cai C, Zhuo Y, Fang S, Xu X, Wang H, Liu P, Zhou S, Wang W, Hu Y, Fang J. An endophenotype network strategy uncovers YangXue QingNao Wan suppresses Aβ deposition, improves mitochondrial dysfunction and glucose metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156158. [PMID: 39447228 DOI: 10.1016/j.phymed.2024.156158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Alzheimer's disease (AD), an escalating global health issue, lacks effective treatments due to its complex pathogenesis. YangXue QingNao Wan (YXQNW) is a China Food and Drug Administration (CFDA)- approved TCM formula that has been repurposed in clinical Phase II for the treatment of AD. Identifying YXQNW's active ingredients and their mechanisms is crucial for developing effective AD treatments. PURPOSE This study aims to elucidate the anti-AD effects of YXQNW and to explore its potential therapeutic mechanisms employing an endophenotype network strategy. METHODS Herein we present an endophenotype network strategy that combines active ingredient identification in rat serum, network proximity prediction, metabolomics, and in vivo experimental validation in two animal models. Specially, utilizing UPLC-Q-TOF-MS/MS, active ingredients are identified in YXQNW to build a drug-target network. We applied network proximity to identify potential AD pathological mechanisms of YXQNW via integration of drug-target network, AD endophenotype gene sets, and human protein interactome, and validated related mechanisms in two animal models. In a d-galactose-induced senescent rat model, YXQNW was administered at varying doses for cognitive and neuronal assessments through behavioral tests, Nissl staining, and transmission electron microscopy (TEM). Metabolomic analysis with LC-MS revealed YXQNW's influence on brain metabolites, suggesting therapeutic pathways. Levels of key proteins and biochemicals were measured by WB and ELISA, providing insights into YXQNW's neuroprotective mechanisms. In addition, 5×FAD model mice were used and administered YXQNW by gavage for 14 days at two doses. Amyloid-β levels, transporter expression, and cerebral blood flow have been detected by MRI and biochemical assays. RESULTS The network proximity analysis showed that the effect of YXQNW on AD was highly correlated with amyloid β, synaptic function, glucose metabolism and mitochondrial function. The results of metabolomics combined with in vivo experimental validation suggest that YXQNW has the potential to ameliorate glucose transport abnormalities in the brain by upregulating the expression of GLUT1 and GLUT3, while further enhancing glucose metabolism through increased O-GlcNAcylation and mitigating mitochondrial dysfunction via the AMPK/Sirt1 pathway, thereby improving d-galactose-induced cognitive deficits in rats. Additionally, YXQNW treatment significantly decreased Aβ1-42 levels and enhanced cerebral blood flow (CBF) in the hippocampus of 5×FAD mice. while mechanistic findings indicated that YXQNW treatment increased the expression of ABCB1, an Aβ transporter, in 5×FAD model mice to promote the clearance of Aβ from the brain and alleviate AD-like symptoms. CONCLUSIONS This study reveals that YXQNW may mitigate AD by inhibiting Aβ deposition and ameliorating mitochondrial dysfunction and glucose metabolism, thus offering a promising therapeutic approach for AD.
Collapse
Affiliation(s)
- Xue Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jinna Yang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin, 300193, China; Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd, China
| | - Xiaolian Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jinyong Cai
- Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Jieqi Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chuipu Cai
- Division of Data Intelligence, Department of Computer Science, Shantou University, Shantou 515063, China
| | - Yue Zhuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xinxin Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Peng Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin, 300193, China; Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Shuiping Zhou
- State Key Laboratory of Chinese Medicine Modernization, Tianjin, 300193, China; Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Wenjia Wang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin, 300193, China; Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd, China
| | - Yunhui Hu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin, 300193, China; Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd, China.
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
7
|
Suzuki H, Murata J, Unekawa M, Kanno I, Izawa Y, Tomita Y, Tanaka KF, Nakahara J, Masamoto K. Microfluctuations in Capillary Lumens Independent of Pericyte Lining Density in the Anesthetized Mouse Cortex. Microcirculation 2024; 31:e12885. [PMID: 39283679 DOI: 10.1111/micc.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVE This study aimed to examine the spatiotemporal coherence of capillary lumen fluctuations in relation to spatial variations in the pericyte lining in the cortex of anesthetized mice. METHODS Two-photon microscopic angiography data (previously published) were reanalyzed, and spatial variations in capillary diameter fluctuations at rest and in capillary lining with vascular mural cells were measured along capillary centerlines. RESULTS Relatively large diameters of the capillaries (5.5 μm) coincided with a dense pericyte lining, while small capillaries (4.3 μm) had a sparse pericyte lining. Temporal variations had a frequency of about 0.1 Hz with an amplitude of 0.5 μm, which were negatively correlated with pericyte lining density. Spatial frequency analysis further revealed a common pattern of spatial variations in capillary diameter and pericyte lining, but temporal variations differed. The temporal variations in capillary lumens were locally distinct from those in neighboring locations, suggesting intrinsic fluctuations independent of the pericyte lining. CONCLUSIONS Capillary lumens in the brain exhibit slow microfluctuations that are independent of pericyte lining. These microfluctuations could affect the distribution of flowing blood cells and may be important for homogenizing their distribution in capillary networks.
Collapse
Affiliation(s)
- Hiroki Suzuki
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Juri Murata
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Tomita Hospital, Nagoya, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
8
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [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: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
9
|
Hao J, Kwapong WR, Shen T, Fu H, Xu Y, Lu Q, Liu S, Zhang J, Liu Y, Zhao Y, Zheng Y, Frangi AF, Zhang S, Qi H, Zhao Y. Early detection of dementia through retinal imaging and trustworthy AI. NPJ Digit Med 2024; 7:294. [PMID: 39428420 PMCID: PMC11491446 DOI: 10.1038/s41746-024-01292-5] [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: 05/22/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024] Open
Abstract
Alzheimer's disease (AD) is a global healthcare challenge lacking a simple and affordable detection method. We propose a novel deep learning framework, Eye-AD, to detect Early-onset Alzheimer's Disease (EOAD) and Mild Cognitive Impairment (MCI) using OCTA images of retinal microvasculature and choriocapillaris. Eye-AD employs a multilevel graph representation to analyze intra- and inter-instance relationships in retinal layers. Using 5751 OCTA images from 1671 participants in a multi-center study, our model demonstrated superior performance in EOAD (internal data: AUC = 0.9355, external data: AUC = 0.9007) and MCI detection (internal data: AUC = 0.8630, external data: AUC = 0.8037). Furthermore, we explored the associations between retinal structural biomarkers in OCTA images and EOAD/MCI, and the results align well with the conclusions drawn from our deep learning interpretability analysis. Our findings provide further evidence that retinal OCTA imaging, coupled with artificial intelligence, will serve as a rapid, noninvasive, and affordable dementia detection.
Collapse
Affiliation(s)
- Jinkui Hao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - William R Kwapong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Shen
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Huazhu Fu
- Institute of High-Performance Computing, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yanwu Xu
- School of Future Technology, South China University of Technology, Guangzhou, China
| | - Qinkang Lu
- Department of Ophthalmology, the Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Shouyue Liu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Jiong Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yonghuai Liu
- Department of Computer Science, Edge Hill University, Ormskirk, UK
| | - Yifan Zhao
- School of Aerospace, Transport and Manufacturing, Cranfield University, Bedfordshire, UK
| | - Yalin Zheng
- Department of Eye and Vision Sciences, University of Liverpool, Liverpool, UK
| | - Alejandro F Frangi
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
- Department of Computer Science, School of Engineering, The University of Manchester, Manchester, United Kingdom
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.
| | - Yitian Zhao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Department of Ophthalmology, the Affiliated People's Hospital of Ningbo University, Ningbo, China.
- Department of Eye and Vision Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
10
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
11
|
Lin H, Wang Z, Liao Y, Yu Z, Xu H, Qin T, Tang J, Yang X, Chen S, Chen X, Zhang X, Shen Y. Super-resolution ultrasound imaging reveals temporal cerebrovascular changes with disease progression in female 5×FAD mouse model of Alzheimer's disease: correlation with pathological impairments. EBioMedicine 2024; 108:105355. [PMID: 39293213 PMCID: PMC11424966 DOI: 10.1016/j.ebiom.2024.105355] [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: 04/12/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Vascular dysfunction is closely associated with the progression of Alzheimer's disease (AD). A critical research gap exists that no studies have explored the in vivo temporal changes of cerebrovascular alterations with AD progression in mouse models, encompassing both structure and flow dynamics at micron-scale resolution across the early, middle, and late stages of the disease. METHODS In this study, ultrasound localisation microscopy (ULM) was applied to image the cerebrovascular alterations of the transgenic female 5×FAD mouse model across different stages of disease progression: early (4 months), moderate (7 months), and late (12 months). Age-matched non-transgenic (non-Tg) littermates were used as controls. Immunohistology examinations were performed to evaluate the influence of disease progression on the β-amyloid (Aβ) load and microvascular alterations, including morphological changes and the blood-brain barrier (BBB) leakage. FINDINGS Our findings revealed a significant decline in both vascular density and flow velocity in the retrosplenial cortex of 5×FAD mice at an early stage, which subsequently became more pronounced in the visual cortex and hippocampus as the disease progressed. Additionally, we observed a reduction in vascular length preceding diminished flow velocities in cortical penetrating arterioles during the early stages. The quantification of vascular metrics derived from ULM imaging showed significant correlations with those obtained from vascular histological images. Immunofluorescence staining identified early vascular abnormalities in the retrosplenial cortex. As the disease advanced, there was an exacerbation of Aβ accumulation and BBB disruption in a regionally variable manner. The vascular changes observed through ULM imaging exhibited a negative correlation with amyloid load and were associated with the compromise of the BBB integrity. INTERPRETATION Through high-resolution, in vivo imaging of cerebrovasculature, this study reveals significant spatiotemporal dysfunction in cerebrovascular dynamics accompanying disease progression in a mouse model of AD, enhancing our understanding of its pathophysiology. FUNDING This study is supported by grants from National Key Research and Development Program of China (2020YFA0908800), National Natural Science Foundation of China (12074269, 82272014, 82327804, 62071310), Shenzhen Basic Science Research (20220808185138001, JCYJ20220818095612027, JCYJ20210324093006017), STI 2030-Major Projects (2021ZD0200500) and Guangdong Natural Science Foundation (2024A1515012591, 2024A1515011342).
Collapse
Affiliation(s)
- Haoming Lin
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Zidan Wang
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Yingtao Liao
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China; Department of Radiation Oncology, Huizhou Central People's Hospital, Huizhou, 516001, Guangdong, China
| | - Zhifan Yu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Huiqin Xu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Ting Qin
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Jianbo Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Centre for Disease Control and Prevention, Shenzhen, 518055, China
| | - Siping Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Xin Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Xinyu Zhang
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China
| | - Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518071, China.
| |
Collapse
|
12
|
Curro KR, van Nispen RMA, den Braber A, van de Giessen EM, van de Kreeke JA, Tan HS, Visser PJ, Bouwman FH, Verbraak FD. Longitudinal Assessment of Retinal Microvasculature in Preclinical Alzheimer's Disease. Invest Ophthalmol Vis Sci 2024; 65:2. [PMID: 39361291 PMCID: PMC11451830 DOI: 10.1167/iovs.65.12.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Purpose To investigate if changes in vessel density (VD) and the foveal avascular zone (FAZ) occur in the preclinical phase of Alzheimer's disease (pAD) over time. Methods Optical coherence tomography angiography (OCTA) was used to image VD and FAZ at baseline and for a follow-up period of 2 years. Positron emission tomography (PET) was used to determine the amyloid beta (Aβ) status of participants. Results The VD and FAZ of 148 participants (54% female) were analyzed at baseline and follow-up (mean time between measurements, 2.24 ± 0.35 years). The mean age of the participants was 68.3 ± 6.0 years at baseline and 70.3 ± 5.9 years at follow-up. Participants were divided into three groups: control group, participants who had negative Aβ status at both measurements (Aβ-, n = 116); converter group, participants who transitioned from negative to positive between baseline and follow-up (Aβ-+, n = 18); and participants who were consistently positive at both visits (Aβ++, n = 14). The VD of both Aβ+ groups demonstrated non-significant increases over time in both macula and optic nerve head (ONH) regions. The Aβ- group was found to be significantly higher in both ONH and macular regions. The VD of the Aβ++ group was significantly higher in the macula inner and outer rings compared to the Aβ-+ and Aβ- groups. No significant change was found in FAZ values over time. Conclusions Alterations in VD seem to manifest already in pAD, exhibiting distinct variations between the ONH and macula. Further longitudinal studies with a longer follow-up design and known amyloid pathology should be undertaken to validate these observations.
Collapse
Affiliation(s)
- Katie R. Curro
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ruth M. A. van Nispen
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | - H. Stevie Tan
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pieter-Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Femke H. Bouwman
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frank D. Verbraak
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Wang J, Wang X, Yang J, Zhen Y, Ban W, Zhu G. Molecular profiling of a rat model of vascular dementia: Evidences from proteomics, metabolomics and experimental validations. Brain Res 2024; 1846:149254. [PMID: 39341485 DOI: 10.1016/j.brainres.2024.149254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Decrease of cerebral blood flow is the primary cause of vascular dementia (VD), but its pathophysiological mechanisms are still not known. This study aims to profile the molecular changes of a rat model of VD induced by bilateral common carotid artery ligation. The Morris water maze and new object recognition tasks were used to test the cognitive function of rats. Hematoxylin and Eosin (HE) staining was used to detect pathological changes in the hippocampus. After confirming the model, proteomics was used to detect differentially expressed proteins in the hippocampus, and metabolomics was used to detect differential metabolites in rat serum. Thereafter, bioinformatics were used to integrate and analyze the potential molecular profile. The results showed that compared with the sham control group, the spatial and recognition memory of the rats were significantly reduced, and pathological changes were observed in the hippocampal CA1 region of the model group. Proteomic analysis suggested 206 differentially expressed proteins in the hippocampus of VD rats, with 117 proteins upregulated and 89 downregulated. Protein-protein interaction network analysis suggested that those differentially expressed proteins might play crucial roles in lipid metabolism, cell adhesion, intracellular transport, and signal transduction. Metabolomics analysis identified 103 differential metabolites, and comparison with the human metabolome database revealed 22 common metabolites, which predicted 265 potential targets. Afterwards, by intersecting the predicted results from metabolomics with the differentially expressed proteins from proteomics, we identified five potential targets, namely ACE, GABBR1, Rock1, Abcc1 and Mapk10. Furthermore, western blotting confirmed that compared with control group, hippocampal GABBR1 and Rock1 were enhanced in the model group. Together, this study showed the molecular profile of VD rats through a combination of proteomics, metabolomics, and experimental confirmation methods, offering crucial molecular targets for the diagnosis and treatment of VD.
Collapse
Affiliation(s)
- Jingji Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China; Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China
| | - Xueqing Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jun Yang
- The First Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230031, China.
| | - Yilan Zhen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wenming Ban
- Taihe County Hospital of Traditional Chinese Medicine, Fuyang 236600, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
14
|
Leitner D, Kavanagh T, Kanshin E, Balcomb K, Pires G, Thierry M, Suazo JI, Schneider J, Ueberheide B, Drummond E, Wisniewski T. Differences in the cerebral amyloid angiopathy proteome in Alzheimer's disease and mild cognitive impairment. Acta Neuropathol 2024; 148:9. [PMID: 39039355 PMCID: PMC11263258 DOI: 10.1007/s00401-024-02767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by amyloid beta (Aβ) deposition in cerebrovasculature. It is prevalent with aging and Alzheimer's disease (AD), associated with intracerebral hemorrhage, and contributes to cognitive deficits. To better understand molecular mechanisms, CAA(+) and CAA(-) vessels were microdissected from paraffin-embedded autopsy temporal cortex of age-matched Control (n = 10), mild cognitive impairment (MCI; n = 4), and sporadic AD (n = 6) cases, followed by label-free quantitative mass spectrometry. 257 proteins were differentially abundant in CAA(+) vessels compared to neighboring CAA(-) vessels in MCI, and 289 in AD (p < 0.05, fold-change > 1.5). 84 proteins changed in the same direction in both groups, and many changed in the same direction among proteins significant in at least one group (p < 0.0001, R2 = 0.62). In CAA(+) vessels, proteins significantly increased in both AD and MCI were particularly associated with collagen-containing extracellular matrix, while proteins associated with ribonucleoprotein complex were significantly decreased in both AD and MCI. In neighboring CAA(-) vessels, 61 proteins were differentially abundant in MCI, and 112 in AD when compared to Control cases. Increased proteins in CAA(-) vessels were associated with extracellular matrix, external encapsulating structure, and collagen-containing extracellular matrix in MCI; collagen trimer in AD. Twenty two proteins were increased in CAA(-) vessels of both AD and MCI. Comparison of the CAA proteome with published amyloid-plaque proteomic datasets identified many proteins similarly enriched in CAA and plaques, as well as a protein subset hypothesized as preferentially enriched in CAA when compared to plaques. SEMA3G emerged as a CAA specific marker, validated immunohistochemically and with correlation to pathology levels (p < 0.0001; R2 = 0.90). Overall, the CAA(-) vessel proteomes indicated changes in vessel integrity in AD and MCI in the absence of Aβ, and the CAA(+) vessel proteome was similar in MCI and AD, which was associated with vascular matrix reorganization, protein translation deficits, and blood brain barrier breakdown.
Collapse
Affiliation(s)
- Dominique Leitner
- Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Tomas Kavanagh
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies and Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Kaleah Balcomb
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Geoffrey Pires
- Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Manon Thierry
- Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jianina I Suazo
- Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Julie Schneider
- Department Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Suite 1000, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Beatrix Ueberheide
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Proteomics Laboratory, Division of Advanced Research Technologies and Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Eleanor Drummond
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Thomas Wisniewski
- Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
15
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Llamas Rodríguez J, van der Kouwe AJW, Oltmer J, Rosenblum E, Mercaldo N, Fischl B, Marshall M, Frosch MP, Augustinack JC. Entorhinal vessel density correlates with phosphorylated tau and TDP-43 pathology. Alzheimers Dement 2024; 20:4649-4662. [PMID: 38877668 PMCID: PMC11247684 DOI: 10.1002/alz.13896] [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: 02/28/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION The entorhinal cortex (EC) and perirhinal cortex (PC) are vulnerable to Alzheimer's disease. A triggering factor may be the interaction of vascular dysfunction and tau pathology. METHODS We imaged post mortem human tissue at 100 μm3 with 7 T magnetic resonance imaging and manually labeled individual blood vessels (mean = 270 slices/case). Vessel density was quantified and compared per EC subfield, between EC and PC, and in relation to tau and TAR DNA-binding protein 43 (TDP-43) semiquantitative scores. RESULTS PC was more vascularized than EC and vessel densities were higher in posterior EC subfields. Tau and TDP-43 strongly correlated with vasculature density and subregions with severe tau at the preclinical stage had significantly greater vessel density than those with low tau burden. DISCUSSION These data impact cerebrovascular maps, quantification of subfield vasculature, and correlation of vasculature and pathology at early stages. The ordered association of vessel density, and tau or TDP-43 pathology, may be exploited in a predictive context. HIGHLIGHTS Vessel density correlates with phosphorylated tau (p-tau) burden in entorhinal and perirhinal cortices. Perirhinal area 35 and posterior entorhinal cortex showed greatest p-tau burden but also the highest vessel density in the preclinical phase of Alzheimer's disease. We combined an ex vivo magnetic resonance imaging model and histopathology to demonstrate the 3D reconstruction of intracortical vessels and its spatial relationship to the pathology.
Collapse
Affiliation(s)
- Josué Llamas Rodríguez
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - André J W van der Kouwe
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jan Oltmer
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Digital Health & Innovation, Vivantes Netzwerk für Gesundheit GmbH, Berlin, Germany
| | - Emma Rosenblum
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Nathaniel Mercaldo
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- MGH Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bruce Fischl
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael Marshall
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew P Frosch
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jean C Augustinack
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| |
Collapse
|
17
|
Mardanyan S, Sharoyan S, Antonyan A. Diversity of amyloid beta peptide actions. Rev Neurosci 2024; 35:387-398. [PMID: 38281140 DOI: 10.1515/revneuro-2023-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/16/2023] [Indexed: 01/30/2024]
Abstract
Fibril formation by amyloidogenic proteins and peptides is considered the cause of a number of incurable diseases. One of the most known amyloid diseases is Alzheimer's disease (AD). Traditionally, amyloidogenic beta peptides Aβ40 and Aβ42 (Aβs) are considered as main causes of AD and the foremost targets in AD fight. The main efforts in pharmacology are aimed at reducing Aβs concentration to prevent their accumulation, aggregation, formation of senile plaques, neuronal death, and neurodegeneration. However, a number of publications have demonstrated certain beneficial physiological effects of Aβs. Simultaneously, it is indicated that the effects of Aβs turn into pathological due to the development of certain diseases in the body. The accumulation of C- and N-terminal truncated Aβs under diverse conditions is supposed to play a role in AD development. The significance of transformation of glutamate residue at positions 3 or 11 of Aβs catalyzed by glutaminyl cyclase making them more degradation resistant, hydrophobic, and prone to aggregation, as well as the participation of dipeptidyl peptidase IV in these transformations are discussed. The experimental data presented confirm the maintenance of physiological, nonaggregated state of Aβs by plant preparations. In conclusion, this review suggests that in the fight against AD, instead of removing Aβs, preference should be given to the treatment of common diseases. Glutaminyl cyclase and dipeptidyl peptidase IV can be considered as targets in AD treatment. Flavonoids and plant preparations that possess antiamyloidogenic propensity are proposed as beneficial neuroprotective, anticancer, and antidiabetic food additives.
Collapse
Affiliation(s)
- Sona Mardanyan
- H. Buniatian Institute of Biochemistry of Armenian National Academy of Sciences, Yerevan 0014, Republic of Armenia
| | - Svetlana Sharoyan
- H. Buniatian Institute of Biochemistry of Armenian National Academy of Sciences, Yerevan 0014, Republic of Armenia
| | - Alvard Antonyan
- H. Buniatian Institute of Biochemistry of Armenian National Academy of Sciences, Yerevan 0014, Republic of Armenia
| |
Collapse
|
18
|
Saleh SR, Abd-Elmegied A, Aly Madhy S, Khattab SN, Sheta E, Elnozahy FY, Mehanna RA, Ghareeb DA, Abd-Elmonem NM. Brain-targeted Tet-1 peptide-PLGA nanoparticles for berberine delivery against STZ-induced Alzheimer's disease in a rat model: Alleviation of hippocampal synaptic dysfunction, Tau pathology, and amyloidogenesis. Int J Pharm 2024; 658:124218. [PMID: 38734273 DOI: 10.1016/j.ijpharm.2024.124218] [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: 02/14/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder that causes severe dementia and memory loss. Surface functionalized poly(lactic-co-glycolic acid) nanoparticles have been reported for better transport through the blood-brain barrier for AD therapy. This study investigated the improved therapeutic potential of berberine-loaded poly(lactic-co-glycolic acid)/Tet-1 peptide nanoparticles (BBR/PLGA-Tet NPs) in a rat model of sporadic AD. BBR was loaded into the PLGA-Tet conjugate. BBR/PLGA-Tet NPs were physicochemically and morphologically characterized. AD was achieved by bilateral intracerebroventricular (ICV) injection of streptozotocin (STZ). Cognitively impaired rats were divided into STZ, STZ + BBR, STZ + BBR/PLGA-Tet NPs, and STZ + PLGA-Tet NPs groups. Cognitive improvement was assessed using the Morris Water Maze. Brain acetylcholinesterase and monoamine oxidase activities, amyloid β42 (Aβ42), and brain glycemic markers were estimated. Further, hippocampal neuroplasticity (BDNF, pCREB, and pERK/ERK), Tau pathogenesis (pGSK3β/GSK3β, Cdk5, and pTau), inflammatory, and apoptotic markers were evaluated. Finally, histopathological changes were monitored. ICV-STZ injection produces AD-like pathologies evidenced by Aβ42 deposition, Tau hyperphosphorylation, impaired insulin signaling and neuroplasticity, and neuroinflammation. BBR and BBR/PLGA-Tet NPs attenuated STZ-induced hippocampal damage, enhanced cognitive performance, and reduced Aβ42, Tau phosphorylation, and proinflammatory responses. BBR/PLGA-Tet NPs restored neuroplasticity, cholinergic, and monoaminergic function, which are critical for cognition and brain function. BBR/PLGA-Tet NPs may have superior therapeutic potential in alleviating sporadic AD than free BBR due to their bioavailability, absorption, and brain uptake.
Collapse
Affiliation(s)
- Samar R Saleh
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Aml Abd-Elmegied
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Somaya Aly Madhy
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Fatma Y Elnozahy
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Radwa A Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Doaa A Ghareeb
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Nihad M Abd-Elmonem
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
19
|
Yang P, Li Y, Qian K, Zhou L, Cheng Y, Wu J, Xu M, Wang T, Yang X, Mu Y, Liu X, Zhang Q. Precise Modulation of Pericyte Dysfunction by a Multifunctional Nanoprodrug to Ameliorate Alzheimer's Disease. ACS NANO 2024; 18:14348-14366. [PMID: 38768086 DOI: 10.1021/acsnano.4c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Pericyte dysfunction severely undermines cerebrovascular integrity and exacerbates neurodegeneration in Alzheimer's disease (AD). However, pericyte-targeted therapy is a yet-untapped frontier for AD. Inspired by the elevation of vascular cell adhesion molecule-1 (VCAM-1) and reactive oxygen species (ROS) levels in pericyte lesions, we fabricated a multifunctional nanoprodrug by conjugating the hybrid peptide VLC, a fusion of the VCAM-1 high-affinity peptide VHS and the neuroprotective apolipoprotein mimetic peptide COG1410, to curcumin (Cur) through phenylboronic ester bond (VLC@Cur-NPs) to alleviate complex pericyte-related pathological changes. Importantly, VLC@Cur-NPs effectively homed to pericyte lesions via VLC and released their contents upon ROS stimulation to maximize their regulatory effects. Consequently, VLC@Cur-NPs markedly increased pericyte regeneration to form a positive feedback loop and thus improved neurovascular function and ultimately alleviated memory defects in APP/PS1 transgenic mice. We present a promising therapeutic strategy for AD that can precisely modulate pericytes and has the potential to treat other cerebrovascular diseases.
Collapse
Affiliation(s)
- Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yongkang Mu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xuan Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| |
Collapse
|
20
|
Niedowicz DM, Gollihue JL, Weekman EM, Phe P, Wilcock DM, Norris CM, Nelson PT. Using digital pathology to analyze the murine cerebrovasculature. J Cereb Blood Flow Metab 2024; 44:595-610. [PMID: 37988134 PMCID: PMC10981399 DOI: 10.1177/0271678x231216142] [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: 06/09/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
Research on the cerebrovasculature may provide insights into brain health and disease. Immunohistochemical staining is one way to visualize blood vessels, and digital pathology has the potential to revolutionize the measurement of blood vessel parameters. These tools provide opportunities for translational mouse model research. However, mouse brain tissue presents a formidable set of technical challenges, including potentially high background staining and cross-reactivity of endogenous IgG. Formalin-fixed paraffin-embedded (FFPE) and fixed frozen sections, both of which are widely used, may require different methods. In this study, we optimized blood vessel staining in mouse brain tissue, testing both FFPE and frozen fixed sections. A panel of immunohistochemical blood vessel markers were tested (including CD31, CD34, collagen IV, DP71, and VWF), to evaluate their suitability for digital pathological analysis. Collagen IV provided the best immunostaining results in both FFPE and frozen fixed murine brain sections, with highly-specific staining of large and small blood vessels and low background staining. Subsequent analysis of collagen IV-stained sections showed region and sex-specific differences in vessel density and vessel wall thickness. We conclude that digital pathology provides a useful tool for relatively unbiased analysis of the murine cerebrovasculature, provided proper protein markers are used.
Collapse
Affiliation(s)
- Dana M Niedowicz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Jenna L Gollihue
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Erica M Weekman
- Stark Neurosciences Research Institute, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Panhavuth Phe
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Donna M Wilcock
- Stark Neurosciences Research Institute, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
21
|
Colvee-Martin H, Parra JR, Gonzalez GA, Barker W, Duara R. Neuropathology, Neuroimaging, and Fluid Biomarkers in Alzheimer's Disease. Diagnostics (Basel) 2024; 14:704. [PMID: 38611617 PMCID: PMC11012058 DOI: 10.3390/diagnostics14070704] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 04/14/2024] Open
Abstract
An improved understanding of the pathobiology of Alzheimer's disease (AD) should lead ultimately to an earlier and more accurate diagnosis of AD, providing the opportunity to intervene earlier in the disease process and to improve outcomes. The known hallmarks of Alzheimer's disease include amyloid-β plaques and neurofibrillary tau tangles. It is now clear that an imbalance between production and clearance of the amyloid beta protein and related Aβ peptides, especially Aβ42, is a very early, initiating factor in Alzheimer's disease (AD) pathogenesis, leading to aggregates of hyperphosphorylation and misfolded tau protein, inflammation, and neurodegeneration. In this article, we review how the AD diagnostic process has been transformed in recent decades by our ability to measure these various elements of the pathological cascade through the use of imaging and fluid biomarkers. The more recently developed plasma biomarkers, especially phosphorylated-tau217 (p-tau217), have utility for screening and diagnosis of the earliest stages of AD. These biomarkers can also be used to measure target engagement by disease-modifying therapies and the response to treatment.
Collapse
Affiliation(s)
- Helena Colvee-Martin
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Juan Rayo Parra
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Gabriel Antonio Gonzalez
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Warren Barker
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| |
Collapse
|
22
|
Pluta R, Czuczwar SJ. Trans- and Cis-Phosphorylated Tau Protein: New Pieces of the Puzzle in the Development of Neurofibrillary Tangles in Post-Ischemic Brain Neurodegeneration of the Alzheimer's Disease-like Type. Int J Mol Sci 2024; 25:3091. [PMID: 38542064 PMCID: PMC10970557 DOI: 10.3390/ijms25063091] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 11/11/2024] Open
Abstract
Recent evidence indicates that experimental brain ischemia leads to dementia with an Alzheimer's disease-like type phenotype and genotype. Based on the above evidence, it was hypothesized that brain ischemia may contribute to the development of Alzheimer's disease. Brain ischemia and Alzheimer's disease are two diseases characterized by similar changes in the hippocampus that are closely related to memory impairment. Following brain ischemia in animals and humans, the presence of amyloid plaques in the extracellular space and intracellular neurofibrillary tangles was revealed. The phenomenon of tau protein hyperphosphorylation is a similar pathological feature of both post-ischemic brain injury and Alzheimer's disease. In Alzheimer's disease, the phosphorylated Thr231 motif in tau protein has two distinct trans and cis conformations and is the primary site of tau protein phosphorylation in the pre-entanglement cascade and acts as an early precursor of tau protein neuropathology in the form of neurofibrillary tangles. Based on the latest publication, we present a similar mechanism of the formation of neurofibrillary tangles after brain ischemia as in Alzheimer's disease, established on trans- and cis-phosphorylation of tau protein, which ultimately influences the development of tauopathy.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland;
| | | |
Collapse
|
23
|
Toribio-Fernandez R, Ceron C, Tristão-Pereira C, Fernandez-Nueda I, Perez-Castillo A, Fernandez-Ferro J, Moro MA, Ibañez B, Fuster V, Cortes-Canteli M. Oral anticoagulants: A plausible new treatment for Alzheimer's disease? Br J Pharmacol 2024; 181:760-776. [PMID: 36633908 DOI: 10.1111/bph.16032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease (AD) and cardiovascular disease (CVD) are strongly associated. Both are multifactorial disorders with long asymptomatic phases and similar risk factors. Indeed, CVD signatures such as cerebral microbleeds, micro-infarcts, atherosclerosis, cerebral amyloid angiopathy and a procoagulant state are highly associated with AD. However, AD and CVD co-development and the molecular mechanisms underlying such associations are not understood. Here, we review the evidence regarding the vascular component of AD and clinical studies using anticoagulants that specifically evaluated the development of AD and other dementias. Most studies reported a markedly decreased incidence of composite dementia in anticoagulated patients with atrial fibrillation, with the highest benefit for direct oral anticoagulants. However, sub-analyses by differential dementia diagnosis were scarce and inconclusive. We finally discuss whether anticoagulation could be a plausible preventive/therapeutic approach for AD and, if so, which would be the best drug and strategy to maximize clinical benefit and minimize potential risks. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Raquel Toribio-Fernandez
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Ceron
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | - Irene Fernandez-Nueda
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Perez-Castillo
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Jose Fernandez-Ferro
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Stroke Unit, Neurology Service, Hospital Universitario Rey Juan Carlos (HURJC), Madrid, Spain
| | - Maria Angeles Moro
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Borja Ibañez
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de enfermedades cardiovasculares (CIBERCV), ISCIII, Madrid, Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marta Cortes-Canteli
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
24
|
Yuyama K, Sun H, Fujii R, Hemmi I, Ueda K, Igeta Y. Extracellular vesicle proteome unveils cathepsin B connection to Alzheimer's disease pathogenesis. Brain 2024; 147:627-636. [PMID: 38071653 PMCID: PMC10834236 DOI: 10.1093/brain/awad361] [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: 04/26/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released extracellularly and considered to be implicated in the pathogenesis of neurodegenerative diseases including Alzheimer's disease. Here, CSF EVs of 16 ATN-classified cases were subjected to quantitative proteome analysis. In these CSF EVs, levels of 11 proteins were significantly altered during the ATN stage transitions (P < 0.05 and fold-change > 2.0). These proteins were thought to be associated with Alzheimer's disease pathogenesis and represent candidate biomarkers for pathogenic stage classification. Enzyme-linked immunosorbent assay analysis of CSF and plasma EVs revealed altered levels of cathepsin B (CatB) during the ATN transition (seven ATN groups in validation set, n = 136). The CSF and plasma EV CatB levels showed a negative correlation with CSF amyloid-β42 concentrations. This proteomic landscape of CSF EVs in ATN classifications can depict the molecular framework of Alzheimer's disease progression, and CatB may be considered a promising candidate biomarker and therapeutic target in Alzheimer's disease amyloid pathology.
Collapse
Affiliation(s)
- Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hui Sun
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Risa Fujii
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Isao Hemmi
- Department of Nursing, Japanese Red Cross College of Nursing, Tokyo 150-0012, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Yukifusa Igeta
- Department of Dementia, Dementia Center, Toranomon Hospital, Tokyo 105-8470, Japan
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, Tokyo 105-8470, Japan
| |
Collapse
|
25
|
Pluta R, Czuczwar SJ. Ischemia-Reperfusion Programming of Alzheimer's Disease-Related Genes-A New Perspective on Brain Neurodegeneration after Cardiac Arrest. Int J Mol Sci 2024; 25:1291. [PMID: 38279289 PMCID: PMC10816023 DOI: 10.3390/ijms25021291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
The article presents the latest data on pathological changes after cerebral ischemia caused by cardiac arrest. The data include amyloid accumulation, tau protein modification, neurodegenerative and cognitive changes, and gene and protein changes associated with Alzheimer's disease. We present the latest data on the dysregulation of genes related to the metabolism of the amyloid protein precursor, tau protein, autophagy, mitophagy, apoptosis, and amyloid and tau protein transport genes. We report that neuronal death after cerebral ischemia due to cardiac arrest may be dependent and independent of caspase. Moreover, neuronal death dependent on amyloid and modified tau protein has been demonstrated. Finally, the results clearly indicate that changes in the expression of the presented genes play an important role in acute and secondary brain damage and the development of post-ischemic brain neurodegeneration with the Alzheimer's disease phenotype. The data indicate that the above genes may be a potential therapeutic target for brain therapy after ischemia due to cardiac arrest. Overall, the studies show that the genes studied represent attractive targets for the development of new therapies to minimize ischemic brain injury and neurological dysfunction. Additionally, amyloid-related genes expression and tau protein gene modification after cerebral ischemia due to cardiac arrest are useful in identifying ischemic mechanisms associated with Alzheimer's disease. Cardiac arrest illustrates the progressive, time- and area-specific development of neuropathology in the brain with the expression of genes responsible for the processing of amyloid protein precursor and the occurrence of tau protein and symptoms of dementia such as those occurring in patients with Alzheimer's disease. By carefully examining the common genetic processes involved in these two diseases, these data may help unravel phenomena associated with the development of Alzheimer's disease and neurodegeneration after cerebral ischemia and may lead future research on Alzheimer's disease or cerebral ischemia in new directions.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland;
| | | |
Collapse
|
26
|
Pluta R, Bogucka-Kocka A, Bogucki J, Kocki J, Czuczwar SJ. Apoptosis, Autophagy, and Mitophagy Genes in the CA3 Area in an Ischemic Model of Alzheimer's Disease with 2-Year Survival. J Alzheimers Dis 2024; 99:1375-1383. [PMID: 38759019 PMCID: PMC11191440 DOI: 10.3233/jad-240401] [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: 04/13/2024] [Indexed: 05/19/2024]
Abstract
Background Currently, no evidence exists on the expression of apoptosis (CASP3), autophagy (BECN1), and mitophagy (BNIP3) genes in the CA3 area after ischemia with long-term survival. Objective The goal of the paper was to study changes in above genes expression in CA3 area after ischemia in the period of 6-24 months. Methods In this study, using quantitative RT-PCR, we present the expression of genes associated with neuronal death in a rat ischemic model of Alzheimer's disease. Results First time, we demonstrated overexpression of the CASP3 gene in CA3 area after ischemia with survival ranging from 0.5 to 2 years. Overexpression of the CASP3 gene was accompanied by a decrease in the activity level of the BECN1 and BNIP3 genes over a period of 0.5 year. Then, during 1-2 years, BNIP3 gene expression increased significantly and coincided with an increase in CASP3 gene expression. However, BECN1 gene expression was variable, increased significantly at 1 and 2 years and was below control values 1.5 years post-ischemia. Conclusions Our observations suggest that ischemia with long-term survival induces neuronal death in CA3 through activation of caspase 3 in cooperation with the pro-apoptotic gene BNIP3. This study also suggests that the BNIP3 gene regulates caspase-independent pyramidal neuronal death post-ischemia. Thus, caspase-dependent and -independent death of neuronal cells occur post-ischemia in the CA3 area. Our data suggest new role of the BNIP3 gene in the regulation of post-ischemic neuronal death in CA3. This suggests the involvement of the BNIP3 together with the CASP3 in the CA3 in neuronal death post-ischemia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Jacek Bogucki
- Faculty of Medicine, Johon Paul II Catholic University of Lublin, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
27
|
Yu B, Wan G, Cheng S, Wen P, Yang X, Li J, Tian H, Gao Y, Zhong Q, Liu J, Li J, Zhu Y. Disruptions of Gut Microbiota are Associated with Cognitive Deficit of Preclinical Alzheimer's Disease: A Cross-Sectional Study. Curr Alzheimer Res 2024; 20:875-889. [PMID: 38529601 DOI: 10.2174/0115672050303878240319054149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most prevalent type of dementia. The early change of gut microbiota is a potential biomarker for preclinical AD patients. OBJECTIVE The study aimed to explore changes in gut microbiota characteristics in preclinical AD patients, including those with Subjective Cognitive Decline (SCD) and Mild Cognitive Impairment (MCI), and detect the correlation between gut microbiota characteristics and cognitive performances. METHODS This study included 117 participants [33 MCI, 54 SCD, and 30 Healthy Controls (HC)]. We collected fresh fecal samples and blood samples from all participants and evaluated their cognitive performance. We analyzed the diversity and structure of gut microbiota in all participants through qPCR, screened characteristic microbial species through machine learning models, and explored the correlations between these species and cognitive performances and serum indicators. RESULTS Compared to the healthy controls, the structure of gut microbiota in MCI and SCD patients was significantly different. The three characteristic microorganisms, including Bacteroides ovatus, Bifidobacterium adolescentis, and Roseburia inulinivorans, were screened based on the best classification model (HC and MCI) having intergroup differences. Bifidobacterium adolescentis is associated with better performance in multiple cognitive scores and several serum indicators. Roseburia inulinivorans showed negative correlations with the scores of the Functional Activities Questionnaire (FAQ). CONCLUSION The gut microbiota in patients with preclinical AD has significantly changed in terms of composition and richness. Correlations have been discovered between changes in characteristic species and cognitive performances. Gut microbiota alterations have shown promise in affecting AD pathology and cognitive deficit.
Collapse
Affiliation(s)
- Binbin Yu
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guomeng Wan
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Shupeng Cheng
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Pengcheng Wen
- Statistics Department, Nanjing Mini Silicon Valley Innovation Group Co., Ltd, Nanjing, China
| | - Xi Yang
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Jiahuan Li
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Huifang Tian
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Yaxin Gao
- Department of Rehabilitation, Suzhou Municipal Hospital, Suzhou, China
| | - Qian Zhong
- Department of Rehabilitation, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jin Liu
- Clinical Medicine Research Institution, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianan Li
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhu
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Wiefels MD, Furar E, Eshraghi RS, Mittal J, Memis I, Moosa M, Mittal R, Eshraghi AA. Targeting Gut Dysbiosis and Microbiome Metabolites for the Development of Therapeutic Modalities for Neurological Disorders. Curr Neuropharmacol 2024; 22:123-139. [PMID: 36200211 PMCID: PMC10716879 DOI: 10.2174/1570159x20666221003085508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/10/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
The gut microbiota, composed of numerous species of microbes, works in synergy with the various organ systems in the body to bolster our overall health and well-being. The most well-known function of the gut microbiome is to facilitate the metabolism and absorption of crucial nutrients, such as complex carbohydrates, while also generating vitamins. In addition, the gut microbiome plays a crucial role in regulating the functioning of the central nervous system (CNS). Host genetics, including specific genes and single nucleotide polymorphisms (SNPs), have been implicated in the pathophysiology of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), and autism spectrum disorder (ASD). The gut microbiome dysbiosis also plays a role in the pathogenesis of these neurodegenerative disorders, thus perturbing the gut-brain axis. Overproduction of certain metabolites synthesized by the gut microbiome, such as short-chain fatty acids (SCFAs) and p-cresyl sulfate, are known to interfere with microglial function and trigger misfolding of alpha-synuclein protein, which can build up inside neurons and cause damage. By determining the association of the gut microbiome and its metabolites with various diseases, such as neurological disorders, future research will pave the way for the development of effective preventive and treatment modalities.
Collapse
Affiliation(s)
- Matthew D Wiefels
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Emily Furar
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rebecca S Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jeenu Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Idil Memis
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Moeed Moosa
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rahul Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Adrien A Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
29
|
Abstract
The deterioration of the brain's microvasculature, particularly in the hippocampus, appears to be a very early event in the development of Alzheimer's disease (AD), preceding even the deposition of amyloid-β. A damaged microvasculature reduces the supply of oxygen and glucose to this region and limits the production of energy, ATP. The damage may be a function of the rise with age in the expression and activity of NADPH oxidase (NOX) in these microvessels. This rise renders these vessels vulnerable to the effects of oxidative stress and inflammation. The rise in NOX activity with age is even more marked in the AD brain where an inverse correlation has been demonstrated between NOX activity and cognitive ability. Apocynin, a putative NOX inhibitor, has been shown to block the damaging effects of NOX activation. Apocynin acts as a strong scavenger of H2O2, and as a weak scavenger of superoxide. Like apocynin, sodium oxybate (SO) has also been shown to block the toxic effects of NOX activation. The application of SO generates NADPH and ATP. SO inhibits oxidative stress and maintains normal cerebral ATP levels under hypoxic conditions. Moreover, it acts epigenetically to attenuate the expression of NOX. SO may delay the onset and slow the progress of AD by suppling energy and maintaining an antioxidative environment in the brain throughout the night. The slow wave activity produced by SO may also activate the glymphatic system and promote the clearance of amyloid-β from the brain.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Poliakova T, Wellington CL. Roles of peripheral lipoproteins and cholesteryl ester transfer protein in the vascular contributions to cognitive impairment and dementia. Mol Neurodegener 2023; 18:86. [PMID: 37974180 PMCID: PMC10652636 DOI: 10.1186/s13024-023-00671-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
This narrative review focuses on the role of cholesteryl ester transfer protein (CETP) and peripheral lipoproteins in the vascular contributions to cognitive impairment and dementia (VCID). Humans have a peripheral lipoprotein profile where low-density lipoproteins (LDL) represent the dominant lipoprotein fraction and high-density lipoproteins (HDL) represent a minor lipoprotein fraction. Elevated LDL-cholesterol (LDL-C) levels are well-established to cause cardiovascular disease and several LDL-C-lowering therapies are clinically available to manage this vascular risk factor. The efficacy of LDL-C-lowering therapies to reduce risk of all-cause dementia and AD is now important to address as recent studies demonstrate a role for LDL in Alzheimer's Disease (AD) as well as in all-cause dementia. The LDL:HDL ratio in humans is set mainly by CETP activity, which exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise LDL and lower HDL as CETP activity increases. Genetic and pharmacological studies support the hypothesis that CETP inhibition reduces cardiovascular risk by lowering LDL, which, by extension, may also lower VCID. Unlike humans, wild-type mice do not express catalytically active CETP and have HDL as their major lipoprotein fraction. As HDL has potent beneficial effects on endothelial cells, the naturally high HDL levels in mice protect them from vascular disorders, likely including VCID. Genetic restoration of CETP expression in mice to generate a more human-like lipid profile may increase the relevance of murine models for VCID studies. The therapeutic potential of existing and emerging LDL-lowering therapies for VCID will be discussed. Figure Legend. Cholesteryl Ester Transfer Protein in Alzheimer's Disease. CETP is mainly produced by the liver, and exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise circulating LDL and lower HDL as CETP activity increases. Low CETP activity is associated with better cardiovascular health, due to decreased LDL and increased HDL, which may also improve brain health. Although most peripheral lipoproteins cannot enter the brain parenchyma due to the BBB, it is increasingly appreciated that direct access to the vascular endothelium may enable peripheral lipoproteins to have indirect effects on brain health. Thus, lipoproteins may affect the cerebrovasculature from both sides of the BBB. Recent studies show an association between elevated plasma LDL, a well-known cardiovascular risk factor, and a higher risk of AD, and considerable evidence suggests that high HDL levels are associated with reduced CAA and lower neuroinflammation. Considering the potential detrimental role of LDL in AD and the importance of HDL's beneficial effects on endothelial cells, high CETP activity may lead to compromised BBB integrity, increased CAA deposits and greater neuroinflammation. Abbreviations: CETP - cholesteryl transfer ester protein; LDL - low-density lipoproteins; HDL - high-density lipoproteins; BBB - blood-brain barrier; CAA - cerebral amyloid angiopathy, SMC - smooth muscle cells, PVM - perivascular macrophages, RBC - red blood cells.
Collapse
Affiliation(s)
- Tetiana Poliakova
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- International Collaboration On Repair Discoveries, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
31
|
Wu W, Huang J, Han P, Zhang J, Wang Y, Jin F, Zhou Y. Research Progress on Natural Plant Molecules in Regulating the Blood-Brain Barrier in Alzheimer's Disease. Molecules 2023; 28:7631. [PMID: 38005352 PMCID: PMC10674591 DOI: 10.3390/molecules28227631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder. With the aging population and the continuous development of risk factors associated with AD, it will impose a significant burden on individuals, families, and society. Currently, commonly used therapeutic drugs such as Cholinesterase inhibitors, N-methyl-D-aspartate antagonists, and multiple AD pathology removal drugs have been shown to have beneficial effects on certain pathological conditions of AD. However, their clinical efficacy is minimal and they are associated with certain adverse reactions. Furthermore, the underlying pathological mechanism of AD remains unclear, posing a challenge for drug development. In contrast, natural plant molecules, widely available, offer multiple targeting pathways and demonstrate inherent advantages in modifying the typical pathologic features of AD by influencing the blood-brain barrier (BBB). We provide a comprehensive review of recent in vivo and in vitro studies on natural plant molecules that impact the BBB in the treatment of AD. Additionally, we analyze their specific mechanisms to offer novel insights for the development of safe and effective targeted drugs as well as guidance for experimental research and the clinical application of drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Weidong Wu
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Jiahao Huang
- Department of Chinese Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Pengfei Han
- Science and Education Section, Zhangjiakou First Hospital, Zhangjiakou 075041, China;
| | - Jian Zhang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Yuxin Wang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Fangfang Jin
- Department of Internal Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yanyan Zhou
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| |
Collapse
|
32
|
Jeong H, Pan Y, Akhter F, Volkow ND, Zhu D, Du C. Impairment of cerebral vascular reactivity and resting blood flow in early-staged transgenic AD mice: in vivo optical imaging studies. RESEARCH SQUARE 2023:rs.3.rs-3579916. [PMID: 37987006 PMCID: PMC10659553 DOI: 10.21203/rs.3.rs-3579916/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder with progressive cognitive decline in aging individuals that poses a significant challenge to patients due to an incomplete understanding of its etiology and lack of effective interventions. While "the Amyloid Cascade Hypothesis," the abnormal accumulation of amyloid-β in the brain, has been the most prevalent theory for AD, mounting evidence from clinical and epidemiological studies suggest that defects in cerebral vessels and hypoperfusion appear prior to other pathological manifestations and might contribute to AD, leading to "the Vascular Hypothesis." However, assessment of structural and functional integrity of the cerebral vasculature in vivo in the brain from AD rodent models has been challenging owing to the limited spatiotemporal resolution of conventional imaging technologies. Methods We employed two in vivo imaging technologies, i.e., Dual-Wavelength Imaging (DWI) and Optical Coherence Tomography (OCT), to evaluate cerebrovascular reactivity (CVR; responsiveness of blood vessels to vasoconstriction as triggered by cocaine) in a relatively large field of view of the cortex in vivo, and 3D quantitative cerebrovascular blood flow (CBF) imaging in living transgenic AD mice at single vessel resolution. Results Our results showed significantly impaired CVR and reduced CBF in basal state in transgenic AD mice compared to non-transgenic littermates in an early stage of AD progression. Changes in total hemoglobin (Δ[HbT]) in response to vasoconstriction were significantly attenuated in AD mice, especially in arteries and tissue, and the recovery time of Δ[HbT] after vasoconstriction was shorter for AD than WT in all types of vessels and cortical tissue, thereby indicating hypoperfusion and reduced vascular flexibility. Additionally, our 3D OCT images revealed that CBF velocities in arteries were slower and that the microvascular network was severely disrupted in the brain of AD mice. Conclusions These results suggest significant vascular impairment in basal CBF and dynamic CVR in the neurovascular network in a rodent model of AD at an early stage of the disease. These cutting-edge in vivo optical imaging tools offer an innovative venue for detecting early neurovascular dysfunction in relation to AD pathology and pave the way for clinical translation of early diagnosis and elucidation of AD pathogenesis in the future.
Collapse
Affiliation(s)
- Hyomin Jeong
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Firoz Akhter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20857, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
33
|
Pastorello Y, Carare RO, Banescu C, Potempa L, Di Napoli M, Slevin M. Monomeric C-reactive protein: A novel biomarker predicting neurodegenerative disease and vascular dysfunction. Brain Pathol 2023; 33:e13164. [PMID: 37158450 PMCID: PMC10580018 DOI: 10.1111/bpa.13164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Circulating C-reactive protein (pCRP) concentrations rise dramatically during both acute (e.g., following stroke) or chronic infection and disease (e.g., autoimmune conditions such as lupus), providing complement fixation through C1q protein binding. It is now known, that on exposure to the membranes of activated immune cells (and microvesicles and platelets), or damaged/dysfunctional tissue, it undergoes lysophosphocholine (LPC)-phospholipase-C-dependent dissociation to the monomeric form (mCRP), concomitantly becoming biologically active. We review histological, immunohistochemical, and morphological/topological studies of post-mortem brain tissue from individuals with neuroinflammatory disease, showing that mCRP becomes stably distributed within the parenchyma, and resident in the arterial intima and lumen, being "released" from damaged, hemorrhagic vessels into the extracellular matrix. The possible de novo synthesis via neurons, endothelial cells, and glia is also considered. In vitro, in vivo, and human tissue co-localization analyses have linked mCRP to neurovascular dysfunction, vascular activation resulting in increased permeability, and leakage, compromise of blood brain barrier function, buildup of toxic proteins including tau and beta amyloid (Aβ), association with and capacity to "manufacture" Aβ-mCRP-hybrid plaques, and, greater susceptibility to neurodegeneration and dementia. Recently, several studies linked chronic CRP/mCRP systemic expression in autoimmune disease with increased risk of dementia and the mechanisms through which this occurs are investigated here. The neurovascular unit mediates correct intramural periarterial drainage, evidence is provided here that suggests a critical impact of mCRP on neurovascular elements that could suggest its participation in the earliest stages of dysfunction and conclude that further investigation is warranted. We discuss future therapeutic options aimed at inhibiting the pCRP-LPC mediated dissociation associated with brain pathology, for example, compound 1,6-bis-PC, injected intravenously, prevented mCRP deposition and associated damage, after temporary left anterior descending artery ligation and myocardial infarction in a rat model.
Collapse
Affiliation(s)
- Ylenia Pastorello
- Department of AnatomyGeorge Emil Palade University of Medicine, Pharmacy, Science and TechnologyTârgu MuresRomania
| | - Roxana O. Carare
- Department of AnatomyGeorge Emil Palade University of Medicine, Pharmacy, Science and TechnologyTârgu MuresRomania
- Clinical and experimental SciencesUniversity of SouthamptonSouthamptonUK
| | - Claudia Banescu
- Department of AnatomyGeorge Emil Palade University of Medicine, Pharmacy, Science and TechnologyTârgu MuresRomania
| | - Lawrence Potempa
- Department of Life Sciences, College of Science, Health and PharmacyRoosevelt UniversitySchaumburgIllinoisUSA
| | - Mario Di Napoli
- Department of Neurology and Stroke UnitSan Camillo de Lellis General HospitalRietiItaly
| | - Mark Slevin
- Department of AnatomyGeorge Emil Palade University of Medicine, Pharmacy, Science and TechnologyTârgu MuresRomania
- Manchester Metropolitan UniversityManchesterUK
| |
Collapse
|
34
|
Tarasova I, Trubnikova O, Kukhareva I, Syrova I, Sosnina A, Kupriyanova D, Barbarash O. A Comparison of Two Multi-Tasking Approaches to Cognitive Training in Cardiac Surgery Patients. Biomedicines 2023; 11:2823. [PMID: 37893196 PMCID: PMC10604887 DOI: 10.3390/biomedicines11102823] [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: 08/21/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The multi-tasking approach may be promising for cognitive rehabilitation in cardiac surgery patients due to a significant effect on attentional and executive functions. This study aimed to compare the neuropsychological changes in patients who have undergone two variants of multi-tasking training and a control group in the early postoperative period of coronary artery bypass grafting (CABG). METHODS One hundred and ten CABG patients were divided into three groups: cognitive training (CT) I (a postural balance task with mental arithmetic, verbal fluency, and divergent tasks) (n = 30), CT II (a simple visual-motor reaction with mental arithmetic, verbal fluency, and divergent tasks) (n = 40), and control (n = 40). RESULTS Two or more cognitive indicators improved in 93.3% of CT I patients, in 72.5% of CT II patients, and in 62.5% of control patients; CT I patients differed from CT II and control (p = 0.04 and p = 0.008, respectively). The improving short-term memory and attention was found more frequently in the CT I group as compared to control (56.7% vs. 15%; p = 0.0005). The cognitive improvement of all domains (psychomotor and executive functions, attention, and short-term memory) was also revealed in CT I patients more frequently than CT II (46.7% vs. 20%; p = 0.02) and control (46.7% vs. 5%; p = 0.0005). CONCLUSIONS The CT I multi-tasking training was more effective at improving the cognitive performance in cardiac surgery patients as compared to CT II training and standard post-surgery management. The findings of this study will be helpful for future studies involving multi-tasking training.
Collapse
Affiliation(s)
- Irina Tarasova
- Department of Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Sosnovy Blvd., 6, 650002 Kemerovo, Russia; (O.T.); (I.S.)
| | | | | | | | | | | | | |
Collapse
|
35
|
Banerjee G, Collinge J, Fox NC, Lashley T, Mead S, Schott JM, Werring DJ, Ryan NS. Clinical considerations in early-onset cerebral amyloid angiopathy. Brain 2023; 146:3991-4014. [PMID: 37280119 PMCID: PMC10545523 DOI: 10.1093/brain/awad193] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/16/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an important cerebral small vessel disease associated with brain haemorrhage and cognitive change. The commonest form, sporadic amyloid-β CAA, usually affects people in mid- to later life. However, early-onset forms, though uncommon, are increasingly recognized and may result from genetic or iatrogenic causes that warrant specific and focused investigation and management. In this review, we firstly describe the causes of early-onset CAA, including monogenic causes of amyloid-β CAA (APP missense mutations and copy number variants; mutations of PSEN1 and PSEN2) and non-amyloid-β CAA (associated with ITM2B, CST3, GSN, PRNP and TTR mutations), and other unusual sporadic and acquired causes including the newly-recognized iatrogenic subtype. We then provide a structured approach for investigating early-onset CAA, and highlight important management considerations. Improving awareness of these unusual forms of CAA amongst healthcare professionals is essential for facilitating their prompt diagnosis, and an understanding of their underlying pathophysiology may have implications for more common, late-onset, forms of the disease.
Collapse
Affiliation(s)
- Gargi Banerjee
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - John Collinge
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, London, W1 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Simon Mead
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - David J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| |
Collapse
|
36
|
Chen L, Zhen Y, Wang X, Wang J, Zhu G. Neurovascular glial unit: A target of phytotherapy for cognitive impairments. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155009. [PMID: 37573807 DOI: 10.1016/j.phymed.2023.155009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/29/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Neurovascular glial unit (NVGU) dysfunction has been reported to be an early and critical event in the pathophysiology of Alzheimer's disease (AD) and vascular dementia (VD). Although herbal medicines, with their favorable safety profiles and low adverse effects, have been suggested to be useful for the treatment of cognitive impairment, the potential role of the NVGU as the target of the effects of herbal medicines is still unclear. PURPOSE This review aimed to retrieve evidence from experimental studies of phytopharmaceuticals targeting the NVGU for the treatment of cognitive impairment in AD and VD, and discussed the potential of phytopharmaceuticals to improve cognitive impairment from the perspective of the NVGU. STUDY DESIGN AND METHODS We systematically searched PubMed, Google Scholar, Web of Science, and CNKI. The keywords used for searching information on the NVGU in the treatment of cognitive impairments included "Alzheimer's disease," "Vascular dementia," "Herbal medicines," "Natural products," "Neurovascular," "Adverse reaction," and "Toxicity, etc." We selected studies on the basis of predefined eligibility criteria. RESULTS NVGU mainly consists of endothelial cells, pericytes, astrocytes, microglia, oligodendrocytes, and neurons, and damage to these cells can induce cognitive impairment by impairing the blood-brain barrier (BBB) and cerebral blood flow (CBF) as well as neuronal function. The active components of herbal medicines, including Ginkgo biloba L., Ginseng Radix et Rhizoma, Epimedium Folium, Chuanxiong Rhizoma, Carthami flos, and Acorus tatarinowii Schott, as well as traditional Chinese medicine prescriptions have shown the potential to improve BBB function and increase CBF to prevent cognitive impairment by inhibiting astrocyte and microglia activation, protecting oligodendrocyte myelin function, reducing neuronal apoptosis, and promoting angiogenesis. CONCLUSIONS Herbal medicines demonstrate great potential to prevent cognitive impairment. Multiple components from herbal medicines may function through different signaling pathways to target the NVGU. Future studies using novel drug-carrier or delivery systems targeting the NVGU will certainly facilitate the development of phytopharmaceuticals for AD and VD.
Collapse
Affiliation(s)
- Lixia Chen
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yilan Zhen
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jingji Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China; The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
37
|
Sharma HS, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma A. Stress induced exacerbation of Alzheimer's disease brain pathology is thwarted by co-administration of nanowired cerebrolysin and monoclonal amyloid beta peptide antibodies with serotonin 5-HT6 receptor antagonist SB-399885. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:3-46. [PMID: 37783559 DOI: 10.1016/bs.irn.2023.05.018] [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: 10/04/2023]
Abstract
Alzheimer's disease is one of the devastating neurodegenerative diseases affecting mankind worldwide with advancing age mainly above 65 years and above causing great misery of life. About more than 7 millions are affected with Alzheimer's disease in America in 2023 resulting in huge burden on health care system and care givers and support for the family. However, no suitable therapeutic measures are available at the moment to enhance quality of life to these patients. Development of Alzheimer's disease may reflect the stress burden of whole life inculcating the disease processes of these neurodegenerative disorders of the central nervous system. Thus, new strategies using nanodelivery of suitable drug therapy including antibodies are needed in exploring neuroprotection in Alzheimer's disease brain pathology. In this chapter role of stress in exacerbating Alzheimer's disease brain pathology is explored and treatment strategies are examined using nanotechnology based on our own investigation. Our observations clearly show that restraint stress significantly exacerbate Alzheimer's disease brain pathology and nanodelivery of a multimodal drug cerebrolysin together with monoclonal antibodies (mAb) to amyloid beta peptide (AβP) together with a serotonin 5-HT6 receptor antagonist SB399885 significantly thwarted Alzheimer's disease brain pathology exacerbated by restraint stress, not reported earlier. The possible mechanisms and future clinical significance is discussed.
Collapse
Affiliation(s)
- 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.
| | - 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, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- 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
| | - 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.
| |
Collapse
|
38
|
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
|
39
|
Cheng J, Wang L, Guttha V, Haugstad G, Kandimalla KK. Delivery of RNA to the Blood-Brain Barrier Endothelium Using Cationic Bicelles. Pharmaceutics 2023; 15:2086. [PMID: 37631300 PMCID: PMC10459289 DOI: 10.3390/pharmaceutics15082086] [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: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction is prevalent in Alzheimer's disease and other neurological disorders. Restoring normal BBB function through RNA therapy is a potential avenue for addressing cerebrovascular changes in these disorders that may lead to cognitive decline. Although lipid nanoparticles have been traditionally used as drug carriers for RNA, bicelles have been emerging as a better alternative because of their higher cellular uptake and superior transfection capabilities. Cationic bicelles composed of DPPC/DC7PC/DOTAP at molar ratios of 63.8/25.0/11.2 were evaluated for the delivery of RNA in polarized hCMEC/D3 monolayers, a widely used BBB cell culture model. RNA-bicelle complexes were formed at five N/P ratios (1:1 to 5:1) by a thin-film hydration method. The RNA-bicelle complexes at N/P ratios of 3:1 and 4:1 exhibited optimal particle characteristics for cellular delivery. The cellular uptake of cationic bicelles laced with 1 mol% DiI-C18 was confirmed by flow cytometry and confocal microscopy. The ability of cationic bicelles (N/P ratio 4:1) to transfect polarized hCMEC/D3 with FITC-labeled control siRNA was tested vis-a-vis commercially available Lipofectamine RNAiMAX. These studies demonstrated the higher transfection efficiency and greater potential of cationic bicelles for RNA delivery to the BBB endothelium.
Collapse
Affiliation(s)
- Joan Cheng
- Department of Pharmaceutics, Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (J.C.); (L.W.); (V.G.)
| | - Lushan Wang
- Department of Pharmaceutics, Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (J.C.); (L.W.); (V.G.)
| | - Vineetha Guttha
- Department of Pharmaceutics, Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (J.C.); (L.W.); (V.G.)
| | - Greg Haugstad
- The Characterization Facility, College of Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Karunya K. Kandimalla
- Department of Pharmaceutics, Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (J.C.); (L.W.); (V.G.)
| |
Collapse
|
40
|
Jullienne A, Szu JI, Quan R, Trinh MV, Norouzi T, Noarbe BP, Bedwell AA, Eldridge K, Persohn SC, Territo PR, Obenaus A. Cortical cerebrovascular and metabolic perturbations in the 5xFAD mouse model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1220036. [PMID: 37533765 PMCID: PMC10392850 DOI: 10.3389/fnagi.2023.1220036] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction The 5xFAD mouse is a popular model of familial Alzheimer's disease (AD) that is characterized by early beta-amyloid (Aβ) deposition and cognitive decrements. Despite numerous studies, the 5xFAD mouse has not been comprehensively phenotyped for vascular and metabolic perturbations over its lifespan. Methods Male and female 5xFAD and wild type (WT) littermates underwent in vivo 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging at 4, 6, and 12 months of age to assess regional glucose metabolism. A separate cohort of mice (4, 8, 12 months) underwent "vessel painting" which labels all cerebral vessels and were analyzed for vascular characteristics such as vessel density, junction density, vessel length, network complexity, number of collaterals, and vessel diameter. Results With increasing age, vessels on the cortical surface in both 5xFAD and WT mice showed increased vessel length, vessel and junction densities. The number of collateral vessels between the middle cerebral artery (MCA) and the anterior and posterior cerebral arteries decreased with age but collateral diameters were significantly increased only in 5xFAD mice. MCA total vessel length and junction density were decreased in 5xFAD mice compared to WT at 4 months. Analysis of 18F-FDG cortical uptake revealed significant differences between WT and 5xFAD mice spanning 4-12 months. Broadly, 5xFAD males had significantly increased 18F-FDG uptake at 12 months compared to WT mice. In most cortical regions, female 5xFAD mice had reduced 18F-FDG uptake compared to WT across their lifespan. Discussion While the 5xFAD mouse exhibits AD-like cognitive deficits as early as 4 months of age that are associated with increasing Aβ deposition, we only found significant differences in cortical vascular features in males, not in females. Interestingly, 5xFAD male and female mice exhibited opposite effects in 18F-FDG uptake. The MCA supplies blood to large portions of the somatosensory cortex and portions of motor and visual cortex and increased vessel length alongside decreased collaterals which coincided with higher metabolic rates in 5xFAD mice. Thus, a potential mismatch between metabolic demand and vascular delivery of nutrients in the face of increasing Aβ deposition could contribute to the progressive cognitive deficits seen in the 5xFAD mouse model.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jenny I. Szu
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Ryan Quan
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Michelle V. Trinh
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Tannoz Norouzi
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Brenda P. Noarbe
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Amanda A. Bedwell
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Kierra Eldridge
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Scott C. Persohn
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Paul R. Territo
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
41
|
Andrade-Guerrero J, Orta-Salazar E, Salinas-Lara C, Sánchez-Garibay C, Rodríguez-Hernández LD, Vargas-Rodríguez I, Barron-Leon N, Ledesma-Alonso C, Diaz-Cintra S, Soto-Rojas LO. Effects of Voluntary Physical Exercise on the Neurovascular Unit in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:11134. [PMID: 37446312 DOI: 10.3390/ijms241311134] [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: 06/17/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Histopathologically, AD presents two pathognomonic hallmarks: (1) neurofibrillary tangles, characterized by intracellular deposits of hyperphosphorylated tau protein, and (2) extracellular amyloid deposits (amyloid plaques) in the brain vasculature (cerebral amyloid angiopathy; CAA). It has been proposed that vascular amyloid deposits could trigger neurovascular unit (NVU) dysfunction in AD. The NVU is composed primarily of astrocytic feet, endothelial cells, pericytes, and basement membrane. Although physical exercise is hypothesized to have beneficial effects against AD, it is unknown whether its positive effects extend to ameliorating CAA and improving the physiology of the NVU. We used the triple transgenic animal model for AD (3xTg-AD) at 13 months old and analyzed through behavioral and histological assays, the effect of voluntary physical exercise on cognitive functions, amyloid angiopathy, and the NVU. Our results show that 3xTg-AD mice develop vascular amyloid deposits which correlate with cognitive deficits and NVU alteration. Interestingly, the physical exercise regimen decreases amyloid angiopathy and correlates with an improvement in cognitive function as well as in the underlying integrity of the NVU components. Physical exercise could represent a key therapeutic approach in cerebral amyloid angiopathy and NVU stability in AD patients.
Collapse
Affiliation(s)
- Jesús Andrade-Guerrero
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Erika Orta-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México 14269, Mexico
| | - Carlos Sánchez-Garibay
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México 14269, Mexico
| | - Luis Daniel Rodríguez-Hernández
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Isaac Vargas-Rodríguez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Nayeli Barron-Leon
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Luis O Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| |
Collapse
|
42
|
Pluta R, Miziak B, Czuczwar SJ. Post-Ischemic Permeability of the Blood-Brain Barrier to Amyloid and Platelets as a Factor in the Maturation of Alzheimer's Disease-Type Brain Neurodegeneration. Int J Mol Sci 2023; 24:10739. [PMID: 37445917 DOI: 10.3390/ijms241310739] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this review is to present evidence of the impact of ischemic changes in the blood-brain barrier on the maturation of post-ischemic brain neurodegeneration with features of Alzheimer's disease. Understanding the processes involved in the permeability of the post-ischemic blood-brain barrier during recirculation will provide clinically relevant knowledge regarding the neuropathological changes that ultimately lead to dementia of the Alzheimer's disease type. In this review, we try to distinguish between primary and secondary neuropathological processes during and after ischemia. Therefore, we can observe two hit stages that contribute to Alzheimer's disease development. The onset of ischemic brain pathology includes primary ischemic neuronal damage and death followed by the ischemic injury of the blood-brain barrier with serum leakage of amyloid into the brain tissue, leading to increased ischemic neuronal susceptibility to amyloid neurotoxicity, culminating in the formation of amyloid plaques and ending in full-blown dementia of the Alzheimer's disease type.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
43
|
Muñoz-Castro C, Mejias-Ortega M, Sanchez-Mejias E, Navarro V, Trujillo-Estrada L, Jimenez S, Garcia-Leon JA, Fernandez-Valenzuela JJ, Sanchez-Mico MV, Romero-Molina C, Moreno-Gonzalez I, Baglietto-Vargas D, Vizuete M, Gutierrez A, Vitorica J. Monocyte-derived cells invade brain parenchyma and amyloid plaques in human Alzheimer's disease hippocampus. Acta Neuropathol Commun 2023; 11:31. [PMID: 36855152 PMCID: PMC9976401 DOI: 10.1186/s40478-023-01530-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Microglia are brain-resident myeloid cells and play a major role in the innate immune responses of the CNS and the pathogenesis of Alzheimer's disease (AD). However, the contribution of nonparenchymal or brain-infiltrated myeloid cells to disease progression remains to be demonstrated. Here, we show that monocyte-derived cells (MDC) invade brain parenchyma in advanced stages of AD continuum using transcriptional analysis and immunohistochemical characterization in post-mortem human hippocampus. Our findings demonstrated that a high proportion (60%) of demented Braak V-VI individuals was associated with up-regulation of genes rarely expressed by microglial cells and abundant in monocytes, among which stands the membrane-bound scavenger receptor for haptoglobin/hemoglobin complexes or Cd163. These Cd163-positive MDC invaded the hippocampal parenchyma, acquired a microglial-like morphology, and were located in close proximity to blood vessels. Moreover, and most interesting, these invading monocytes infiltrated the nearby amyloid plaques contributing to plaque-associated myeloid cell heterogeneity. However, in aged-matched control individuals with hippocampal amyloid pathology, no signs of MDC brain infiltration or plaque invasion were found. The previously reported microglial degeneration/dysfunction in AD hippocampus could be a key pathological factor inducing MDC recruitment. Our data suggest a clear association between MDC infiltration and endothelial activation which in turn may contribute to damage of the blood brain barrier integrity. The recruitment of monocytes could be a consequence rather than the cause of the severity of the disease. Whether monocyte infiltration is beneficial or detrimental to AD pathology remains to be fully elucidated. These findings open the opportunity to design targeted therapies, not only for microglia but also for the peripheral immune cell population to modulate amyloid pathology and provide a better understanding of the immunological mechanisms underlying the progression of AD.
Collapse
Affiliation(s)
- Clara Muñoz-Castro
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Marina Mejias-Ortega
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Elisabeth Sanchez-Mejias
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Victoria Navarro
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Laura Trujillo-Estrada
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Sebastian Jimenez
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Juan Antonio Garcia-Leon
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Juan Jose Fernandez-Valenzuela
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Maria Virtudes Sanchez-Mico
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Carmen Romero-Molina
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Ines Moreno-Gonzalez
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - David Baglietto-Vargas
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Marisa Vizuete
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Antonia Gutierrez
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain. .,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| | - Javier Vitorica
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain. .,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain. .,Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
44
|
Sheikh AM, Yano S, Tabassum S, Mitaki S, Michikawa M, Nagai A. Alzheimer's Amyloid β Peptide Induces Angiogenesis in an Alzheimer's Disease Model Mouse through Placental Growth Factor and Angiopoietin 2 Expressions. Int J Mol Sci 2023; 24:ijms24054510. [PMID: 36901941 PMCID: PMC10003449 DOI: 10.3390/ijms24054510] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Increased angiogenesis, especially the pathological type, has been documented in Alzheimer's disease (AD) brains, and it is considered to be activated due to a vascular dysfunction-mediated hypoxic condition. To understand the role of the amyloid β (Aβ) peptide in angiogenesis, we analyzed its effects on the brains of young APP transgenic AD model mice. Immunostaining results revealed that Aβ was mainly localized intracellularly, with very few immunopositive vessels, and there was no extracellular deposition at this age. Solanum tuberosum lectin staining demonstrated that compared to their wild-type littermates, the vessel number was only increased in the cortex of J20 mice. CD105 staining also showed an increased number of new vessels in the cortex, some of which were partially positive for collagen4. Real-time PCR results demonstrated that placental growth factor (PlGF) and angiopoietin 2 (AngII) mRNA were increased in both the cortex and hippocampus of J20 mice compared to their wild-type littermates. However, vascular endothelial growth factor (VEGF) mRNA did not change. Immunofluorescence staining confirmed the increased expression of PlGF and AngII in the cortex of the J20 mice. Neuronal cells were positive for PlGF and AngII. Treatment of a neural stem cell line (NMW7) with synthetic Aβ1-42 directly increased the expression of PlGF and AngII, at mRNA levels, and AngII at protein levels. Thus, these pilot data indicate that pathological angiogenesis exists in AD brains due to the direct effects of early Aβ accumulation, suggesting that the Aβ peptide regulates angiogenesis through PlGF and AngII expression.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Correspondence: (A.M.S.); (A.N.); Tel.: +81-0853-20-2306 (A.M.S.); +81-0853-20-2198 (A.N.)
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shatera Tabassum
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Atsushi Nagai
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Correspondence: (A.M.S.); (A.N.); Tel.: +81-0853-20-2306 (A.M.S.); +81-0853-20-2198 (A.N.)
| |
Collapse
|
45
|
Qiu Y, Mo C, Xu S, Chen L, Ye W, Kang Y, Chen G, Zhu T. Research progress on perioperative blood-brain barrier damage and its potential mechanism. Front Cell Dev Biol 2023; 11:1174043. [PMID: 37101615 PMCID: PMC10124715 DOI: 10.3389/fcell.2023.1174043] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
The blood-brain barrier (BBB) is an important barrier separating the central nervous system from the periphery. The composition includes endothelial cells, pericytes, astrocytes, synapses and tight junction proteins. During the perioperative period, anesthesia and surgical operations are also a kind of stress to the body, which may be accompanied by blood-brain barrier damage and brain metabolism dysfunction. Perioperative blood-brain barrier destruction is closely associated with cognitive impairment and may increase the risk of postoperative mortality, which is not conducive to enhanced recovery after surgery. However, the potential pathophysiological process and specific mechanism of blood-brain barrier damage during the perioperative period have not been fully elucidated. Changes in blood-brain barrier permeability, inflammation and neuroinflammation, oxidative stress, ferroptosis, and intestinal dysbiosis may be involved in blood-brain barrier damage. We aim to review the research progress of perioperative blood-brain barrier damage and its potential adverse effects and potential molecular mechanisms, and provide ideas for the study of homeostasis maintenance of brain function and precision anesthesia.
Collapse
Affiliation(s)
- Yong Qiu
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shiyu Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lu Chen
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wanlin Ye
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Kang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Guo Chen
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Guo Chen, ; Tao Zhu,
| | - Tao Zhu
- Department of Anesthesiology, National Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012), West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Guo Chen, ; Tao Zhu,
| |
Collapse
|
46
|
Sousa JA, Bernardes C, Bernardo-Castro S, Lino M, Albino I, Ferreira L, Brás J, Guerreiro R, Tábuas-Pereira M, Baldeiras I, Santana I, Sargento-Freitas J. Reconsidering the role of blood-brain barrier in Alzheimer's disease: From delivery to target. Front Aging Neurosci 2023; 15:1102809. [PMID: 36875694 PMCID: PMC9978015 DOI: 10.3389/fnagi.2023.1102809] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The existence of a selective blood-brain barrier (BBB) and neurovascular coupling are two unique central nervous system vasculature features that result in an intimate relationship between neurons, glia, and blood vessels. This leads to a significant pathophysiological overlap between neurodegenerative and cerebrovascular diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease whose pathogenesis is still to be unveiled but has mostly been explored under the light of the amyloid-cascade hypothesis. Either as a trigger, bystander, or consequence of neurodegeneration, vascular dysfunction is an early component of the pathological conundrum of AD. The anatomical and functional substrate of this neurovascular degeneration is the BBB, a dynamic and semi-permeable interface between blood and the central nervous system that has consistently been shown to be defective. Several molecular and genetic changes have been demonstrated to mediate vascular dysfunction and BBB disruption in AD. The isoform ε4 of Apolipoprotein E is at the same time the strongest genetic risk factor for AD and a known promoter of BBB dysfunction. Low-density lipoprotein receptor-related protein 1 (LRP-1), P-glycoprotein, and receptor for advanced glycation end products (RAGE) are examples of BBB transporters implicated in its pathogenesis due to their role in the trafficking of amyloid-β. This disease is currently devoid of strategies that change the natural course of this burdening illness. This unsuccess may partly be explained by our misunderstanding of the disease pathogenesis and our inability to develop drugs that are effectively delivered to the brain. BBB may represent a therapeutic opportunity as a target itself or as a therapeutic vehicle. In this review, we aim to explore the role of BBB in the pathogenesis of AD including the genetic background and detail how it can be targeted in future therapeutic research.
Collapse
Affiliation(s)
- João André Sousa
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Catarina Bernardes
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sara Bernardo-Castro
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Albino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Miguel Tábuas-Pereira
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
47
|
Joseph CR, Kreilach A, Reyna VA, Kepler TA, Taylor BV, Kang J, McCorkle D, Rider NL. Utilizing Reduced Labeled Proton Clearance to Identify Preclinical Alzheimer Disease with 3D ASL MRI. Case Rep Neurol 2023; 15:177-186. [PMID: 37901133 PMCID: PMC10603764 DOI: 10.1159/000530980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/02/2023] [Indexed: 10/31/2023] Open
Abstract
Addressing the seminal pathophysiology in Alzheimer disease (AD) is the next logical focus for effective intervention, given the initial disappointing and more recent possibly encouraging results of monoclonal antibody trials. Endothelial cell dysfunction-induced blood-brain barrier leak with associated prolonged capillary mean transit time (cMTT) and glymphatic outflow dysfunction is the most proximal events in the degeneration cascade. Sensitive and reproducible markers are required to both identify early disease and assess future treatment trial outcomes. Two participants, with mild cognitive impairment (MCI) and one with AD, were evaluated clinically prior to MRI in this small case series report. From seven 3D turbo gradient and spin echo (TGSE) pulsed arterial spin echo (PASL) MRI sequences six homologous region of interest in bitemporal, bifrontal, and biparietal lobes for each sequence were examined and plotted against time. By choosing late perfusion times during cMTT phase of perfusion linear analysis of signal decay could be utilized. A reference axial FLAIR sequence was also obtained. Slope of the linear analysis correlated to the rate of labeled proton clearance with reduced clearance occurring in AD participants compared to normal participants in our previous study. Whether similar differences in clearance rate extend to either MCI or early AD was investigated. Participants were categorized by clinical phenotype before MRI and compared to previously published phenotype cohorts: n = 18 normal/healthy, n = 6 AD, n = 3 MCI. Significant differences in labeled proton clearance rates between AD and MCI/control phenotypes within bilateral temporal lobes (left p = 0.004, right p = 0.002) and within bilateral frontal lobes AD versus controls (left p = 0.001, right p = 0.008) and AD versus MCI (left p = 0.001, right p = 0.001) were found. This noninvasive MRI technique has potential for identifying MCI transition to AD.
Collapse
Affiliation(s)
- Charles R. Joseph
- Department of Neurology, Liberty University College of Osteopathic Medicine (LUCOM) Lynchburg, VA, USA
| | | | | | | | | | - Jubin Kang
- LUCOM medical student, Lynchburg, VA, USA
| | | | - Nicholas L. Rider
- Department of Bioinformatics and Immunology, LUCOM, Lynchburg, VA, USA
| |
Collapse
|
48
|
Li W, Jiang J, Zou X, Zhang Y, Sun M, Jia Z, Li W, Xu J. The characteristics of arterial spin labeling cerebral blood flow in patients with subjective cognitive decline: The Chinese imaging, biomarkers, and lifestyle study. Front Neurosci 2022; 16:961164. [PMID: 35983224 PMCID: PMC9379247 DOI: 10.3389/fnins.2022.961164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveWe aimed to characterize the potential risk factors and cerebral perfusion of patients with subjective cognitive decline (SCD).MethodsThis prospective study enrolled consecutive patients from the Chinese Imaging, Biomarkers, and Lifestyle (CIBL) Cohort of Alzheimer’s disease between February 2021 and March 2022. Patients who met the SCD diagnostic criteria were categorized into the SCD group, while those without cognitive complaints or any concerns were assigned to the healthy control (HC) group. The demographic and clinical characteristics and cerebral blood flow (CBF) from pseudo-continuous arterial spin labeling (pCASL) in standard cognitive regions were compared between these two groups. A multivariate analysis was performed to identify independent factors associated with SCD.ResultsThe frequency of family history of dementia in the SCD group was higher compared with the HC group (p = 0.016). The CBF of left hippocampus (p = 0.023), left parahippocampal gyrus (p = 0.004), left precuneus (p = 0.029), left middle temporal gyrus (p = 0.022), right parahippocampal gyrus (p = 0.018), and right precuneus (p = 0.024) in the SCD group were significantly increased than those in the HC group. The multivariate logistic regression analyses demonstrated that the family history of dementia [OR = 4.284 (1.096–16.747), p = 0.036] and the CBF of left parahippocampal gyrus [OR = 1.361 (1.006–1.840), p = 0.045] were independently associated with SCD.ConclusionThis study demonstrated that the family history of dementia and the higher CBF within the left parahippocampal gyrus were independent risk factors associated with patients with SCD, which could help in the early identification of the SCD and in intervening during this optimal period.
Collapse
Affiliation(s)
- Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- *Correspondence: Jun Xu,
| |
Collapse
|