1
|
Mohammadi S, Dolatshahi M, Rahmani F, Raji CA. Altered Clearance in Alzheimer's Disease and Cerebral Amyloid Angiopathy. Neuroimaging Clin N Am 2025; 35:277-286. [PMID: 40210383 DOI: 10.1016/j.nic.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
With our focus on Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA), the authors herein provided a comprehensive overview of impaired amyloid beta and tau clearance pathways observed through advanced neuroimaging techniques such as dynamic contrast-enhanced MR imaging, arterial spin labeling, phase contrast MR imaging, PET, and functional MR imaging. The findings suggest the role of impaired degradation clearance, blood-brain barrier clearance, perivascular clearance, glymphatic system clearance, and cerebrospinal fluid dynamics in AD and CAA pathogenesis.
Collapse
Affiliation(s)
- Soheil Mohammadi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Bankolé A, Srivastava A, Shihavuddin A, Tighanimine K, Faucourt M, Koka V, Weill S, Nemazanyy I, Nelson AJ, Stokes MP, Delgehyr N, Genovesio A, Meunier A, Fumagalli S, Pende M, Spassky N. mTOR controls ependymal cell differentiation by targeting the alternative cell cycle and centrosomal proteins. EMBO Rep 2025:10.1038/s44319-025-00460-2. [PMID: 40307619 DOI: 10.1038/s44319-025-00460-2] [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: 10/21/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Ependymal cells are multiciliated glial cells lining the ventricles of the mammalian brain. Their differentiation from progenitor cells involves cell enlargement and progresses through centriole amplification phases and ciliogenesis. These phases are accompanied by the sharp up-regulation of mTOR Complex 1 activity (mTORC1), a master regulator of macromolecule biosynthesis and cell growth, whose function in ependymal cell differentiation is unknown. We demonstrate that mTORC1 inhibition by rapamycin preserves the progenitor pool by reinforcing quiescence and preventing alternative cell cycle progression for centriole amplification. Overexpressing E2F4 and MCIDAS circumvents mTORC1-regulated processes, enabling centriole amplification despite rapamycin, and enhancing mTORC1 activity through positive feedback. Acute rapamycin treatment in multicentriolar cells during the late phases of differentiation causes centriole regrouping, indicating a direct role of mTORC1 in centriole dynamics. By phosphoproteomic and phosphomutant analysis, we reveal that the mTORC1-mediated phosphorylation of GAS2L1, a centrosomal protein that links actin and microtubule cytoskeletons, participates in centriole disengagement. This multilayered and sequential control of ependymal development by mTORC1, from the progenitor pool to centriolar function, has implications for pathophysiological conditions like aging and hydrocephalus-prone genetic diseases.
Collapse
Affiliation(s)
- Alexia Bankolé
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Asm Shihavuddin
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
- Department of EEE, Presidency University, Dhaka, Bangladesh
| | - Khaled Tighanimine
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Vonda Koka
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Solene Weill
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Alissa J Nelson
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Matthew P Stokes
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Auguste Genovesio
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Stefano Fumagalli
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Mario Pende
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France.
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France.
| |
Collapse
|
3
|
Bai W, Chang T, Halike K, Li J, Huang X, Zhang X, Wang J. Cerebrospinal Fluid Biomarkers in Idiopathic Normal Pressure Hydrocephalus: Bibliometric Analysis and Literature Review. World Neurosurg 2025; 198:123981. [PMID: 40250527 DOI: 10.1016/j.wneu.2025.123981] [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: 02/07/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is a progressive neurological disorder characterized by normal intracranial pressure, ventriculomegaly, gait disturbances, cognitive impairment, and urinary dysfunction, with an unknown etiology and predominantly affecting elderly populations. iNPH frequently coexists with Alzheimer disease (AD), and differentiating dementia-subtype iNPH from AD remains clinically challenging. In this review, we demonstrate that cerebrospinal fluid biomarkers may aid differential diagnosis, correlate with cognitive impairment, and predict shunt surgery outcomes. The cerebrospinal fluid biomarkers investigated include AD-associated markers (amyloid-β and tau proteins). Our analysis integrates bibliometric methodologies to map research trends. Advancements in understanding amyloid-β and tau pathology may offer novel diagnostic and therapeutic strategies for iNPH.
Collapse
Affiliation(s)
- Wenju Bai
- Department of Neurosurgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Tengwu Chang
- Department of Neurosurgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Kamiran Halike
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jinyong Li
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaoyuan Huang
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xv Zhang
- Department of Systematic Anatomy, Xinjiang Second Medical College, Karamay, Xinjiang, China
| | - Jichao Wang
- Department of Neurosurgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China.
| |
Collapse
|
4
|
Mondal R, Deb S, Shome G, Sarkar V, Lahiri D, Datta SS, Benito-León J. Molecular dynamics of amyloid-β transport in Alzheimer's disease: Exploring therapeutic plasma exchange with albumin replacement - Current insights and future perspectives. Neurologia 2025; 40:306-328. [PMID: 40280630 DOI: 10.1016/j.nrleng.2025.03.008] [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: 05/04/2023] [Accepted: 11/07/2023] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The complex process of amyloid-β (Aβ) transportation across the blood-brain and blood-cerebrospinal fluid barriers is crucial for preventing Aβ accumulation, which linked to dementia and neurodegeneration. This review explores therapeutic plasma exchange with albumin replacement in Alzheimer's disease, based on the dynamics of amyloid-β between the brain, plasma, and cerebrospinal fluid. METHODOLOGY A comprehensive literature review was conducted using PubMed/Medline, Cochrane Library, and open databases (bioRxiv, MedRixv, preprint.org) up to April 30, 2023. The first search utilized the following MeSH terms and keywords: 'Plasma Exchange', 'Plasmapheresis', 'Therapeutic plasma exchange', 'Apheresis', 'Aβ', 'p-tau', 'Total-tau', 'Alzheimer's disease', 'Cognitive dysfunction', 'neurodegenerative diseases', 'centrifugation', 'membranous', and 'filtration' in the Title/Abstract, yielding 146 results. A second search with the keywords: 'Albumin', 'Aβ', 'BBB', 'Alzheimer's dementia', and 'Nerve degeneration' resulted in 125 additional articles for analysis. Finally, a third search using keywords: 'Albumin structural domains', 'Albumin-Aβ interactions', 'Albumin-endothelial interactions', and 'Post-Translational Modification' produced 193 results for further review. RESULTS/DISCUSSION Therapeutic plasma exchange shows potential as a disease-modifying therapy for dementia, specifically for Alzheimer's disease. Additionally, the promising role of albumin supplementation in cognitive improvement has attracted attention. However, clinical evidence supporting therapeutic plasma exchange for dementia remains limited, necessitating further research and development to mitigate potential adverse effects. A deeper understanding of the molecular dynamics of Aβ transportation and the mechanisms of therapeutic plasma exchange is essential. A critical evaluation of existing evidence highlights the importance of balancing potential benefits with associated risks, which will guide the development and application of these treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- R Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - S Deb
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - G Shome
- Department of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - V Sarkar
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - D Lahiri
- Baycrest Academy of Research and Education, Toronto, Canada; Rotman Research Institute, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Canada; Department of Neurology, Institute of Neurosciences, Kolkata, India
| | - S S Datta
- Department of Transfusion Medicine, Tata Medical Center, Kolkata 700160, India
| | - J Benito-León
- Department of Neurology, University Hospital "12 de Octubre", Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
5
|
Yamada S, Yuzawa T, Ito H, Iseki C, Kondo T, Yamanaka T, Tanikawa M, Otani T, Ii S, Ohta Y, Watanabe Y, Wada S, Oshima M, Mase M. Regional brain volume changes in Hakim's disease versus Alzheimer's and mild cognitive impairment. Brain Commun 2025; 7:fcaf122. [PMID: 40235958 PMCID: PMC11997787 DOI: 10.1093/braincomms/fcaf122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/16/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
Idiopathic normal-pressure hydrocephalus (Hakim's disease) is characterized by ventricular enlargement and disproportionately enlarged subarachnoid space hydrocephalus, leading to localized brain deformation. Differentiating regional brain volume changes in Hakim's disease from those in Alzheimer's disease, Hakim's disease with Alzheimer's disease, and mild cognitive impairment provides insights into disease-specific mechanisms. This study aimed to identify disease-specific patterns of brain volume changes in Hakim's disease, Alzheimer's disease, Hakim's disease with Alzheimer's disease, and mild cognitive impairment and compare them with those in cognitively healthy individuals using an advanced artificial intelligence-based brain segmentation tool. The study included 970 participants, comprising 52 patients with Hakim's disease, 256 with Alzheimer's disease, 25 with Hakim's disease with Alzheimer's disease, 163 with mild cognitive impairment, and 474 healthy controls. The intracranial spaces were segmented into 100 brain and 7 CSF subregions from 3D T1-weighted MRIs using brain subregion analysis. The volume ratios of these regions were compared among the groups using Glass's Δ, referencing 400 healthy controls aged ≥50 years. Hakim's disease exhibited significant volume reduction in the supramarginal gyrus of the parietal lobe and the paracentral gyrus of the frontal lobe. Alzheimer's disease exhibited prominent volume loss in the hippocampus and temporal lobe, particularly in the entorhinal cortex, fusiform gyrus, and inferior temporal gyrus. Hakim's disease with Alzheimer's disease showed significant volume reductions in the supramarginal gyrus of the parietal lobe, similar to Hakim's disease, whereas temporal lobe volumes were relatively preserved compared with those in Alzheimer's disease. Patients with mild cognitive impairment aged ≥70 years had comparable regional brain volume ratios with healthy controls in the same age group. The Hakim's disease and Hakim's disease with Alzheimer's disease groups were characterized by volume reductions in the frontal and parietal lobes caused by disproportionately enlarged subarachnoid space hydrocephalus-related compression compared with temporal lobe atrophy observed in the Alzheimer's disease group. These disease-specific morphological changes highlight the need for longitudinal studies to clarify the causes of compression and atrophy.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, 467-8601, Japan
- Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, 153-8505, Japan
| | - Takuya Yuzawa
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, 107-0052, Japan
| | - Hirotaka Ito
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, 107-0052, Japan
| | - Chifumi Iseki
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
- Division of Neurology and Clinical Neuroscience, Department of Internal Medicine III, Yamagata University School of Medicine, Yamagata, 990-9585, Japan
| | - Toshiyuki Kondo
- Division of Neurology and Clinical Neuroscience, Department of Internal Medicine III, Yamagata University School of Medicine, Yamagata, 990-9585, Japan
| | - Tomoyasu Yamanaka
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, 467-8601, Japan
| | - Motoki Tanikawa
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, 467-8601, Japan
| | - Tomohiro Otani
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, 560-8531, Japan
| | - Satoshi Ii
- Department of Mechanical Engineering, School of Engineering, Institute of Science Tokyo, Tokyo, 145-0061, Japan
| | - Yasuyuki Ohta
- Division of Neurology and Clinical Neuroscience, Department of Internal Medicine III, Yamagata University School of Medicine, Yamagata, 990-9585, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Shigeo Wada
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, 560-8531, Japan
| | - Marie Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, 153-8505, Japan
| | - Mitsuhito Mase
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, 467-8601, Japan
| |
Collapse
|
6
|
Deng Z, Wang H, Zhong K, Li Y, Deng H, Gao B, Huang K, Tong A, Zhou L. The Role of Choroid Plexus in Hydrocephalus from the Perspective of Structure and Function: a Therapeutic Target. Mol Neurobiol 2025:10.1007/s12035-025-04823-7. [PMID: 40085357 DOI: 10.1007/s12035-025-04823-7] [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: 10/08/2024] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Hydrocephalus is one of the most common neurological diseases, characterized by abnormal excessive accumulation of cerebrospinal fluid (CSF) in the ventricular system. Its pathophysiological mechanism is believed to be related to the imbalance of CSF circulation and homeostasis. As the main source of CSF secretion, the choroid plexus is closely related to hydrocephalus. The choroid plexus is a specialized vascularized tissue located within the cerebral ventricles. It has multiple physiological functions including regulating CSF, immune response, endocrine metabolism, etc. Strategies that reduce choroid plexus CSF secretion have been shown to be effective in the treatment of hydrocephalus. However, the role of other physiological functions of the choroid plexus in hydrocephalus is still unclear. Recent studies on the choroid plexus and the blood-CSF barrier have deepened our understanding of the structure and function of the choroid plexus. The idea of targeting the choroid plexus to treat hydrocephalus has spawned many branches: choroid plexus epithelial cells, choroid plexus immune cells, choroid plexus peptides, and choroid plexus cilia, etc. This review introduces the basic structure and function of the choroid plexus, summarizes their changes in hydrocephalus, and analyzes the possibility of the choroid plexus as a therapeutic target for hydrocephalus.
Collapse
Affiliation(s)
- Ziang Deng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haoxiang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyou Li
- Department of Pediatric Neurosurgery, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Huajiang Deng
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Sichuan Province, Luzhou City, China
| | - Baocheng Gao
- Department of Neurosurgery, The First People'S Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology) Kunming, Yunan, China
| | - Keru Huang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China.
- Department of Neurosurgery, The Fifth People's Hospital of Ningxia, Shizuishan, China.
| |
Collapse
|
7
|
Bateman GA, Bateman AR. Brain Ischemia in Alzheimer's Disease May Partly Counteract the Disruption of the Blood-Brain Barrier. Brain Sci 2025; 15:269. [PMID: 40149790 PMCID: PMC11940560 DOI: 10.3390/brainsci15030269] [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: 02/11/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND In normal pressure hydrocephalus (NPH) there is blood-brain barrier (BBB) disruption, which should increase the CSF formation rate (CSFfr) and, therefore, also increase the intracranial pressure (ICP). However, the ICP is normal in NPH. A lumped parameter study was performed to look at the interrelation between the ICP, cerebral blood flow (CBF), and the degree of BBB disruption in NPH. The model suggested that the CSFfr could be reduced in this condition if the BBB disruption was moderated by a reduction in the capillary transmural pressure (TMP) secondary to arteriolar constriction and a reduced CBF. In early Alzheimer's disease (AD), there is BBB disruption, reduced ICP, and global ischemia. This raises the possibility that the same physiology may occur in AD as occurs in NPH. METHODS A lumped parameter model previously used to describe the hydrodynamics of NPH was modified to investigate the effects of changes in CSF pressure and blood flow in patients with mild cognitive impairment (MCI) and AD. RESULTS The model indicates that the average capillary TMP is normal in MCI, but decreases as AD progresses. Removing CSF in AD patients during a tap test initially increases the capillary TMP. The brain in AD responds to a tap test by increasing its level of ischemia, and this reduces the capillary TMP. CONCLUSIONS A hypothesis is put forward that the BBB disruption in AD is partially mitigated by the brain making itself ischemic. Modelling gives support to this hypothesis. The model can suggest a cause for the development of ischemic neuronal loss and amyloid accumulation secondary to glymphatic flow disruption as AD progresses.
Collapse
Affiliation(s)
- Grant A. Bateman
- Department of Medical Imaging, John Hunter Hospital, Newcastle, NSW 2310, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, Newcastle University, Callaghan Campus, Newcastle, NSW 2308, Australia
| | - Alexander R. Bateman
- School of Engineering, College of Engineering, Science and Environment, Newcastle University, Callaghan Campus, Newcastle, NSW 2308, Australia;
| |
Collapse
|
8
|
Ko AWK, Abdelmonem A, Taheri MR. Arachnoid granulations: Dynamic nature and review. Curr Probl Diagn Radiol 2025; 54:265-272. [PMID: 39676028 DOI: 10.1067/j.cpradiol.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Arachnoid granulations have been known for centuries yet remain incompletely understood. While traditionally associated with cerebrospinal fluid transport, the precise mechanism remains uncertain. This manuscript reviews the literature on the anatomy, histology, and imaging findings of arachnoid granulations and their mimickers and anomalous variations. We highlight variations in incidence, size, and characteristics of arachnoid granulations on imaging, and hypothesize that these variations may be explained by arachnoid granulations being dynamic secondary to varying functionality. We review the pathophysiologic role of arachnoid granulations in pathologies related to hydrocephalus, neurodegenerative disorders, and intracranial hypertension and hypotension. A further understanding of arachnoid granulations, their mechanism in cerebrospinal fluid transport, and change over time may provide a basis for future imaging markers and therapies.
Collapse
Affiliation(s)
- Andrew Wai Kei Ko
- Department of Radiology, George Washington University Hospital, 900 23rd St NW, Washington, DC 20037, USA.
| | - Ahmed Abdelmonem
- Department of Radiology, George Washington University Hospital, 900 23rd St NW, Washington, DC 20037, USA.
| | - M Reza Taheri
- Department of Radiology, George Washington University Hospital, 900 23rd St NW, Washington, DC 20037, USA.
| |
Collapse
|
9
|
Li T, Wang Q, Yang B, Qu X, Chen W, Wang H, Wang N, Xian J. Glymphatic system impairment in normal tension glaucoma evaluated by diffusion tensor image analysis along the perivascular space. Brain Res 2025; 1850:149450. [PMID: 39793917 DOI: 10.1016/j.brainres.2025.149450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Disruption of the glymphatic system plays a vital role in pathogenesis of neurodegeneration in normal tension glaucoma (NTG). We evaluated the impairment of glymphatic system of NTG patients by diffusion tensor image analysis along the perivascular space (DTI-ALPS), and explored the correlation between the ALPS index and dysfunction of visual cortices in resting state. DTI-ALPS was applied to 37 normal controls (NCs) and 37 NTG patients. Multidirectional diffusivity maps and fractional anisotropy (FA) maps were reconstructed to calculate ALPS index. The Amplitude of low-frequency fluctuation (ALFF) in visual cortices (V1-V5) were calculated using resting-state fMRI. Clinical data and ALPS indexes were compared between the groups. Lateralization of ALPS indexes and differences in visual field of two eyes were analyzed. Subsequently, regression analyses between ALPS indexes and mean deviation (MD) values of bilateral eyes and ALFF of visual cortices were performed. The bilateral ALPS indexes of NTG patients decreased significantly. In NCs and NTG patients, ALPS indexes in right hemisphere were lower than that in left hemisphere. The right ALPS indexes of NTG patients were positively correlated with the MD values of the left eyes. In NTG patients, decreased ALFF was detected in right V1 and bilateral V2-5, and the left ALPS indexes were positively correlated with ALFF in bilateral V1, V2, V5, and right V3V area. The ALPS index decreased in NTG patients, correlated with visual defects and ALFF, indicating impairment of the glymphatic system and the potential to be a biomarker in the future.
Collapse
Affiliation(s)
- Ting Li
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qian Wang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Bingbing Yang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaoxia Qu
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Weiwei Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing Tongren Hospital, Capital Medical University, China
| | - Huaizhou Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing Tongren Hospital, Capital Medical University, China
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing Tongren Hospital, Capital Medical University, China.
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Zampogna A, Patera M, Falletti M, Pinola G, Asci F, Suppa A. Technological Advances for Gait and Balance in Normal Pressure Hydrocephalus: A Systematic Review. Bioengineering (Basel) 2025; 12:135. [PMID: 40001656 PMCID: PMC11852021 DOI: 10.3390/bioengineering12020135] [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: 12/23/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Normal pressure hydrocephalus (NPH) is a recognized cause of reversible cognitive and motor decline, with gait and balance impairments often emerging early. Technologies providing gait and balance measures can aid in early detection, diagnosis, and prognosis of the disease. This systematic review comprehensively discusses previous studies on the instrumental assessment of gait and balance in NPH. A PubMed search following PRISMA guidelines identified studies published between 2000 and 2024 that used laboratory instruments to assess gait and balance in NPH. Studies underwent quality assessment for internal, statistical, and external validity. Methodological details such as motor tasks, instruments, analytical approaches, and main findings were summarized. Overall, this review includes 41 studies on gait and 17 on balance, most of which used observational, cross-sectional designs. These studies employed various tools, such as pressure-sensitive platforms, optoelectronic motion-capture systems, and wearable inertial sensors. Significant differences in kinematic measures of gait and balance have been found in NPH patients compared to healthy controls and individuals with other neurological conditions. Finally, this review explores potential pathophysiological mechanisms underlying the kinematic changes in gait and balance in NPH and emphasizes the absence of longitudinal data, which hinders drawing definitive conclusions for prognostic purposes.
Collapse
Affiliation(s)
- Alessandro Zampogna
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
- IRCCS Neuromed Institute, 86077 Pozzilli, Italy
| | - Martina Patera
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
| | - Marco Falletti
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
| | - Giulia Pinola
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
| | - Francesco Asci
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
| | - Antonio Suppa
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (A.Z.); (M.P.); (M.F.); (G.P.); (F.A.)
- IRCCS Neuromed Institute, 86077 Pozzilli, Italy
| |
Collapse
|
11
|
Mravinacová S, Bergström S, Olofsson J, de San José NG, Anderl-Straub S, Diehl-Schmid J, Fassbender K, Fliessbach K, Jahn H, Kornhuber J, Landwehrmeyer GB, Lauer M, Levin J, Ludolph AC, Prudlo J, Schneider A, Schroeter ML, Wiltfang J, Steinacker P, Otto M, Nilsson P, Månberg A. Addressing inter individual variability in CSF levels of brain derived proteins across neurodegenerative diseases. Sci Rep 2025; 15:668. [PMID: 39753643 PMCID: PMC11698900 DOI: 10.1038/s41598-024-83281-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Accurate diagnosis and monitoring of neurodegenerative diseases require reliable biomarkers. Cerebrospinal fluid (CSF) proteins are promising candidates for reflecting brain pathology; however, their diagnostic utility may be compromised by natural variability between individuals, weakening their association with disease. Here, we measured the levels of 69 pre-selected proteins in cerebrospinal fluid using antibody-based suspension bead array technology in a multi-disease cohort of 499 individuals with neurodegenerative disorders including Alzheimer's disease (AD), behavioral variant frontotemporal dementia, primary progressive aphasias, amyotrophic lateral sclerosis (ALS), corticobasal syndrome, primary supranuclear palsy, along with healthy controls. We identify significant inter-individual variability in overall CSF levels of brain-derived proteins, which could not be attributed to specific disease associations. Using linear modelling, we show that adjusting for median CSF levels of brain-derived proteins increases the diagnostic accuracy of proteins previously identified as altered in CSF in the context of neurodegenerative disorders. We further demonstrate a simplified approach for the adjustment using pairs of correlated proteins with opposite alteration in the diseases. With this approach, the proteins adjust for each other and further increase the biomarker performance through additive effect. When comparing the diseases, two proteins-neurofilament medium and myelin basic protein-showed increased levels in ALS compared to other diseases, and neurogranin showed a specific increase in AD. Several other proteins showed similar trends across the studied diseases, indicating that these proteins likely reflect shared processes related to neurodegeneration. Overall, our findings suggest that accounting for inter-individual variability is crucial in future studies to improve the identification and performance of relevant biomarkers. Importantly, we highlight the need for multi-disease studies to identify disease-specific biomarkers.
Collapse
Affiliation(s)
- Sára Mravinacová
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Sofia Bergström
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jennie Olofsson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | | | - Janine Diehl-Schmid
- Department of Psychiatry, Technical University of Munich, Munich, Germany
- Kbo-Inn-Salzach-Klinikum Gemeinnützige GmbH, Wasserburg Am Inn, Germany
| | | | - Klaus Fliessbach
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn and DZNE Bonn, Bonn, Germany
| | - Holger Jahn
- Department of Psychiatry, University Hospital, Hamburg, Germany
| | - Johannes Kornhuber
- Department of Psychiatry, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | - Martin Lauer
- Center for Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Albert C Ludolph
- Department of Neurology, University Hospital Ulm (UKU), Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany
| | - Johannes Prudlo
- Rostock University Medical Center and German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Anja Schneider
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn and DZNE Bonn, Bonn, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University Clinic Leipzig, and Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, and DZNE, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Petra Steinacker
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Markus Otto
- Department of Neurology, University Hospital Ulm (UKU), Ulm, Germany
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Nilsson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Månberg
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
12
|
Sun Z, Li C, Muccio M, Jiang L, Masurkar A, Buch S, Chen Y, Zhang J, Haacke EM, Wisniewski T, Ge Y. Vascular Aging in the Choroid Plexus: A 7T Ultrasmall Superparamagnetic Iron Oxide (USPIO)-MRI Study. J Magn Reson Imaging 2024; 60:2564-2575. [PMID: 38587279 PMCID: PMC11458823 DOI: 10.1002/jmri.29381] [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: 01/31/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND The choroid plexus (ChP), a densely vascularized structure, has drawn increasing attention for its involvement in brain homeostasis and waste clearance. While the volumetric changes have been explored in many imaging studies, few studies have investigated the vascular degeneration associated with aging in the ChP. PURPOSE To investigate the sub-structural characteristics of the ChP, particularly the vascular compartment using high-resolution 7T imaging enhanced with Ferumoxytol, an ultrasmall super-paramagnetic iron oxide, which greatly increase the susceptibility contrast for vessels. STUDY TYPE Prospective. SUBJECTS Forty-nine subjects without neurological disorders (age: 21-80 years; 42 ± 17 years; 20 females). FIELD STRENGTH/SEQUENCE 7-T with 2D and 3D T2* GRE, 3D MPRAGE T1, 2D TSE T2, and 2D FLAIR. ASSESSMENT The vascular and stromal compartments of the ChP were segmented using K-means clustering on post-contrast 2D GRE images. Visual and qualitative assessment of ChP vascular characteristics were conducted independently by three observers. Vascular density (Volvessel/VolChP ratio) and susceptibility change (Δχ) induced by Ferumoxytol were analyzed on 3D GRE-derived susceptibility-weighted imaging and quantitative susceptibility mapping, respectively. STATISTICAL TESTS Independent t-test, Mann-Whitney U test, and Chi-square test were utilized for group comparisons. The relationship between age and ChP's vascular alterations was examined using Pearson's correlation. Intra-class coefficient was calculated for inter-observer agreement. A P value <0.05 was considered statistically significant. RESULTS 2D GRE images demonstrated superior contrast and accurate delineation of ChP substructures (ICC = 0.86). Older subjects exhibited a significantly smaller vascular density (16.5 ± 4.34%) and lower Δχ (22.10 ± 12.82 ppb) compared to younger subjects (24.85 ± 6.84% and 34.64 ± 12.69 ppb). Vascular density and mean Δχ within the ChP negatively correlated with age (r = -0.48, and r = -0.45). DATA CONCLUSION Ferumoxytol-enhanced 7T images can demonstrate ChP alterations in elderly with decreased vascular density and expansion of nonvascular compartment. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
- Vilcek Institute of Graduate Medical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Chenyang Li
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
- Vilcek Institute of Graduate Medical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Marco Muccio
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Li Jiang
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Arjun Masurkar
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sagar Buch
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Yongsheng Chen
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Jiangyang Zhang
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - E. Mark Haacke
- Department of Radiology, Wayne State University, Detroit, MI, USA
| | - Thomas Wisniewski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Departments of Pathology and Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Yulin Ge
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
13
|
Bruno MK, Dhall R, Duquette A, Haq IU, Honig LS, Lamotte G, Mari Z, McFarland NR, Montaser-Kouhsari L, Rodriguez-Porcel F, Shurer J, Siddiqui J, Spears CC, Wills AMA, Diaz K, Golbe LI. A General Neurologist's Practical Diagnostic Algorithm for Atypical Parkinsonian Disorders: A Consensus Statement. Neurol Clin Pract 2024; 14:e200345. [PMID: 39185098 PMCID: PMC11341009 DOI: 10.1212/cpj.0000000000200345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/16/2024] [Indexed: 08/27/2024]
Abstract
Purpose of Review The most common four neurodegenerative atypical parkinsonian disorders (APDs) are progressive supranuclear palsy (PSP), multiple system atrophy (MSA), corticobasal syndrome (CBS), and dementia with Lewy bodies (DLB). Their formal diagnostic criteria often require subspecialty experience to implement as designed and all require excluding competing diagnoses without clearly specifying how to do that. Validated diagnostic criteria are not available at all for many of the other common APDs, including normal pressure hydrocephalus (NPH), vascular parkinsonism (VP), or drug-induced parkinsonism (DIP). APDs also include conditions of structural, genetic, vascular, toxic/metabolic, infectious, and autoimmune origin. Their differential diagnosis can be challenging early in the course, if the presentation is atypical, or if a rare or non-neurodegenerative condition is present. This review equips community general neurologists to make an early provisional diagnosis before, or in place of, referral to a tertiary center. Early diagnosis would allay diagnostic uncertainty, allow prompt symptomatic management, provide disease-specific information and support resources, avoid further pointless testing and treatments, and create the possibility of trial referral. Recent Findings We address 64 APDs using one over-arching flow diagram and a series of detailed tables. Most instances of APDs can be diagnosed with a careful history and neurological exam, along with a non-contrast brain MRI. Additional diagnostic tests are rarely needed but are delineated where applicable. Our diagnostic algorithm encourages referral to a tertiary center whenever the general neurologist feels it would be in the patient's best interest. Our algorithm emphasizes that the diagnosis of APDs is an iterative process, refined with the appearance of new diagnostic features, availability of new technology, and advances in scientific understanding of the disorders. Clinicians' proposals for all diagnostic tests for the APDs, including repeat visits, should be discussed with patients and their families to ensure that the potential information to be gained aligns with their larger clinical goals. Summary We designed this differential diagnostic algorithm for the APDs to enhance general neurologists' diagnostic skills and confidence and to help them address the less common or more ambiguous cases.
Collapse
Affiliation(s)
- Michiko K Bruno
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Rohit Dhall
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Antoine Duquette
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Ihtsham U Haq
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Lawrence S Honig
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Guillaume Lamotte
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Zoltan Mari
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Nikolaus R McFarland
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Leila Montaser-Kouhsari
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Federico Rodriguez-Porcel
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Jessica Shurer
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Junaid Siddiqui
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Christopher C Spears
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Anne-Marie A Wills
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Kristophe Diaz
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Lawrence I Golbe
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
14
|
Mishra KA, Sethi KK. Unveiling tomorrow: Carbonic anhydrase activators and inhibitors pioneering new frontiers in Alzheimer's disease. Arch Pharm (Weinheim) 2024:e2400748. [PMID: 39506506 DOI: 10.1002/ardp.202400748] [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: 09/16/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and a principal basis of dementia in the elderly population globally. Recently, human carbonic anhydrases (hCAs, EC 4.2.1.1) were demonstrated as possible new targets for treating AD. hCAs are vital for maintaining pH balance and performing other physiological processes as they catalyze the reversible hydration of carbon dioxide to bicarbonate and a proton. Current research indicates that hCA plays a role in brain functions critical for transmitting neural signals. Activation of carbonic anhydrase (CA) has emerged as a promising avenue in addressing memory loss and cognitive issues. Conversely, the exploration of CA inhibition represents a novel frontier in this field. By enhancing glial fitness and cerebrovascular health and blocking amyloid-β (Aβ)-induced mitochondrial dysfunction pathways, cytochrome C (CytC) release, caspase 9 activation, and H2O2 generation in neurons, CA inhibitors improve cognition and lessen the pathology caused by Aβ. Recent research has pushed hCAs into the spotlight as critical players in AD pathogenesis and precise therapeutic targets. The captivating dilemma of choosing between hCA inhibitors and activators looms large, as inhibitors reduce Aβ aggregation and improve cerebral blood flow, while activators enhance cerebrovascular functions and restore pH balance. The current review sheds light on the clinical evidence for hCAs and the roles of inhibitors and activators in AD. Additionally, this review offers a fascinating outlook on the data that may aid medicinal chemists in designing and developing new leads that are more effective and selective for upcoming in vitro and in vivo studies, allowing for the discovery and introduction of novel drug candidates for the treatment of AD to the market and into the clinical pipeline.
Collapse
Affiliation(s)
- Km Abha Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| | - Kalyan K Sethi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| |
Collapse
|
15
|
Farke D, Olszewska A, Büttner K, Schmidt MJ. Association among raised intraventricular pressure, clinical signs, and magnetic resonance imaging findings in dogs with congenital internal hydrocephalus. J Vet Intern Med 2024; 38:3119-3128. [PMID: 39482254 PMCID: PMC11586545 DOI: 10.1111/jvim.17235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Dogs with internal hydrocephalus do not necessarily have high intraventricular pressure (IVP). HYPOTHESIS/OBJECTIVES Not all reported MRI findings indicate high IVP and some clinical signs might be associated with elevated IVP and syringomyelia. ANIMALS Fifty-three dogs. MATERIALS AND METHODS Cross-sectional study. Clinical signs and MRI findings were evaluated for an association of IVP >12 mm Hg and syringomyelia. RESULTS High IVP was associated with obtundation OR 4.64 (95% CI 1.27-16.93) (P = .02), head tilt OR 6.42 (95% CI 1.08-37.97) (P = .04) and nystagmus OR 8.24 (95% CI 1.44-47.07) (P = .02). Pain was associated with syringomyelia OR 3.4 (95% CI 0.98-11.78) (P = .05). The number of affected ventricles was associated with high IVP OR 2.85 (95% CI 0.97-8.33) (P = .05) and syringomyelia OR 12.74 (95% CI 2.93-55.4) (P = .0007). Periventricular edema OR 24.46 (95% CI 4.54-131.77), OR 7.61 (95% CI 1.91-30.32) (P < .0002, P = .004) and signal void sign OR 17.34 (95% CI 4.01-74.95), OR 4.18 (95% CI 1.16-15.02) (P < .0001, P = .03) were associated with high IVP and syringomyelia. The probability for syringomyelia is lower with disruption of the internal capsule OR 0.19 (95% CI 0.05-0.72) (P = .01) and higher VBR OR 0.25 (95% CI 0.1-0.63) (P = .004). CONCLUSIONS AND CLINICAL IMPORTANCE Previously reported MRI findings are not predictive of high IVP. Clinical signs and MRI findings should be used to make a diagnosis of internal hydrocephalus in dogs with or without high IVP.
Collapse
Affiliation(s)
- Daniela Farke
- Department of Veterinary Clinical Sciences, Small Animal ClinicJustus‐Liebig‐University, Frankfurter Strasse 108Giessen 35392Germany
| | - Agnieszka Olszewska
- Department of Veterinary Clinical Sciences, Small Animal ClinicJustus‐Liebig‐University, Frankfurter Strasse 108Giessen 35392Germany
| | - Kathrin Büttner
- Unit for Biomathematics and Data Processing, Faculty of Veterinary MedicineJustus Liebig‐University‐GiessenGiessenGermany
| | - Martin J. Schmidt
- Department of Veterinary Clinical Sciences, Small Animal ClinicJustus‐Liebig‐University, Frankfurter Strasse 108Giessen 35392Germany
| |
Collapse
|
16
|
Hu Y, Cao C, Li M, He H, Luo L, Guo Y. Association between idiopathic normal pressure hydrocephalus and Alzheimer's disease: a bidirectional Mendelian randomization study. Sci Rep 2024; 14:22744. [PMID: 39349954 PMCID: PMC11443089 DOI: 10.1038/s41598-024-72559-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Observational studies have suggested a bidirectional relationship between idiopathic normal pressure hydrocephalus (iNPH) and Alzheimer's disease (AD). However, the causal association between these two neurodegenerative disorders remains unclear. This study aimed to explore the causal relationship between iNPH and AD using a two-sample bidirectional Mendelian randomization (MR) method. Large-scale genome-wide association studies of iNPH (Ncase = 767, Ncontrol = 375,610) and AD (Ncase/proxy = 111,326, Ncontrol = 677,663) in European individuals were used to screen genetic instruments for MR analysis. Inverse variance-weighted (IVW) method was used as the main analysis, other MR methods and a series of sensitivity analyses were performed to ensure the reliability. In the forward MR analysis, genetic predisposition to iNPH had no effects on the risk of AD development. Likewise, in the reverse MR analysis, AD did not demonstrate a significant causal effect on iNPH. Sensitivity analyses bolstered the reliability of the MR results. Our MR study indicated no genetic evidence supporting a suggestive association between AD and iNPH in either direction, and provided evidence on the dichotomy between true iNPH and neurodegenerative NPH.
Collapse
Affiliation(s)
- Yuanjun Hu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Chengan Cao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manting Li
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Haiyong He
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Lun Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
17
|
Sadanandan J, Sathyanesan M, Newton SS. Aging alters the expression of trophic factors and tight junction proteins in the mouse choroid plexus. Fluids Barriers CNS 2024; 21:77. [PMID: 39334352 PMCID: PMC11438291 DOI: 10.1186/s12987-024-00574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The choroid plexus (CP) is an understudied tissue in the central nervous system and is primarily implicated in cerebrospinal fluid (CSF) production. CP also produces numerous neurotrophic factors (NTF) which circulate to different brain regions. Regulation of NTFs in the CP during natural aging is largely unknown. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and the water channel protein Aquaporin (AQP1). METHODS Male and female mice were used for our study. Age-related transcriptional changes were analyzed using quantitative PCR at three different time points: mature adult, middle-aged, and aged. Transcriptional changes during aging were further confirmed with digital droplet PCR. Additionally, we used immunohistochemical analysis (IHC) for the evaluation of in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP, and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, Plectin. RESULTS Aging significantly altered NTF gene expression in the CP. Brain-derived neurotrophic factor (BDNF), Midkine (MDK), VGF, Insulin-like growth factor (IGF1), IGF2, Klotho (KL), Erythropoietin (EPO), and its receptor (EPOR) were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression was unchanged in the aged CP, while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2 and CLAUDIN5 were reduced in aged mice. CONCLUSIONS Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
18
|
Pearce RKB, Gontsarova A, Richardson D, Methley AM, Watt HC, Tsang K, Carswell C. Shunting for idiopathic normal pressure hydrocephalus. Cochrane Database Syst Rev 2024; 8:CD014923. [PMID: 39105473 PMCID: PMC11301990 DOI: 10.1002/14651858.cd014923.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
BACKGROUND Normal pressure hydrocephalus (NPH) occurs when the brain ventricles expand, causing a triad of gait, cognitive, and urinary impairment. It can occur after a clear brain injury such as trauma, but can also occur without a clear cause (termed idiopathic, or iNPH). Non-randomised studies have shown a benefit from surgically diverting ventricular fluid to an area of lower pressure by cerebrospinal fluid (CSF)-shunting in iNPH, but historically there have been limited randomised controlled trial (RCT) data to confirm this. OBJECTIVES To determine the effect of CSF-shunting versus no CSF-shunting in people with iNPH and the frequency of adverse effects of CSF-shunting in iNPH. SEARCH METHODS We searched the Cochrane Dementia and Cognitive Improvement Group's register, Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (Ovid SP), Embase (Ovid SP), PsycINFO (Ovid SP), CINAHL (EBSCOhost), Web of Science Core Collection (Clarivate), LILACS (BIREME), ClinicalTrials.gov, and the World Health Organization International Clinical Trials Registry Platform on 15 February 2023. SELECTION CRITERIA We included only RCTs of people who had symptoms of gait, cognitive, or urinary impairment with communicating hydrocephalus (Evans index of > 0.3) and normal CSF pressure. Control groups included those with no CSF shunts or those with CSF shunts that were in 'inactive' mode. DATA COLLECTION AND ANALYSIS We used standard Cochrane methodological procedures. Where necessary, we contacted study authors requesting data not provided in the papers. We assessed the overall certainty of the evidence using GRADE. MAIN RESULTS We included four RCTs, of which three were combined in a meta-analysis. The four RCTs included 140 participants (73 with immediate CSF-shunting and 67 controls who had delayed CSF-shunting) with an average age of 75 years. Risk of bias was low in all parallel-group outcomes evaluated apart from gait speed, cognitive function (general cognition and Symbol Digit Test) (some concerns) and adverse events, which were not blind-assessed. CSF-shunting probably improves gait speed at less than six months post-surgery (standardised mean difference (SMD) 0.62, 95% confidence interval (CI) 0.24 to 0.99; 3 studies, 116 participants; moderate-certainty evidence). CSF-shunting may improve qualitative gait function at less than six months post-surgery by an uncertain amount (1 study, 88 participants; low-certainty evidence). CSF-shunting probably results in a large reduction of disability at less than six months post-surgery (risk ratio 2.08, 95% CI 1.31 to 3.31; 3 studies, 118 participants; moderate-certainty evidence). The evidence is very uncertain about the effect of CSF-shunting on cognitive function at less than six months post-CSF-shunt surgery (SMD 0.35, 95% CI -0.04 to 0.74; 2 studies, 104 participants; very low-certainty evidence). The evidence is also very uncertain about the effect of CSF-shunt surgery on adverse events (1 study, 88 participants; very low-certainty evidence). There were no data regarding the effect of CSF-shunting on quality of life. AUTHORS' CONCLUSIONS We found moderate-certainty evidence that CSF-shunting likely improves gait speed and disability in iNPH in the relative short term. The evidence is very uncertain regarding cognition and adverse events. There were no longer-term RCT data for any of our prespecified outcomes. More studies are required to improve the certainty of these findings. In addition, more information is required regarding patient ethnicity and the effect of CSF-shunting on quality of life.
Collapse
Affiliation(s)
- Ronald K B Pearce
- Department of Neurology, Imperial College Healthcare NHS Trust, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Davina Richardson
- Department of Neurophysiotherapy, Imperial College Healthcare NHS Trust, London, UK
| | - Abigail M Methley
- Department of Clinical Neuropsychology, North Staffordshire Combined Healthcare NHS Trust, Stoke-On-Trent, UK
| | - Hilary Clare Watt
- Department of Primary Care and Public Health, Imperial College London, London, UK
| | - Kevin Tsang
- Department of Neurosurgery, Imperial College Healthcare NHS Trust, London, UK
| | - Christopher Carswell
- Department of Neurology, Imperial College Healthcare NHS Trust, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
19
|
Ho K, Bodi NE, Sharma TP. Normal-Tension Glaucoma and Potential Clinical Links to Alzheimer's Disease. J Clin Med 2024; 13:1948. [PMID: 38610712 PMCID: PMC11012506 DOI: 10.3390/jcm13071948] [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: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Glaucoma is a group of optic neuropathies and the world's leading cause of irreversible blindness. Normal-tension glaucoma (NTG) is a subtype of glaucoma that is characterized by a typical pattern of peripheral retinal loss, in which the patient's intraocular pressure (IOP) is considered within the normal range (<21 mmHg). Currently, the only targetable risk factor for glaucoma is lowering IOP, and patients with NTG continue to experience visual field loss after IOP-lowering treatments. This demonstrates the need for a better understanding of the pathogenesis of NTG and underlying mechanisms leading to neurodegeneration. Recent studies have found significant connections between NTG and cerebral manifestations, suggesting NTG as a neurodegenerative disease beyond the eye. Gaining a better understanding of NTG can potentially provide new Alzheimer's Disease diagnostics capabilities. This review identifies the epidemiology, current biomarkers, altered fluid dynamics, and cerebral and ocular manifestations to examine connections and discrepancies between the mechanisms of NTG and Alzheimer's Disease.
Collapse
Affiliation(s)
- Kathleen Ho
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Nicole E. Bodi
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tasneem P. Sharma
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
20
|
Sadanandan J, Sathyanesan M, Newton SS. Regulation of trophic factors in the choroid plexus of aged mice. RESEARCH SQUARE 2024:rs.3.rs-4123786. [PMID: 38562722 PMCID: PMC10984084 DOI: 10.21203/rs.3.rs-4123786/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: 04/04/2024]
Abstract
Background The choroid plexus (CP) is an understudied tissue in the central nervous system (CNS), primarily implicated in cerebrospinal fluid (CSF) production. Additionally, CP produces numerous neurotrophic factors (NTF), which circulate to different regions of the brain. Regulation of NTF in the CP during natural aging has yet to be discovered. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and water channel protein Aquaporin (AQP1). Methods We used male and female mice for our study. We analyzed neurotrophic factor gene expression patterns using quantitative and digital droplet PCR at three different time points: mature adult, middle-aged, and aged. Additionally, we used immunohistochemical analysis (IHC) to evaluate in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, plectin. Results Aging significantly altered the NTF's gene expression in the CP Brain-derived neurotrophic factor (BDNF), Midkine, VGF, Insulin-like growth factor (IGF1), IGF2, klotho, Erythropoietin, and its receptor were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression is unchanged in the aged CP while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2, and CLAUDIN5 were reduced in aged mice. Conclusions Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
|
21
|
Tseng PH, Huang WT, Wang JH, Huang BR, Huang HY, Tsai ST. Cerebrospinal fluid shunt surgery reduces the risk of developing dementia and Alzheimer's disease in patients with idiopathic normal pressure hydrocephalus: a nationwide population-based propensity-weighted cohort study. Fluids Barriers CNS 2024; 21:16. [PMID: 38355601 PMCID: PMC10868070 DOI: 10.1186/s12987-024-00517-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Patients with idiopathic normal-pressure hydrocephalus (iNPH) are predisposed to developing dementing disorders. Cerebrospinal fluid (CSF) shunt implantation is a treatment used to improve the motor and cognitive disabilities of these patients; however, its effect on the risk of developing dementing disorders remains unclear. We conducted a population-based propensity-weighted cohort study to investigate whether CSF shunt surgery may reduce the risk of subsequently developing dementia, Alzheimer's disease (AD), and vascular dementia in iNPH patients. METHODS Patients aged ≥ 60 years who were diagnosed with iNPH (n = 2053) between January 2001 and June 2018 were identified from the Taiwan National Health Insurance Research Database. Various demographic characteristics (age, sex, and monthly income) and clinical data (incidence year, comorbidities, and Charlson comorbidity index) were collected and divided into the shunt surgery group (SSG) and the non-shunt surgery group (NSSG). Stabilized inverse probability of treatment weighting by using the propensity score was performed to achieve a balanced distribution of confounders across the two study groups. The cumulative incidence rate and risk of dementing disorders were estimated during a 16-year follow-up period. RESULTS After weighting, the data of 375.0 patients in SSG and 1677.4 patients in NSSG were analyzed. Kaplan-Meier curve analysis indicated that the cumulative incidence rate of AD (p = 0.009), but not dementia (p = 0.241) and vascular dementia (p = 0.761), in SSG was significantly lower than that in NSSG over the 16-year follow-up period. Cox proportional hazards regression analysis revealed that SSG had a reduced hazard ratio (HR) for developing AD [HR (95% CI) 0.17 (0.04-0.69)], but not for dementia [HR (95% CI) 0.83 (0.61-1.12)] and vascular dementia [HR (95% CI) 1.18 (0.44-3.16)], compared with NSSG. Further Fine-Gray hazard regression analysis with death as a competing event demonstrated that SSG had a reduced subdistribution HR (sHR) for developing dementia [sHR (95% CI) 0.74 (0.55-0.99)] and AD [sHR (95% CI) 0.15 (0.04-0.61)], but not for vascular dementia [sHR (95% CI) 1.07 (0.40-2.86)]. CONCLUSION CSF shunt surgery is associated with reduced risks of the subsequent development of dementia and AD in iNPH patients. Our findings may provide valuable information for assessing the benefit-to-risk profile of CSF shunt surgery.
Collapse
Affiliation(s)
- Pao-Hui Tseng
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
- Department of Nursing, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970, Taiwan
| | - Wan-Ting Huang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Sec 3, Zhongyang Road, Hualien, 970, Taiwan
| | - Jen-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Sec 3, Zhongyang Road, Hualien, 970, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Hsin-Yi Huang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Sec 3, Zhongyang Road, Hualien, 970, Taiwan.
| | - Sheng-Tzung Tsai
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan.
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970, Taiwan.
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan.
| |
Collapse
|
22
|
Yang X, Gan J, Ji Y. Association between cerebrospinal fluid pressure and cognition in patients with Alzheimer's disease and Lewy body dementia. BMC Neurol 2024; 24:35. [PMID: 38243235 PMCID: PMC10797877 DOI: 10.1186/s12883-023-03502-1] [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: 04/09/2023] [Accepted: 12/09/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The relationship between cerebrospinal fluid pressure (CSFP) and cognition has received little research attention. The purpose of this study was to explore the relationship between CSFP and cognition in patients with Alzheimer's disease (AD) and patients with Lewy body dementia (LBD). METHOD We included 178 participants, including 137 patients with AD and 41 patients with LBD (including dementia with Lewy bodies (DLBs) and Parkinson's disease dementia (PDD)). CSFP was measured by lumbar puncture, and a patient-reported history and laboratory test data were collected. Logistic and linear regression analyses were used to evaluate the associations between CSFP and cognition, the cerebrospinal fluid (CSF) / serum albumin ratio (Qalb), and CSF biomarkers of AD. RESULTS The mean age of the included patients was 63.58 ± 8.77 years old, and the mean CSFP was 121 ± 33.72 mmH2O. A total of 76.9% of the patients had a CSFP distribution of [90-170) mmH2O, 46 patients (25.8%) had severe dementia, 83 patients (46.6%) had moderate dementia, 28 patients (15.7%) had mild dementia, and 21 patients (11.8%) had mild cognitive impairment (MCI) (including 16 patients with MCI due to AD and 5 patients with MCI due to LBD). In all patients (p value < 0.001) and in patients with AD (p value = 0.01), the mean cerebrospinal fluid pressure (CSFP) was higher in patients with MCI than in patients with dementia. In multivariate analysis, in all patients (OR: 6.37, 95% confidential interval (CI): 1.76-23.04, p = 0.005) and patients with AD (odds ratio (OR): 5.43, 95% CI: 1.41-20.87, p = 0.005), a CSFP in the lowest quartile ([50-90) mmH2O) was associated with a higher level of severe dementia than a CSFP in the highest quartile ([170-210) mmH2O). In addition, there was a significant linear correlation between CSFP and the Mini-Mental State Examination (MMSE) score in all patients with dementia (r = 0.43, p = 0.04, Durbin-Watson test (D-W test) = 0.75). CONCLUSION In patients with AD, the mean cerebrospinal fluid pressure was higher in patients with MCI than in patients with dementia, and the decrease in CSFP was related to a more serious dementia level. However, no such relationship was found in patients with LBD.
Collapse
Affiliation(s)
- Xia Yang
- Department of Neurology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China
| | - Jinghuan Gan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yong Ji
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China.
| |
Collapse
|
23
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [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: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
24
|
Paul D, Agrawal R, Singh S. Alzheimer's disease and clinical trials. J Basic Clin Physiol Pharmacol 2024; 35:31-44. [PMID: 38491747 DOI: 10.1515/jbcpp-2023-0264] [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] [Accepted: 02/28/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD) is spreading its root disproportionately among the worldwide population. Many genes have been identified as the hallmarks of AD. Based upon the knowledge, many clinical trials have been designed and conducted. Attempts have been made to alleviate the pathology associated with AD by targeting the molecular products of these genes. Irrespective of the understanding on the genetic component of AD, many clinical trials have failed and imposed greater challenges on the path of drug discovery. Therefore, this review aims to identify research and review articles to pinpoint the limitations of drug candidates (thiethylperazine, CT1812, crenezumab, CNP520, and lecanemab), which are under or withdrawn from clinical trials. Thorough analysis of the cross-talk pathways led to the identification of many confounding factors, which could interfere with the success of clinical trials with drug candidates such as thiethylperazine, CT1812, crenezumab, and CNP520. Though these drug candidates were enrolled in clinical trials, yet literature review shows many limitations. These limitations raise many questions on the rationale behind the enrollments of these drug candidates in clinical trials. A meticulous prior assessment of the outcome of clinical studies may stop risky clinical trials at their inceptions. This may save time, money, and resources.
Collapse
Affiliation(s)
- Deepraj Paul
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| | - Rohini Agrawal
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| | - Swati Singh
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| |
Collapse
|
25
|
Maugeri A, Russo C, Patanè GT, Barreca D, Mandalari G, Navarra M. The Inhibition of Mitogen-Activated Protein Kinases (MAPKs) and NF-κB Underlies the Neuroprotective Capacity of a Cinnamon/Curcumin/Turmeric Spice Blend in Aβ-Exposed THP-1 Cells. Molecules 2023; 28:7949. [PMID: 38138438 PMCID: PMC10745857 DOI: 10.3390/molecules28247949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by an increased level of β-amyloid (Aβ) protein deposition in the brain, yet the exact etiology remains elusive. Nowadays, treatments only target symptoms, thus the search for novel strategies is constantly stimulated, and looking to natural substances from the plant kingdom. The aim of this study was to investigate the neuroprotective effects of a spice blend composed of cinnamon bark and two different turmeric root extracts (CCSB) in Aβ-exposed THP-1 cells as a model of neuroinflammation. In abiotic assays, CCSB demonstrated an antioxidant capacity up to three times stronger than Trolox in the ORAC assay, and it reduced reactive oxygen species (ROS) induced by the amyloid fragment in THP-1 cells by up to 39.7%. Moreover, CCSB lowered the Aβ stimulated secretion of the pro-inflammatory cytokines IL-1β and IL-6 by up to 24.9% and 43.4%, respectively, along with their gene expression by up to 25.2% and 43.1%, respectively. The mechanism involved the mitogen-activated protein kinases ERK, JNK and p38, whose phosphorylation was reduced by up to 51.5%, 73.7%, and 58.2%, respectively. In addition, phosphorylation of p65, one of the five components forming NF-κB, was reduced by up to 86.1%. Our results suggest that CCSB can counteract the neuroinflammatory stimulus induced by Aβ-exposure in THP-1 cells, and therefore can be considered a potential candidate for AD management.
Collapse
Affiliation(s)
- Alessandro Maugeri
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Caterina Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (C.R.); (G.T.P.); (D.B.)
| | - Giuseppe Tancredi Patanè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (C.R.); (G.T.P.); (D.B.)
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (C.R.); (G.T.P.); (D.B.)
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (C.R.); (G.T.P.); (D.B.)
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (C.R.); (G.T.P.); (D.B.)
| |
Collapse
|
26
|
Yamada S, Ito H, Tanikawa M, Ii S, Otani T, Wada S, Oshima M, Watanabe Y, Mase M. Age-Related Changes in Cerebrospinal Fluid Dynamics in the Pathogenesis of Chronic Hydrocephalus in Adults. World Neurosurg 2023; 178:351-358. [PMID: 37516143 DOI: 10.1016/j.wneu.2023.07.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023]
Abstract
Cerebrospinal fluid (CSF) dynamics has dramatically changed in this century. In the latest concept of CSF dynamics, CSF is thought to be produced mainly from interstitial fluid excreted from the brain parenchyma and is absorbed in the meningeal lymphatics. Moreover, CSF does not always flow from the ventricles to the subarachnoid space unidirectionally through the foramina of Magendie and Luschka. In an environment of increased intracranial CSF in idiopathic normal pressure hydrocephalus, CSF freely moves through the inferior choroidal point of the choroidal fissure, which interfaces between the inferior horn of the lateral ventricles and the ambient cistern and through the velum interpositum between the third ventricle and the quadrigeminal cistern. The structure of the hippocampus adjacent to the inferior part of the choroidal fissure may be important in preventing the accumulation of waste products in the hippocampus. A recent imaging technology for CSF dynamics, such as four-dimensional flow and intravoxel incoherent motion magnetic resonance imaging, can visualize and quantify the pulsatile complex CSF motion in clinical usage. We present the current concepts of CSF dynamics with advanced magnetic resonance imaging techniques, which will be helpful in the management and understanding of the pathogenesis of chronic hydrocephalus in adults.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan; Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan; Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan.
| | - Hirotaka Ito
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan
| | - Motoki Tanikawa
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Satoshi Ii
- Faculty of System Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Tomohiro Otani
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Shigeo Wada
- Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Marie Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Shiga, Japan
| | - Mitsuhito Mase
- Department of Neurosurgery, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| |
Collapse
|
27
|
Hubert M, Homeyer P, Brandt MD, Donix M, Haußmann R. [Coincidence of normal pressure hydrocephalus and Alzheimer`s disease: therapeutic implications and open questions]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:414-418. [PMID: 37493623 DOI: 10.1055/a-2107-9983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Normal pressure hydrocephalus (NPH) is prevalent in aging patient populations. Despite its clinical relevance, many patients with NPH may not receive adequate treatment. Because of the frequency of Alzheimer`s disease in these patients, there could be overlapping pathophysiological mechanisms that are as yet incompletely understood. Cerebral comorbidities seem to have negative effects on therapeutic response to ventriculoperitoneal shunting. In order to avoid unnecessary and unsuccessful surgery in highly vulnerable elderly patients, they have to be taken into consideration in the diagnostic process.
Collapse
Affiliation(s)
- Max Hubert
- Psychiatrie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Patricia Homeyer
- Psychiatrie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Moritz D Brandt
- Neurologie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Markus Donix
- Psychiatrie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Robert Haußmann
- Psychiatrie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
28
|
Toader C, Tataru CP, Florian IA, Covache-Busuioc RA, Dumitrascu DI, Glavan LA, Costin HP, Bratu BG, Ciurea AV. From Homeostasis to Pathology: Decoding the Multifaceted Impact of Aquaporins in the Central Nervous System. Int J Mol Sci 2023; 24:14340. [PMID: 37762642 PMCID: PMC10531540 DOI: 10.3390/ijms241814340] [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: 09/02/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Aquaporins (AQPs), integral membrane proteins facilitating selective water and solute transport across cell membranes, have been the focus of extensive research over the past few decades. Particularly noteworthy is their role in maintaining cellular homeostasis and fluid balance in neural compartments, as dysregulated AQP expression is implicated in various degenerative and acute brain pathologies. This article provides an exhaustive review on the evolutionary history, molecular classification, and physiological relevance of aquaporins, emphasizing their significance in the central nervous system (CNS). The paper journeys through the early studies of water transport to the groundbreaking discovery of Aquaporin 1, charting the molecular intricacies that make AQPs unique. It delves into AQP distribution in mammalian systems, detailing their selective permeability through permeability assays. The article provides an in-depth exploration of AQP4 and AQP1 in the brain, examining their contribution to fluid homeostasis. Furthermore, it elucidates the interplay between AQPs and the glymphatic system, a critical framework for waste clearance and fluid balance in the brain. The dysregulation of AQP-mediated processes in this system hints at a strong association with neurodegenerative disorders such as Parkinson's Disease, idiopathic normal pressure hydrocephalus, and Alzheimer's Disease. This relationship is further explored in the context of acute cerebral events such as stroke and autoimmune conditions such as neuromyelitis optica (NMO). Moreover, the article scrutinizes AQPs at the intersection of oncology and neurology, exploring their role in tumorigenesis, cell migration, invasiveness, and angiogenesis. Lastly, the article outlines emerging aquaporin-targeted therapies, offering a glimpse into future directions in combatting CNS malignancies and neurodegenerative diseases.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Calin Petru Tataru
- Department of Opthamology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Central Military Emergency Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan-Alexandru Florian
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
29
|
Ullah R, Lee EJ. Advances in Amyloid-β Clearance in the Brain and Periphery: Implications for Neurodegenerative Diseases. Exp Neurobiol 2023; 32:216-246. [PMID: 37749925 PMCID: PMC10569141 DOI: 10.5607/en23014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
This review examines the role of impaired amyloid-β clearance in the accumulation of amyloid-β in the brain and the periphery, which is closely associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). The molecular mechanism underlying amyloid-β accumulation is largely unknown, but recent evidence suggests that impaired amyloid-β clearance plays a critical role in its accumulation. The review provides an overview of recent research and proposes strategies for efficient amyloid-β clearance in both the brain and periphery. The clearance of amyloid-β can occur through enzymatic or non-enzymatic pathways in the brain, including neuronal and glial cells, blood-brain barrier, interstitial fluid bulk flow, perivascular drainage, and cerebrospinal fluid absorption-mediated pathways. In the periphery, various mechanisms, including peripheral organs, immunomodulation/immune cells, enzymes, amyloid-β-binding proteins, and amyloid-β-binding cells, are involved in amyloid-β clearance. Although recent findings have shed light on amyloid-β clearance in both regions, opportunities remain in areas where limited data is available. Therefore, future strategies that enhance amyloid-β clearance in the brain and/or periphery, either through central or peripheral clearance approaches or in combination, are highly encouraged. These strategies will provide new insight into the disease pathogenesis at the molecular level and explore new targets for inhibiting amyloid-β deposition, which is central to the pathogenesis of sporadic AD (amyloid-β in parenchyma) and CAA (amyloid-β in blood vessels).
Collapse
Affiliation(s)
- Rahat Ullah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Neurology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
30
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 328] [Impact Index Per Article: 164.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
31
|
Kopeć K, Szleszkowski S, Koziorowski D, Szlufik S. Glymphatic System and Mitochondrial Dysfunction as Two Crucial Players in Pathophysiology of Neurodegenerative Disorders. Int J Mol Sci 2023; 24:10366. [PMID: 37373513 PMCID: PMC10299586 DOI: 10.3390/ijms241210366] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Neurodegenerative diseases are a complex problem affecting millions of people around the world. The pathogenesis is not fully understood, but it is known that both insufficiency of the glymphatic system and mitochondrial disorders affect the development of pathology. It appears that these are not just two independent factors that coexist in the processes of neurodegeneration, but that they often interact and drive each other. Bioenergetics disturbances are potentially associated with the accumulation of protein aggregates and impaired glymphatic clearance. Furthermore, sleep disorders characteristic of neurodegeneration may impair the work of both the glymphatic system and the activity of mitochondria. Melatonin may be one of the elements linking sleep disorders with the function of these systems. Moreover, noteworthy in this context is the process of neuroinflammation inextricably linked to mitochondria and its impact not only on neurons, but also on glia cells involved in glymphatic clearance. This review only presents possible direct and indirect connections between the glymphatic system and mitochondria in the process of neurodegeneration. Clarifying the connection between these two areas in relation to neurodegeneration could lead to the development of new multidirectional therapies, which, due to the complexity of pathogenesis, seems to be worth considering.
Collapse
Affiliation(s)
| | | | | | - Stanislaw Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (S.S.); (D.K.)
| |
Collapse
|
32
|
Eide PK, Lashkarivand A, Pripp A, Valnes LM, Hovd MH, Ringstad G, Blennow K, Zetterberg H. Plasma neurodegeneration biomarker concentrations associate with glymphatic and meningeal lymphatic measures in neurological disorders. Nat Commun 2023; 14:2084. [PMID: 37045847 PMCID: PMC10097687 DOI: 10.1038/s41467-023-37685-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Clearance of neurotoxic brain proteins via cerebrospinal fluid (CSF) to blood has recently emerged to be crucial, and plasma biomarkers of neurodegeneration were newly introduced to predict neurological disease. This study examines in 106 individuals with neurological disorders associations between plasma biomarkers [40 and 42 amino acid-long amyloid-β (Aβ40 and Aβ42), total-tau, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL)] and magnetic resonance imaging measures of CSF-mediated clearance from brain via extra-vascular pathways (proxy of glymphatic function) and CSF-to-blood clearance variables from pharmacokinetic modeling (proxy of meningeal lymphatic egress). We also examine how biomarkers vary during daytime and associate with subjective sleep quality. Plasma concentrations of neurodegeneration markers associate with indices of glymphatic and meningeal lymphatic functions in individual- and disease-specific manners, vary during daytime, but are unaffected by sleep quality. The results suggest that plasma concentrations of neurodegeneration biomarkers associate with measures of glymphatic and meningeal lymphatic function.
Collapse
Affiliation(s)
- Per Kristian Eide
- Dept. of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Aslan Lashkarivand
- Dept. of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Are Pripp
- Oslo Centre of Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Lars Magnus Valnes
- Dept. of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Markus Herberg Hovd
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Geir Ringstad
- Dept. of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Department of Geriatrics and Internal medicine, Sorlandet Hospital, Arendal, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
33
|
Hoshi K, Kanno M, Goto A, Ugawa Y, Furukawa K, Arai H, Miyajima M, Takahashi K, Hattori K, Kan K, Saito T, Yamaguchi Y, Mitsufuji T, Araki N, Hashimoto Y. Brain-Derived Major Glycoproteins Are Possible Biomarkers for Altered Metabolism of Cerebrospinal Fluid in Neurological Diseases. Int J Mol Sci 2023; 24:ijms24076084. [PMID: 37047057 PMCID: PMC10094273 DOI: 10.3390/ijms24076084] [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: 12/28/2022] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 04/14/2023] Open
Abstract
Cerebrospinal fluid (CSF) plays an important role in the homeostasis of the brain. We previously reported that CSF major glycoproteins are biosynthesized in the brain, i.e., lipocalin-type prostaglandin D2 synthase (L-PGDS) and transferrin isoforms carrying unique glycans. Although these glycoproteins are secreted from distinct cell types, their CSF levels have been found to be highly correlated with each other in cases of neurodegenerative disorders. The aim of this study was to examine these marker levels and their correlations in other neurological diseases, such as depression and schizophrenia, and disorders featuring abnormal CSF metabolism, including spontaneous intracranial hypotension (SIH) and idiopathic normal pressure hydrocephalus (iNPH). Brain-derived marker levels were found to be highly correlated with each other in the CSF of depression and schizophrenia patients. SIH is caused by CSF leakage, which is suspected to induce hypovolemia and a compensatory increase in CSF production. In SIH, the brain-derived markers were 2-3-fold higher than in other diseases, and, regardless of their diverse levels, they were found to be correlated with each other. Another abnormality of the CSF metabolism, iNPH, is possibly caused by the reduced absorption of CSF, which secondarily induces CSF accumulation in the ventricle; the excess CSF compresses the brain's parenchyma to induce dementia. One potential treatment is a "shunt operation" to bypass excess CSF from the ventricles to the peritoneal cavity, leading to the attenuation of dementia. After the shunt operation, marker levels began to increase within a week and then further increased by 2-2.5-fold at three, six, and twelve months post-operation, at which point symptoms had gradually attenuated. Notably, the marker levels were found to be correlated with each other in the post-operative period. In conclusion, the brain-derived major glycoprotein markers were highly correlated in the CSF of patients with different neurological diseases, and their correlations were maintained even after surgical intervention. These results suggest that brain-derived proteins could be biomarkers of CSF production.
Collapse
Grants
- 16hm0102042h0001, 17hm0102042h0002, 18hm0102042h0003, 19dk0310099h0001, 20dk0310099h0002, 21dk0310099h0003, 20dm0307003h0003, 21dm0307003h0004 the Japan Agency for Medical Research and Development (AMED)
- 16hm0102042h0001, 17hm0102042h0002, 18hm0102042h0003, 19dk0310099h0001, 20dk0310099h0002, 21dk0310099h0003, 20dm0307003h0003, 21dm0307003h0004 the Japan Agency for Medical Research and Development (AMED)
Collapse
Affiliation(s)
- Kyoka Hoshi
- Department of Biochemistry, Fukushima Medical University, Fukushima City 960-1295, Fukushima, Japan
| | - Mayumi Kanno
- Department of Forensic Medicine, Fukushima Medical University, Fukushima City 960-1295, Fukushima, Japan
| | - Aya Goto
- Center for Integrated Science and Humanities, Fukushima Medical University, Fukushima City 960-1295, Fukushima, Japan
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, Fukushima Medical University, Fukushima City 960-1295, Fukushima, Japan
| | - Katsutoshi Furukawa
- Division of Community Medicine, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Miyagi, Japan
| | - Hiroyuki Arai
- Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Miyagi, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University, Bunkyo City 113-8421, Tokyo, Japan
| | - Koichi Takahashi
- Department of Neurosurgery, Sanno Hospital, Minato City 107-0052, Tokyo, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira 187-8502, Tokyo, Japan
| | - Keiichi Kan
- Department of Anesthesiology, Southern Tohoku General Hospital, Koriyama 963-8052, Fukushima, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Nagoya City University, Nagoya 467-8601, Aichi, Japan
| | - Yoshiki Yamaguchi
- Laboratory of Pharmaceutical Physical Chemistry, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Miyagi, Japan
| | - Takashi Mitsufuji
- Department of Neurology, Saitama Medical University Hospital, Koshigaya 350-0495, Saitama, Japan
| | - Nobuo Araki
- Department of Neurology, Saitama Medical University Hospital, Koshigaya 350-0495, Saitama, Japan
| | - Yasuhiro Hashimoto
- Department of Forensic Medicine, Fukushima Medical University, Fukushima City 960-1295, Fukushima, Japan
| |
Collapse
|
34
|
Zaksaite T, Loveday C, Edginton T, Spiers HJ, Smith AD. Hydrocephalus: A neuropsychological and theoretical primer. Cortex 2023; 160:67-99. [PMID: 36773394 DOI: 10.1016/j.cortex.2023.01.001] [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: 04/22/2022] [Revised: 09/09/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Hydrocephalus is a common neurological condition, the hallmark feature of which is an excess in production, or accumulation, of cerebrospinal fluid in the ventricles. Although it is associated with diffuse damage to paraventricular brain areas, patients are broadly typified by a particular pattern of cognitive impairments that include deficits in working memory, attention, and spatial abilities. There have, however, been relatively few neuropsychological accounts of the condition. Moreover, theories of the relationship between aetiology and impairment appear to have emerged in isolation of each other, and proffer fundamentally different accounts. In this primer, we aim to provide a comprehensive and contemporary overview of hydrocephalus for the neuropsychologist, covering cognitive sequelae and theoretical interpretations of their origins. We review clinical and neuropsychological assays of cognitive profiles, along with the few studies that have addressed more integrative behaviours. In particular, we explore the distinction between congenital or early-onset hydrocephalus with a normal-pressure variant that can be acquired later in life. The relationship between these two populations is a singularly interesting one in neuropsychology since it can allow for the examination of typical and atypical developmental trajectories, and their interaction with chronic and acute impairment, within the same broad neurological condition. We reflect on the ramifications of this for our subject and suggest avenues for future research.
Collapse
Affiliation(s)
- Tara Zaksaite
- School of Psychology, University of Plymouth, Plymouth, PL4 8AA, UK.
| | - Catherine Loveday
- School of Social Sciences, University of Westminster, 115 New Cavendish St, London W1W 6UW, UK
| | - Trudi Edginton
- Department of Psychology, City, University of London, Northampton Square, London, EC1V 0HB, UK
| | - Hugo J Spiers
- Department of Experimental Psychology, Division of Psychology and Language Sciences, University College London, 26 Bedford Way, London, WC1H 0AP, UK
| | - Alastair D Smith
- School of Psychology, University of Plymouth, Plymouth, PL4 8AA, UK; Brain Research and Imaging Centre, University of Plymouth, 7 Derriford Rd, Plymouth, PL6 8BU, UK.
| |
Collapse
|
35
|
van der Thiel MM, Backes WH, Ramakers IHGB, Jansen JFA. Novel developments in non-contrast enhanced MRI of the perivascular clearance system: What are the possibilities for Alzheimer's disease research? Neurosci Biobehav Rev 2023; 144:104999. [PMID: 36529311 DOI: 10.1016/j.neubiorev.2022.104999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
The cerebral waste clearance system (i.e, glymphatic or intramural periarterial drainage) works through a network of perivascular spaces (PVS). Dysfunction of this system likely contributes to aggregation of Amyloid-β and subsequent toxic plaques in Alzheimer's disease (AD). A promising, non-invasive technique to study this system is MRI, though applications in dementia are still scarce. This review focusses on recent non-contrast enhanced (non-CE) MRI techniques which determine and visualise physiological aspects of the clearance system at multiple levels, i.e., cerebrospinal fluid flow, PVS-flow and interstitial fluid movement. Furthermore, various MRI studies focussing on aspects of the clearance system which are relevant to AD are discussed, such as studies on ageing, sleep alterations, and cognitive decline. Additionally, the complementary function of non-CE to CE methods is elaborated upon. We conclude that non-CE studies have great potential to determine which parts of the waste clearance system are affected by AD and in which stages of cognitive impairment dysfunction of this system occurs, which could allow future clinical trials to target these specific mechanisms.
Collapse
Affiliation(s)
- Merel M van der Thiel
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Inez H G B Ramakers
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
36
|
Cerebrospinal Fluid Biomarkers in iNPH: A Narrative Review. Diagnostics (Basel) 2022; 12:diagnostics12122976. [PMID: 36552981 PMCID: PMC9777226 DOI: 10.3390/diagnostics12122976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is a neurological syndrome characterized by the clinical triad of gait disorder, cognitive impairment and urinary incontinence. It has attracted interest because of the possible reversibility of symptoms, especially with timely treatment. The main pathophysiological theory is based on a vicious circle of disruption in circulation of cerebrospinal fluid (CSF) that leads to the deceleration of its absorption. Data regarding CSF biomarkers in iNPH are contradictory and no definite CSF biomarker profile has been recognized as in Alzheimer's disease (AD), which often co-exists with iNPH. In this narrative review, we investigated the literature regarding CSF biomarkers in iNPH, both the established biomarkers total tau protein (t-tau), phosphorylated tau protein (p-tau) and amyloid peptide with 42 amino acids (Aβ42), and other molecules, which are being investigated as emerging biomarkers. The majority of studies demonstrate differences in CSF concentrations of Aβ42 and tau-proteins (t-tau and p-tau) among iNPH patients, healthy individuals and patients with AD and vascular dementia. iNPH patients present with lower CSF Aβ42 and p-tau concentrations than healthy individuals and lower t-tau and p-tau concentrations than AD patients. This could prove helpful for improving diagnosis, differential diagnosis and possibly prognosis of iNPH patients.
Collapse
|
37
|
Liu R, Zhang Z, Chen Y, Liao J, Wang Y, Liu J, Lin Z, Xiao G. Choroid plexus epithelium and its role in neurological diseases. Front Mol Neurosci 2022; 15:949231. [PMID: 36340696 PMCID: PMC9633854 DOI: 10.3389/fnmol.2022.949231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/15/2022] [Indexed: 02/16/2024] Open
Abstract
Choroid plexus epithelial cells can secrete cerebrospinal fluid into the ventricles, serving as the major structural basis of the selective barrier between the neurological system and blood in the brain. In fact, choroid plexus epithelial cells release the majority of cerebrospinal fluid, which is connected with particular ion channels in choroid plexus epithelial cells. Choroid plexus epithelial cells also produce and secrete a number of essential growth factors and peptides that help the injured cerebrovascular system heal. The pathophysiology of major neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, as well as minor brain damage diseases like hydrocephalus and stroke is still unknown. Few studies have previously connected choroid plexus epithelial cells to the etiology of these serious brain disorders. Therefore, in the hopes of discovering novel treatment options for linked conditions, this review extensively analyzes the association between choroid plexus epithelial cells and the etiology of neurological diseases such as Alzheimer's disease and hydrocephalus. Finally, we review CPE based immunotherapy, choroid plexus cauterization, choroid plexus transplantation, and gene therapy.
Collapse
Affiliation(s)
- Ruizhen Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiping Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yibing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuchang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jingping Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Lin
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Han J, Kim MN, Lee HW, Jeong SY, Lee SW, Yoon U, Kang K. Distinct volumetric features of cerebrospinal fluid distribution in idiopathic normal-pressure hydrocephalus and Alzheimer's disease. Fluids Barriers CNS 2022; 19:66. [PMID: 36045420 PMCID: PMC9434899 DOI: 10.1186/s12987-022-00362-8] [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: 03/04/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Objective The aims of the study were to measure the cerebrospinal fluid (CSF) volumes in the lateral ventricle, high-convexity subarachnoid space, and Sylvian fissure region in patients with idiopathic normal-pressure hydrocephalus (INPH) and Alzheimer’s disease (AD), and to evaluate differences in these volumes between INPH and AD groups and healthy controls. Methods Forty-nine INPH patients, 59 AD patients, and 26 healthy controls were imaged with automated three-dimensional volumetric MRI. Results INPH patients had larger lateral ventricles and CSF spaces of the Sylvian fissure region and smaller high-convexity subarachnoid spaces than other groups, and AD patients had larger lateral ventricles and CSF spaces of the Sylvian fissure region than the control group. The INPH group showed a negative correlation between lateral ventricle and high-convexity subarachnoid space volumes, while the AD group showed a positive correlation between lateral ventricle volume and volume for CSF spaces of the Sylvian fissure region. The ratio of lateral ventricle to high-convexity subarachnoid space volumes yielded an area under the curve of 0.990, differentiating INPH from AD. Conclusions Associations between CSF volumes suggest that there might be different mechanisms between INPH and AD to explain their respective lateral ventricular dilations. The ratio of lateral ventricle to high-convexity subarachnoid space volumes distinguishes INPH from AD with good diagnostic sensitivity and specificity. We propose to refer to this ratio as the VOSS (ventricle over subarachnoid space) index.
Collapse
Affiliation(s)
- Jaehwan Han
- Department of Biomedical Engineering, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Myoung Nam Kim
- Department of Biomedical Engineering, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ho-Won Lee
- Department of Neurology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Uicheul Yoon
- Department of Biomedical Engineering, Daegu Catholic University, 13-13 Hayang- ro, Hayang-eup, Gyeongsan, Gyeongbuk, 38430, South Korea.
| | - Kyunghun Kang
- Department of Neurology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, South Korea.
| |
Collapse
|
39
|
Yoshida H, Ishida S, Yamamoto T, Ishikawa T, Nagata Y, Takeuchi K, Ueno H, Imai Y. Effect of cilia-induced surface velocity on cerebrospinal fluid exchange in the lateral ventricles. JOURNAL OF THE ROYAL SOCIETY, INTERFACE 2022; 19:20220321. [PMID: 35919976 PMCID: PMC9346361 DOI: 10.1098/rsif.2022.0321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ciliary motility disorders are known to cause hydrocephalus. The instantaneous velocity of cerebrospinal fluid (CSF) flow is dominated by artery pulsation, and it remains unclear why ciliary dysfunction results in hydrocephalus. In this study, we investigated the effects of cilia-induced surface velocity on CSF flow using computational fluid dynamics. A geometric model of the human ventricles was constructed using medical imaging data. The CSF produced by the choroid plexus and cilia-induced surface velocity were given as the velocity boundary conditions at the ventricular walls. We developed healthy and reduced cilia motility models based on experimental data of cilia-induced velocity in healthy wild-type and Dpcd-knockout mice. The results indicate that there is almost no difference in intraventricular pressure between healthy and reduced cilia motility models. Additionally, it was found that newly produced CSF from the choroid plexus did not spread to the anterior and inferior horns of the lateral ventricles in the reduced cilia motility model. These findings suggest that a ciliary motility disorder could delay CSF exchange in the anterior and inferior horns of the lateral ventricles.
Collapse
Affiliation(s)
- Haruki Yoshida
- Graduate School of Engineering, Kobe University, Kobe 657-8501, Japan
| | - Shunichi Ishida
- Graduate School of Engineering, Kobe University, Kobe 657-8501, Japan
| | - Taiki Yamamoto
- Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Takayuki Ishikawa
- Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuichi Nagata
- Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Kazuhito Takeuchi
- Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Hironori Ueno
- Aichi University of Education, Kariya 448-8542, Japan
| | - Yohsuke Imai
- Graduate School of Engineering, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
40
|
Goh ET, Lock C, Tan AJL, Tan BL, Liang S, Pillay R, Kumar S, Ahmad-Annuar A, Narayanan V, Kwok J, Tan YJ, Ng ASL, Tan EK, Czosnyka Z, Czosnyka M, Pickard JD, Keong NC. Clinical Outcomes After Ventriculo-Peritoneal Shunting in Patients With Classic vs. Complex NPH. Front Neurol 2022; 13:868000. [PMID: 35903111 PMCID: PMC9315242 DOI: 10.3389/fneur.2022.868000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/13/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Normal pressure hydrocephalus (NPH) is a neurological condition characterized by a clinical triad of gait disturbance, cognitive impairment, and urinary incontinence in conjunction with ventriculomegaly. Other neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and vascular dementia share some overlapping clinical features. However, there is evidence that patients with comorbid NPH and Alzheimer's or Parkinson's disease may still exhibit good clinical response after CSF diversion. This study aims to evaluate clinical responses after ventriculo-peritoneal shunt (VPS) in a cohort of patients with coexisting NPH and neurodegenerative disease. Methods The study has two components; (i) a pilot study was performed that specifically focused upon patients with Complex NPH and following the inclusion of the Complex NPH subtype into consideration for the clinical NPH programme, (ii) a retrospective snapshot study was performed to confirm and characterize differences between Classic and Complex NPH patients being seen consecutively over the course of 1 year within a working subspecialist NPH clinic. We studied the characteristics of patients with Complex NPH, utilizing clinical risk stratification and multimodal biomarkers. Results There was no significant difference between responders and non-responders to CSF diversion on comorbidity scales. After VPS insertion, significantly more Classic NPH patients had improved cognition compared to Complex NPH patients (p = 0.005). Improvement in gait and urinary symptoms did not differ between the groups. 26% of the Classic NPH group showed global improvement of the triad, and 42% improved in two domains. Although only 8% showed global improvement of the triad, all Complex NPH patients improved in gait. Conclusions Our study has demonstrated that the presence of neurodegenerative disorders co-existing with NPH should not be the sole barrier to the consideration of high-volume tap test or lumbar drainage via a specialist NPH programme. Further characterization of distinct cohorts of NPH with differing degrees of CSF responsiveness due to overlay from neurodegenerative or comorbidity risk burden may aid toward more precise prognostication and treatment strategies. We propose a simplistic conceptual framework to describe NPH by its Classic vs. Complex subtypes to promote the clinical paradigm shift toward subspecialist geriatric neurosurgery by addressing needs for rapid screening tools at the clinical-research interface.
Collapse
Affiliation(s)
- Eng Tah Goh
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Christine Lock
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Audrey Jia Luan Tan
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Bee Ling Tan
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Sai Liang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Robin Pillay
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Sumeet Kumar
- Department of Neuroradiology, National Neuroscience Institute, Singapore, Singapore
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Vairavan Narayanan
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Janell Kwok
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Adeline SL Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Zofia Czosnyka
- Neurosurgical Division, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Marek Czosnyka
- Neurosurgical Division, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - John D. Pickard
- Neurosurgical Division, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Nicole C. Keong
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- *Correspondence: Nicole C. Keong
| |
Collapse
|
41
|
Liu S, Fan M, Zheng Q, Hao S, Yang L, Xia Q, Qi C, Ge J. MicroRNAs in Alzheimer's disease: Potential diagnostic markers and therapeutic targets. Biomed Pharmacother 2022; 148:112681. [PMID: 35177290 DOI: 10.1016/j.biopha.2022.112681] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with cognitive decline as the primary clinical feature. According to epidemiological statistics, 50 million people worldwide are currently affected by Alzheimer's disease. Although new drugs such as aducanumab have been approved for use in the treatment of AD, none of them have reversed the progression of AD. MicroRNAs (miRNAs) are small molecule RNAs that exert their biological functions by regulating the expression of intracellular proteins, and differential abundance and varieties are found between the central and peripheral tissues of AD patients and healthy controls. This article will summarise the changes of miRNAs in the AD process, and the potential role of diagnostic markers and therapeutic targets in AD will be explored.
Collapse
Affiliation(s)
- Sen Liu
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Min Fan
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Qiang Zheng
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Longjun Yang
- Chaohu Clinical Medical College, Anhui Medical University, Hefei, China
| | - Qingrong Xia
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China
| | - Congcong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, China.
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.
| |
Collapse
|
42
|
Darbandi AD, Saadat GH, Alsoof D, Rebic A, Siddiqi A, Butler BA, Bokhari F. Effects of Delayed Hip Fracture Surgery on Severely Ill Patients: Defining the Time to Medical Optimization. Am Surg 2022:31348221080425. [PMID: 35324321 DOI: 10.1177/00031348221080425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Patients with multiple comorbidities often have delayed hip fracture surgery due to medical optimization. The goal of this study is to identify the allowable time for medical optimization in severely ill hip fracture patients. METHODS The 2016-2019 NSQIP database was used to identify patients over age 60 with ASA classification scores 3 and 4 for severe and life-threatening systemic diseases. Patients were divided into immediate (<24 hours), early (24-48 hours), or late (>48 hours) groups based on time to surgery (TTS). Risk-adjusted multivariable logistic regressions were conducted to compare relationships between 30-day postoperative outcomes and TTS. RESULTS 43,071 hip fracture cases were analyzed for the purposes of this study. Compared to patients who underwent surgery immediately, patients who had surgeries between 24 and 48 hours were associated with higher rates of pneumonia (OR 1.357, CI 1.194-1.542), UTIs (OR 1.155, CI 1.000-1.224), readmission (OR 1.136, CI 1.041-1.240), postoperative LOS beyond 6 days (OR 1.249, CI 1.165-1.340), and mortality (OR 1.205, CI 1.084-1.338). Patients with surgeries delayed beyond 48 hours were associated with higher rates of CVA (OR 1.542, CI 1.048-2.269), pneumonia (OR 1.886, CI 1.611-2.209), UTIs (OR 1.546, CI 1.283-1.861), readmission (OR 1.212, CI 1.074-1.366), postoperative LOS beyond 6 days (OR 1.829, CI 1.670-2.003), and mortality (OR 1.475, CI 1.286-1.693) compared to patients with immediate surgery. DISCUSSION Severely ill patients with the hip fracture may have a 24-hour window for medical optimization. Hip fracture surgery performed beyond 48 hours is associated with higher complication rates and mortality among those who are severely ill. Further prospective studies are warranted to examine the effects of early surgical intervention among severely ill patients.
Collapse
Affiliation(s)
| | - Ghulam H Saadat
- Department of Trauma and Burn Surgery, John H Stroger Hospital of Cook County, Chicago, IL, USA
| | | | - Ante Rebic
- 32959Kansas City University, Kansas City, MO, USA
| | - Ahmed Siddiqi
- Orthopedic Institute of Central Jersey, a division of Ortho Alliance NJ, Manasquan, NJ, USA; and Department of Orthopedic Surgery, Hackensack Meridian School of Medicine, Jersey Shore University Medical Center
| | - Bennet A Butler
- Department of Trauma and Burn Surgery, John H Stroger Hospital of Cook County, Chicago, IL, USA
| | - Faran Bokhari
- Department of Orthopedic Surgery, Northwestern Memorial Hospital, Chicago, IL, USA
| |
Collapse
|
43
|
The Influence of the Ventricular-Lumbar Gradient on Cerebrospinal Fluid Analysis in Serial Samples. Brain Sci 2022; 12:brainsci12030410. [PMID: 35326365 PMCID: PMC8946585 DOI: 10.3390/brainsci12030410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Cerebrospinal fluid (CSF) samples from patients with non-inflammatory neurological diseases are used for control groups in biomarker studies. Since large amounts of CSF are withdrawn, patients with idiopathic intracranial hypertension (IIH) or normal pressure hydrocephalus (NPH) are especially suitable. The serially taken CSF portions are usually collected in different tubes. We aimed to investigate whether the later random choice of one of these tubes for CSF investigations might harbor the risk of different CSF protein findings due to the so-called ventriculo-lumbar CSF gradient. Methods: Patients with IIH (9) and NPH (7) were included. CSF was serially taken and collected in six tubes of 5 mL each. Concentrations and CSF-serum quotients of immunoglobulins, albumin and the virus-specific antibody index (AI) were determined in the first, fourth and sixth CSF fraction. Results: CSF immunoglobulin and albumin concentrations and CSF-serum protein quotients were significantly lower in the fourth and sixth CSF fraction compared with the first CSF fraction. Virus-specific AI did not significantly differ in the different CSF fractions. Conclusions: CSF protein analytics should be performed in the first CSF fraction in order to avoid different measurement results and achieve comparability within a control group and between different control and patient groups.
Collapse
|
44
|
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To determine the effect of cerebrospinal fluid (CSF) shunting versus no CSF shunting in people with idiopathic normal pressure hydrocephalus (iNPH). To determine the frequency of adverse effects of CSF shunting in iNPH
Collapse
|
45
|
Killer HE. Special Cerebral and Cerebrospinal Features in Primary Open Angle Glaucoma and Normal Tension Glaucoma. Klin Monbl Augenheilkd 2022; 239:177-181. [PMID: 35211940 DOI: 10.1055/a-1699-2911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In addition to aqueous humour and blood, cerebrospinal fluid also plays an important part in the pathophysiology of primary open-angle glaucoma (POAG) and, in particular, normal-tension glaucoma (NTG). Apart from the important role of CSF pressure in papillary congestion, the composition of the CSF and its flow rate are relevant. CSF is in contact with the brain, the spinal canal and the optic nerve. In neurodegenerative disease, one potential pathophysiological factor, apart from an altered composition of the CSF, is a decrease in flow rate. Changes in CSF composition and flow rate have also been described in the perioptic subarachnoid space of the optic nerve in patients with normal tension glaucoma. Such findings indicate that primary open angle glaucoma and normal tension glaucoma especially, might be due to a neurodegenerative process.
Collapse
|
46
|
Jang H, Park YH, Choe YS, Kang SH, Kang ES, Lee S, Seo SW, Kim HJ, Na DL. Amyloid Positive Hydrocephalus: A Hydrocephalic Variant of Alzheimer's Disease? J Alzheimers Dis 2021; 85:1467-1479. [PMID: 34958024 DOI: 10.3233/jad-215110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) and normal pressure hydrocephalus (NPH) commonly coexist. OBJECTIVE We aimed to characterize an overlapping syndrome of AD and NPH that presents with gait disturbance, ventriculomegaly on magnetic resonance imaging, and significant amyloid deposition on positron emission tomography (PET). METHODS Of 114 patients who underwent cerebrospinal fluid (CSF) drainage for a possible diagnosis of NPH between 2015 and 2020 in Samsung Medical Center, we identified 24 patients (21.1%) with the NPH patients with amyloid deposition on PET, which we referred to as hydrocephalic AD in this study. We compared their clinical and imaging findings with those of 123 typical AD without hydrocephalic signs/symptoms. We also investigated the frequency and potential predictors of the tap test response in hydrocephalic AD. RESULTS Evans' index was 0.36±0.03, and a disproportionately enlarged subarachnoid space was present in 54.2% of the hydrocephalic AD patients. The mean age (75.2±7.3 years) and the APOE4 frequency (68.2%) did not differ from those of AD controls. However, the hydrocephalic AD patients showed better memory and language performance, and a thinner cingulate cortex. About 42% of the hydrocephalic AD patients responded to the tap test, of whom seven underwent shunt surgery. Cognition did not improve, whereas gait improved after shunt surgery in all. CONCLUSION Hydrocephalic AD has different neuropsychological and imaging characteristics from typical AD. Future studies are warranted to further investigate the effect of CSF removal on their clinical course and to elucidate the pathophysiological interaction between amyloid and NPH.
Collapse
Affiliation(s)
- Hyemin Jang
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yu-Hyun Park
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Sim Choe
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea.,Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun-Sook Kang
- Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seunghoon Lee
- Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Hee Jin Kim
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk L Na
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
47
|
Wang Z, Sharda N, Curran GL, Li L, Lowe VJ, Kandimalla KK. Semimechanistic Population Pharmacokinetic Modeling to Investigate Amyloid Beta Trafficking and Accumulation at the BBB Endothelium. Mol Pharm 2021; 18:4148-4161. [PMID: 34664956 DOI: 10.1021/acs.molpharmaceut.1c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Elevated exposure to toxic amyloid beta (Aβ) peptides and consequent blood-brain barrier (BBB) dysfunction are believed to promote vasculopathy in Alzheimer's disease (AD). However, the accumulation kinetics of different Aβ isoforms within the BBB endothelium and how it drives BBB dysfunction are not clearly characterized. Using single positron emission computed tomography (SPECT)-computed tomography (CT) dynamic imaging coupled with population pharmacokinetic modeling, we investigated the accumulation kinetics of Aβ40 and Aβ42 in the BBB endothelium. Brain clearance was quantified after intracerebral administration of 125I-Aβ, and BBB-mediated transport was shown to account for 54% of 125I-Aβ40 total clearance. A brain influx study demonstrated lower values of both maximal rate (Vmax) and Michaelis constant (Km) for 125I-Aβ42 compared to 125I-Aβ40. Validated by a transcytosis study in polarized human BBB endothelial cell (hCMEC/D3) monolayers, model simulations demonstrated impaired exocytosis was responsible for inefficient permeability and enhanced accumulation of Aβ42 in the BBB endothelium. Further, both isoforms were shown to disrupt the exocytosis machinery of BBB endothelial cells so that a vicious cycle could be generated. The validated model was able to capture changes in Aβ steady-state levels in plasma as well as the brain during AD progression and allowed us to predict the kinetics of Aβ accumulation in the BBB endothelium.
Collapse
Affiliation(s)
- Zengtao Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United Sates
| | - Nidhi Sharda
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United Sates
| | - Geoffry L Curran
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, United Sates
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United Sates
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, United Sates
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United Sates
| |
Collapse
|
48
|
Ullah R, Park TJ, Huang X, Kim MO. Abnormal amyloid beta metabolism in systemic abnormalities and Alzheimer's pathology: Insights and therapeutic approaches from periphery. Ageing Res Rev 2021; 71:101451. [PMID: 34450351 DOI: 10.1016/j.arr.2021.101451] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated, multifactorial neurodegenerative disorder that is incurable. Despite recent success in treatments that partially improve symptomatic relief, they have failed in most clinical trials. Re-holding AD for accurate diagnosis and treatment is widely known as a challenging task. Lack of knowledge of basic molecular pathogenesis might be a possible reason for ineffective AD treatment. Historically, a majority of therapy-based studies have investigated the role of amyloid-β (Aβ peptide) in the central nervous system (CNS), whereas less is known about Aβ peptide in the periphery in AD. In this review, we provide a comprehensive summary of the current understanding of Aβ peptide metabolism (anabolism and catabolism) in the brain and periphery. We show that the abnormal metabolism of Aβ peptide is significantly linked with central-brain and peripheral abnormalities; the interaction between peripheral Aβ peptide metabolism and peripheral abnormalities affects central-brain Aβ peptide metabolism, suggesting the existence of significant communication between these two pathways of Aβ peptide metabolism. This close interaction between the central brain and periphery in abnormal Aβ peptide metabolism plays a key role in the development and progression of AD. In conclusion, we need to obtain a full understanding of the dynamic roles of Aβ peptide at the molecular level in both the brain and periphery in relation to the pathology of AD. This will not only provide new information regarding the complex disease pathology, but also offer potential new clues to improve therapeutic strategies and diagnostic biomarkers for the successful treatment of AD.
Collapse
|
49
|
Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, Villemagne VL, Aisen P, Vendruscolo M, Iwatsubo T, Masters CL, Cho M, Lannfelt L, Cummings JL, Vergallo A. The Amyloid-β Pathway in Alzheimer's Disease. Mol Psychiatry 2021; 26:5481-5503. [PMID: 34456336 PMCID: PMC8758495 DOI: 10.1038/s41380-021-01249-0] [Citation(s) in RCA: 873] [Impact Index Per Article: 218.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer's disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| | - John Hardy
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christopher Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Aisen
- USC Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Colin L Masters
- Laureate Professor of Dementia Research, Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Min Cho
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA
| | - Lars Lannfelt
- Uppsala University, Department of of Public Health/Geriatrics, Uppsala, Sweden
- BioArctic AB, Stockholm, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Andrea Vergallo
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| |
Collapse
|
50
|
Kawamura K, Miyajima M, Nakajima M, Kanai M, Motoi Y, Nojiri S, Akiba C, Ogino I, Xu H, Kamohara C, Yamada S, Karagiozov K, Ikeuchi T, Kondo A, Arai H. Cerebrospinal Fluid Amyloid-β Oligomer Levels in Patients with Idiopathic Normal Pressure Hydrocephalus. J Alzheimers Dis 2021; 83:179-190. [PMID: 34275898 PMCID: PMC8461658 DOI: 10.3233/jad-210226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The amyloid-β oligomers, consisting of 10-20 monomers (AβO10-20), have strong neurotoxicity and are associated with cognitive impairment in Alzheimer's disease (AD). However, their role in patients with idiopathic normal pressure hydrocephalus (iNPH) is poorly understood. OBJECTIVE We hypothesized that cerebrospinal fluid (CSF) AβO10-20 accumulates in patients with iNPH, and its clearance after CSF shunting contributes to neurological improvement. We measured CSF AβO10-20 levels before and after CSF shunting in iNPH patients evaluating their diagnostic and prognostic role. METHODS We evaluated two iNPH cohorts: "evaluation" (cohort-1) with 32 patients and "validation" (cohort-2) with 13 patients. Comparison cohorts included: 27 neurologically healthy controls (HCs), and 16 AD, 15 Parkinson's disease (PD), and 14 progressive supranuclear palsy (PSP) patients. We assessed for all cohorts CSF AβO10-20 levels and their comprehensive clinical data. iNPH cohort-1 pre-shunting data were compared with those of comparison cohorts, using cohort-2 for validation. Next, we compared cohort-1's clinical and CSF data: 1) before and after CSF shunting, and 2) increased versus decreased AβO10-20 levels at baseline, 1 and 3 years after shunting. RESULTS Cohort-1 had higher CSF AβO10-20 levels than the HCs, PD, and PSP cohorts. This result was validated with data from cohort-2. CSF AβO10-20 levels differentiated cohort-1 from the PD and PSP groups, with an area under receiver operating characteristic curve of 0.94. AβO10-20 levels in cohort-1 decreased after CSF shunting. Patients with AβO10-20 decrease showed better cognitive outcome than those without. CONCLUSION AβO10-20 accumulates in patients with iNPH and is eliminated by CSF shunting. AβO10-20 can be an applicable diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Kaito Kawamura
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo Tokyo Koto Geriatric Medical Centre, Shinsuna Koto-ku, Tokyo, Japan
| | - Madoka Nakajima
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Mitsuyasu Kanai
- Department of Neurology, Mihara Memorial Hospital, Ota-cho, Isesaki-shi, Gunma, Japan
| | - Yumiko Motoi
- Department of Neurology, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Chihiro Akiba
- Department of Neurosurgery, Juntendo Tokyo Koto Geriatric Medical Centre, Shinsuna Koto-ku, Tokyo, Japan
| | - Ikuko Ogino
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Hanbing Xu
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Chihiro Kamohara
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Shinya Yamada
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan.,Department of Neurosurgery, Kugayama Hospital, Kita-Karasuyama, Setagaya-ku, Tokyo, Japan
| | - Kostadin Karagiozov
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Asahimachi, Niigata, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University, Hongo Bunkyo-ku, Tokyo, Japan
| |
Collapse
|