151
|
Systemic Inflammation Predicts Alzheimer Pathology in Community Samples without Dementia. Biomedicines 2022; 10:biomedicines10061240. [PMID: 35740262 PMCID: PMC9219863 DOI: 10.3390/biomedicines10061240] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation and oxidative stress (OS) are implicated in the pathophysiology of Alzheimer’s disease (AD). However, it is unclear at what stage of the disease process inflammation first becomes manifest. The aim of this study was to investigate the associations between specific plasma markers of inflammation and OS, tau, and Amyloid-β 38, 40, and 42 levels in cognitively unimpaired middle-age and older individuals. Associations between inflammatory states identified through principal component analysis and AD biomarkers were investigated in middle-age (52–56 years, n = 335, 52% female) and older-age (72–76 years, n = 351, 46% female) participants without dementia. In middle-age, a component reflecting variation in OS was most strongly associated with tau and to a lesser extent amyloid-β levels. In older-age, a similar component to that observed in middle-age was only associated with tau, while another component reflecting heightened inflammation independent of OS, was associated with all AD biomarkers. In middle and older-age, inflammation and OS states are associated with plasma AD biomarkers.
Collapse
|
152
|
Lee MK, Cho SJ, Bae YJ, Kim JM. MRI-Based Demonstration of the Normal Glymphatic System in a Human Population: A Systematic Review. Front Neurol 2022; 13:827398. [PMID: 35693018 PMCID: PMC9174517 DOI: 10.3389/fneur.2022.827398] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background The glymphatic system has been described as one that facilitates the exchange between the cerebrospinal fluid (CSF) and interstitial fluid, and many recent studies have demonstrated glymphatic flow based on magnetic resonance imaging (MRI). We aim to systematically review the studies demonstrating a normal glymphatic flow in a human population using MRI and to propose a detailed glymphatic imaging protocol. Methods We searched the MEDLINE and EMBASE databases to identify studies with human participants involving MRI-based demonstrations of the normal glymphatic flow. We extracted data on the imaging sequence, imaging protocol, and the targeted anatomical structures on each study. Results According to contrast-enhanced MRI studies, peak enhancement was sequentially detected first in the CSF space, followed by the brain parenchyma, the meningeal lymphatic vessel (MLV), and, finally, the cervical lymph nodes, corresponding with glymphatic flow and explaining the drainage into the MLV. Non-contrast flow-sensitive MRI studies revealed similar glymphatic inflow from the CSF space to the brain parenchyma and efflux of exchanged fluid from the brain parenchyma to the MLV. Conclusion We may recommend T1-weighted contrast-enhanced MRI for visualizing glymphatic flow. Our result can increase understanding of the glymphatic system and may lay the groundwork for establishing central nervous system fluid dynamic theories and developing standardized imaging protocols.
Collapse
Affiliation(s)
- Min Kyoung Lee
- Department of Radiology, College of Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Soeul, South Korea
| | - Se Jin Cho
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Yun Jung Bae
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
- *Correspondence: Yun Jung Bae
| | - Jong-Min Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
- Jong-Min Kim
| |
Collapse
|
153
|
Tithof J, Boster KA, Bork PA, Nedergaard M, Thomas JH, Kelley DH. A network model of glymphatic flow under different experimentally-motivated parametric scenarios. iScience 2022; 25:104258. [PMID: 35521514 PMCID: PMC9062681 DOI: 10.1016/j.isci.2022.104258] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022] Open
Abstract
Flow of cerebrospinal fluid (CSF) through perivascular spaces (PVSs) in the brain delivers nutrients, clears metabolic waste, and causes edema formation. Brain-wide imaging cannot resolve PVSs, and high-resolution methods cannot access deep tissue. However, theoretical models provide valuable insight. We model the CSF pathway as a network of hydraulic resistances, using published parameter values. A few parameters (permeability of PVSs and the parenchyma, and dimensions of PVSs and astrocyte endfoot gaps) have wide uncertainties, so we focus on the limits of their ranges by analyzing different parametric scenarios. We identify low-resistance PVSs and high-resistance parenchyma as the only scenario that satisfies three essential criteria: that the flow be driven by a small pressure drop, exhibit good CSF perfusion throughout the cortex, and exhibit a substantial increase in flow during sleep. Our results point to the most important parameters, such as astrocyte endfoot gap dimensions, to be measured in future experiments. We model the CSF pathway as a network of hydraulic resistances Predictions are bracketed by analyzing parametric scenarios for unknown parameters Low-resistance PVSs and high-resistance parenchyma produce realistic flows Astrocyte endfoot gap size is among the important parameters to be measured
Collapse
Affiliation(s)
- Jeffrey Tithof
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis 55455, MN, USA
- Corresponding author
| | - Kimberly A.S. Boster
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| | - Peter A.R. Bork
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester 14642, NY, USA
| | - John H. Thomas
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| |
Collapse
|
154
|
Vacas S, Canales C, Deiner SG, Cole DJ. Perioperative Brain Health in the Older Adult: A Patient Safety Imperative. Anesth Analg 2022; 135:316-328. [PMID: 35584550 PMCID: PMC9288500 DOI: 10.1213/ane.0000000000006090] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
While people 65 years of age and older represent 16% of the population in the United States, they account for >40% of surgical procedures performed each year. Maintaining brain health after anesthesia and surgery is not only important to our patients, but it is also an increasingly important patient safety imperative for the specialty of anesthesiology. Aging is a complex process that diminishes the reserve of every organ system and often results in a patient who is vulnerable to the stress of surgery. The brain is no exception, and many older patients present with preoperative cognitive impairment that is undiagnosed. As we age, a number of changes occur in the human brain, resulting in a patient who is less resilient to perioperative stress, making older adults more susceptible to the phenotypic expression of perioperative neurocognitive disorders. This review summarizes the current scientific and clinical understanding of perioperative neurocognitive disorders and recommends patient-centered, age-focused interventions that can better mitigate risk, prevent harm, and improve outcomes for our patients. Finally, it discusses the emerging topic of sleep and cognitive health and other future frontiers of scientific inquiry that might inform clinical best practices.
Collapse
Affiliation(s)
- Susana Vacas
- From the Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Cecilia Canales
- From the Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Stacie G Deiner
- Department of Anesthesiology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Daniel J Cole
- From the Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
155
|
Pendlebury GA, Oro P, Haynes W, Byrnes TR, Keane J, Goldstein L. Advocacy for Change: An Osteopathic Review of Traumatic Brain Injury Among Combat Veterans. Cureus 2022; 14:e25051. [PMID: 35719755 PMCID: PMC9199571 DOI: 10.7759/cureus.25051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
As a "signature injury" of the Iraq and Afghanistan wars, traumatic brain injury (TBI) remains a major health concern among military service members. Traumatic brain injury is associated with a wide range of symptoms which may be cognitive, emotional, psychological, biochemical, and social in nature. Mild TBI (mTBI) ranks as the most common traumatic brain injury among veterans. Due to the absence of specific symptoms, mTBI diagnosis may be challenging in acute settings. Repetitive traumatic brain injury during combat deployments can lead to devastating chronic neurodegenerative diseases and other major life disruptions. Many cases of TBI remain undetected in veterans and may lead to long-term adverse comorbidities such as post-traumatic stress disorder (PTSD), suicide, alcohol disorders, psychiatric diagnoses, and service-related somatic dysfunctions. Veterans with TBI are almost twice as likely to die from suicide in comparison to veterans without a history of TBI. Veterans diagnosed with TBI experience significant comorbid conditions and thus advocacy for improved care is justified and necessary. Given the complexity and variation in the symptomatology of TBI, a personalized, multimodal approach is warranted in the evaluation and treatment of veterans with TBI and other associated conditions. As such, this review provides a broad overview of treatment options, with an emphasis on advocacy and osteopathic integration in the standard of care for veterans.
Collapse
|
156
|
Yu L, Hu X, Li H, Zhao Y. Perivascular Spaces, Glymphatic System and MR. Front Neurol 2022; 13:844938. [PMID: 35592469 PMCID: PMC9110928 DOI: 10.3389/fneur.2022.844938] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
The importance of the perivascular space (PVS) as one of the imaging markers of cerebral small vessel disease (CSVD) has been widely appreciated by the neuroradiologists. The PVS surrounds the small blood vessels in the brain and has a signal consistent with the cerebrospinal fluid (CSF) on MR. In a variety of physio-pathological statuses, the PVS may expand. The discovery of the cerebral glymphatic system has provided a revolutionary perspective to elucidate its pathophysiological mechanisms. Research on the function and pathogenesis of this system has become a prevalent topic among neuroradiologists. It is now believed that this system carries out the similar functions as the lymphatic system in other parts of the body and plays an important role in the removal of metabolic waste and the maintenance of homeostatic fluid circulation in the brain. In this article, we will briefly describe the composition of the cerebral glymphatic system, the influencing factors, the MR manifestations of the PVS and the related imaging technological advances. The aim of this research is to provide a reference for future clinical studies of the PVS and glymphatic system.
Collapse
Affiliation(s)
- Linya Yu
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaofei Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haitao Li
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Haitao Li
| | - Yilei Zhao
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Yilei Zhao
| |
Collapse
|
157
|
Silva J, Patricio F, Patricio-Martínez A, Santos-López G, Cedillo L, Tizabi Y, Limón ID. Neuropathological Aspects of SARS-CoV-2 Infection: Significance for Both Alzheimer's and Parkinson's Disease. Front Neurosci 2022; 16:867825. [PMID: 35592266 PMCID: PMC9111171 DOI: 10.3389/fnins.2022.867825] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Evidence suggests that SARS-CoV-2 entry into the central nervous system can result in neurological and/or neurodegenerative diseases. In this review, routes of SARS-Cov-2 entry into the brain via neuroinvasive pathways such as transcribrial, ocular surface or hematogenous system are discussed. It is argued that SARS-Cov-2-induced cytokine storm, neuroinflammation and oxidative stress increase the risk of developing neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Further studies on the effects of SARS-CoV-2 and its variants on protein aggregation, glia or microglia activation, and blood-brain barrier are warranted.
Collapse
Affiliation(s)
- Jaime Silva
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Felipe Patricio
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Gerardo Santos-López
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Atlixco, Mexico
| | - Lilia Cedillo
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
158
|
Lazic A, Balint V, Stanisavljevic Ninkovic D, Peric M, Stevanovic M. Reactive and Senescent Astroglial Phenotypes as Hallmarks of Brain Pathologies. Int J Mol Sci 2022; 23:ijms23094995. [PMID: 35563385 PMCID: PMC9100382 DOI: 10.3390/ijms23094995] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes, as the most abundant glial cells in the central nervous system, are tightly integrated into neural networks and participate in numerous aspects of brain physiology and pathology. They are the main homeostatic cells in the central nervous system, and the loss of astrocyte physiological functions and/or gain of pro-inflammatory functions, due to their reactivation or cellular senescence, can have profound impacts on the surrounding microenvironment with pathological outcomes. Although the importance of astrocytes is generally recognized, and both senescence and reactive astrogliosis have been extensively reviewed independently, there are only a few comparative overviews of these complex processes. In this review, we summarize the latest data regarding astrocyte reactivation and senescence, and outline similarities and differences between these phenotypes from morphological, functional, and molecular points of view. A special focus has been given to neurodegenerative diseases, where these phenotypic alternations of astrocytes are significantly implicated. We also summarize current perspectives regarding new advances in model systems based on astrocytes as well as data pointing to these glial cells as potential therapeutic targets.
Collapse
Affiliation(s)
- Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Correspondence:
| | - Vanda Balint
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Mina Peric
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11001 Belgrade, Serbia
| |
Collapse
|
159
|
de Donato A, Buonincontri V, Borriello G, Martinelli G, Mone P. The dopamine system: insights between kidney and brain. Kidney Blood Press Res 2022; 47:493-505. [PMID: 35378538 DOI: 10.1159/000522132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/21/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is one of the most common diseases in adult age and it is typical of older adults. Recent data suggest that almost half of the elders have CKD. It is now clear that CKD is accompanied, in the early stages, by cognitive impairment, together with depression and subtle abnormalities in motor control (such as gait and balance alterations). SUMMARY Several data suggest a link between brain dopamine and kidney diseases. Metabolic syndrome and diabetes can affect dopamine neuron survival (leading to Parkinson's Disease). Several uremic toxins in CKD (uric acid, indoxyl sulphate) and trace elements accumulating in CKD (aluminium, manganese) can also modify the dopaminergic system. Hormones produced by the kidney such as vitamin D are neuroprotective for dopamine neurons. Dopaminergic drugs are useful for the treatment of a common sleep disorder in CKD, the restless legs syndrome. However, experiments on animal models of CKD show conflicting results regarding a modification of dopamine neurons. KEY MESSAGES Several observations suggest a limited relevance of the dopaminergic system in CKD-related cognitive impairment. However, a common sleep disturbance in CKD, the restless leg syndrome, improves with dopaminergic drugs. Therefore, it remains to be established the role of the dopamine system in subtle motor dysfunction observed in CKD, such as tremors, gait alterations, and central sleep apnea.
Collapse
Affiliation(s)
- Antonio de Donato
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
| | - Veronica Buonincontri
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
| | - Gianmarco Borriello
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
| | - Giuseppe Martinelli
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
- ASL Napoli, Naples, Italy
| | - Pasquale Mone
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
- ASL Avellino, Avellino, Italy
| |
Collapse
|
160
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
161
|
Bonini F, Mosser S, Mor FM, Boutabla A, Burch P, Béduer A, Roux A, Braschler T. The Role of Interstitial Fluid Pressure in Cerebral Porous Biomaterial Integration. Brain Sci 2022; 12:417. [PMID: 35447953 PMCID: PMC9040716 DOI: 10.3390/brainsci12040417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/05/2023] Open
Abstract
Recent advances in biomaterials offer new possibilities for brain tissue reconstruction. Biocompatibility, provision of cell adhesion motives and mechanical properties are among the present main design criteria. We here propose a radically new and potentially major element determining biointegration of porous biomaterials: the favorable effect of interstitial fluid pressure (IFP). The force applied by the lymphatic system through the interstitial fluid pressure on biomaterial integration has mostly been neglected so far. We hypothesize it has the potential to force 3D biointegration of porous biomaterials. In this study, we develop a capillary hydrostatic device to apply controlled in vitro interstitial fluid pressure and study its effect during 3D tissue culture. We find that the IFP is a key player in porous biomaterial tissue integration, at physiological IFP levels, surpassing the known effect of cell adhesion motives. Spontaneous electrical activity indicates that the culture conditions are not harmful for the cells. Our work identifies interstitial fluid pressure at physiological negative values as a potential main driver for tissue integration into porous biomaterials. We anticipate that controlling the IFP level could narrow the gap between in vivo and in vitro and therefore decrease the need for animal screening in biomaterial design.
Collapse
Affiliation(s)
- Fabien Bonini
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| | - Sébastien Mosser
- Neurix SA, Avenue de la Roseraie 64, CH-1022 Geneva, Switzerland;
| | - Flavio Maurizio Mor
- Haute École du Paysage, d’Ingénierie et d’Architecture de Genève, Haute École Spécialisée de Suisse Occidentale (HEPIA HES-SO), University of Applied Sciences and Arts Western Switzerland, CH-1202 Geneva, Switzerland; (F.M.M.); (A.R.)
| | - Anissa Boutabla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| | - Patrick Burch
- Volumina-Medical SA, Route de la Corniche 5, CH-1066 Epalinges, Switzerland;
| | - Amélie Béduer
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
- Volumina-Medical SA, Route de la Corniche 5, CH-1066 Epalinges, Switzerland;
| | - Adrien Roux
- Haute École du Paysage, d’Ingénierie et d’Architecture de Genève, Haute École Spécialisée de Suisse Occidentale (HEPIA HES-SO), University of Applied Sciences and Arts Western Switzerland, CH-1202 Geneva, Switzerland; (F.M.M.); (A.R.)
| | - Thomas Braschler
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| |
Collapse
|
162
|
Onoda A, Hagiwara S, Kubota N, Yanagita S, Takeda K, Umezawa M. A Novel Staining Method for Detection of Brain Perivascular Injuries Induced by Nanoparticle: Periodic Acid-Schiff and Immunohistochemical Double-Staining. FRONTIERS IN TOXICOLOGY 2022; 4:825984. [PMID: 35391824 PMCID: PMC8979793 DOI: 10.3389/ftox.2022.825984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: To protect developing brain from any unfavorable effects, it is necessary to construct experimental techniques that can sensitively detect and evaluate developmental toxicity. We have previously shown that brain perivascular tissues, especially perivascular macrophages (PVMs), respond sensitively even to weak stimuli by foreign toxicants such as low-dose exposure to nanoparticle. This paper shows the protocol of a novel staining method that enables easy detection and rapid evaluation of brain perivascular abnormalities.Methods: As weak stimulus, low-dose of carbon black nanoparticle (95 μg/kg) or titanium dioxide nanoparticle (100 μg/kg) was intranasally administered to pregnant mice at gestational days 5 and 9. The offspring brains were used to confirm the properties of PVMs and to find suitable protocols for the detection and evaluation of the mild denaturation of PVMs. Furthermore, various procedures of novel combinational double staining including periodic acid-Schiff (PAS) staining and immunohistochemistry were examined. In addition, we checked the alterations in neurotransmitter levels and the behaviors of the offspring.Results and discussion: Maternal exposure to low-dose of nanoparticle at levels where no significant effects on the brain were observed, such as abnormal behavior, alteration of neurotransmitter levels, or microglial activation, resulted in mild denaturation of the PVMs, which was captured by PAS staining. However, it was difficult to detect and determine slight histopathological alterations. Therefore, we established PAS-immunohistochemical double-staining method for the brain. This double staining method enabled easy detection and rapid evaluation of brain perivascular abnormalities and the relationship between PVMs and the surrounding cells. In addition, this double staining allows evaluation of the histopathological denaturation of the PVMs and the associated abnormalities in the surrounding tissues in the same section.Conclusion: The slight responses of brain perivascular tissues, such as mild denaturation of PVMs, were sensitively and easily determined by the PAS-immunohistochemical double-staining method. This double staining method is a powerful tool to assess brain perivascular injuries including PVM denaturation and the relationship between the expression of various molecules and the morphology of PVMs. We propose that the observation of the tissue around brain blood vessels using the double staining provides potential endpoints to evaluate developmental neurotoxicity.
Collapse
Affiliation(s)
- Atsuto Onoda
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Japan
- *Correspondence: Atsuto Onoda,
| | - Shin Hagiwara
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Natsuko Kubota
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan
- Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Japan
- Institute of Arts and Sciences, Tokyo University of Science, Noda, Japan
| | - Shinya Yanagita
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan
- Institute of Arts and Sciences, Tokyo University of Science, Noda, Japan
| | - Ken Takeda
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Masakazu Umezawa
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
163
|
Li L, Ding G, Zhang L, Davoodi-Bojd E, Chopp M, Li Q, Zhang ZG, Jiang Q. Aging-Related Alterations of Glymphatic Transport in Rat: In vivo Magnetic Resonance Imaging and Kinetic Study. Front Aging Neurosci 2022; 14:841798. [PMID: 35360203 PMCID: PMC8960847 DOI: 10.3389/fnagi.2022.841798] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Impaired glymphatic waste clearance function during brain aging leads to the accumulation of metabolic waste and neurotoxic proteins (e.g., amyloid-β, tau) which contribute to neurological disorders. However, how the age-related glymphatic dysfunction exerts its effects on different cerebral regions and affects brain waste clearance remain unclear. Methods We investigated alterations of glymphatic transport in the aged rat brain using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and advanced kinetic modeling. Healthy young (3-4 months) and aged (18-20 months) male rats (n = 12/group) underwent the identical MRI protocol, including T2-weighted imaging and 3D T1-weighted imaging with intracisternal administration of contrast agent (Gd-DTPA). Model-derived parameters of infusion rate and clearance rate, characterizing the kinetics of cerebrospinal fluid (CSF) tracer transport via the glymphatic system, were evaluated in multiple representative brain regions. Changes in the CSF-filled cerebral ventricles were measured using contrast-induced time signal curves (TSCs) in conjunction with structural imaging. Results Compared to the young brain, an overall impairment of glymphatic transport function was detected in the aged brain, evidenced by the decrease in both infusion and clearance rates throughout the brain. Enlarged ventricles in parallel with reduced efficiency in CSF transport through the ventricular regions were present in the aged brain. While the age-related glymphatic dysfunction was widespread, our kinetic quantification demonstrated that its impact differed considerably among cerebral regions with the most severe effect found in olfactory bulb, indicating the heterogeneous and regional preferential alterations of glymphatic function. Conclusion The robust suppression of glymphatic activity in the olfactory bulb, which serves as one of major efflux routes for brain waste clearance, may underlie, in part, age-related neurodegenerative diseases associated with neurotoxic substance accumulation. Our data provide new insight into the cerebral regional vulnerability to brain functional change with aging.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | | | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
164
|
Salehpour F, Khademi M, Bragin DE, DiDuro JO. Photobiomodulation Therapy and the Glymphatic System: Promising Applications for Augmenting the Brain Lymphatic Drainage System. Int J Mol Sci 2022; 23:ijms23062975. [PMID: 35328396 PMCID: PMC8950470 DOI: 10.3390/ijms23062975] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
The glymphatic system is a glial-dependent waste clearance pathway in the central nervous system, devoted to drain away waste metabolic products and soluble proteins such as amyloid-beta. An impaired brain glymphatic system can increase the incidence of neurovascular, neuroinflammatory, and neurodegenerative diseases. Photobiomodulation (PBM) therapy can serve as a non-invasive neuroprotective strategy for maintaining and optimizing effective brain waste clearance. In this review, we discuss the crucial role of the glymphatic drainage system in removing toxins and waste metabolites from the brain. We review recent animal research on the neurotherapeutic benefits of PBM therapy on glymphatic drainage and clearance. We also highlight cellular mechanisms of PBM on the cerebral glymphatic system. Animal research has shed light on the beneficial effects of PBM on the cerebral drainage system through the clearance of amyloid-beta via meningeal lymphatic vessels. Finally, PBM-mediated increase in the blood–brain barrier permeability with a subsequent rise in Aβ clearance from PBM-induced relaxation of lymphatic vessels via a vasodilation process will be discussed. We conclude that PBM promotion of cranial and extracranial lymphatic system function might be a promising strategy for the treatment of brain diseases associated with cerebrospinal fluid outflow abnormality.
Collapse
Affiliation(s)
- Farzad Salehpour
- College for Light Medicine and Photobiomodulation, D-82319 Starnberg, Germany;
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
| | - Mahsa Khademi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51666, Iran;
| | - Denis E. Bragin
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| | - Joseph O. DiDuro
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
- Correspondence: ; Tel.: +1-(845)-203-9204
| |
Collapse
|
165
|
Buckley MW, McGavern DB. Immune dynamics in the CNS and its barriers during homeostasis and disease. Immunol Rev 2022; 306:58-75. [PMID: 35067941 PMCID: PMC8852772 DOI: 10.1111/imr.13066] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
The central nervous system (CNS) has historically been viewed as an immunologically privileged site, but recent studies have uncovered a vast landscape of immune cells that reside primarily along its borders. While microglia are largely responsible for surveying the parenchyma, CNS barrier sites are inhabited by a plethora of different innate and adaptive immune cells that participate in everything from the defense against microbes to the maintenance of neural function. Static and dynamic imaging studies have revolutionized the field of neuroimmunology by providing detailed maps of CNS immune cells as well as information about how these cells move, organize, and interact during steady-state and inflammatory conditions. These studies have also redefined our understanding of neural-immune interactions at a cellular level and reshaped our conceptual view of immune privilege in this specialized compartment. This review will focus on insights gained using imaging techniques in the field of neuroimmunology, with an emphasis on anatomy and CNS immune dynamics during homeostasis, infectious diseases, injuries, and aging.
Collapse
Affiliation(s)
- Monica W. Buckley
- Viral Immunology and Intravital Imaging Section National Institute of Neurological Disorders and Stroke National Institutes of Health Bethesda Maryland USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section National Institute of Neurological Disorders and Stroke National Institutes of Health Bethesda Maryland USA
| |
Collapse
|
166
|
Perosa V, Oltmer J, Munting LP, Freeze WM, Auger CA, Scherlek AA, van der Kouwe AJ, Iglesias JE, Atzeni A, Bacskai BJ, Viswanathan A, Frosch MP, Greenberg SM, van Veluw SJ. Perivascular space dilation is associated with vascular amyloid-β accumulation in the overlying cortex. Acta Neuropathol 2022; 143:331-348. [PMID: 34928427 PMCID: PMC9047512 DOI: 10.1007/s00401-021-02393-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/10/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022]
Abstract
Perivascular spaces (PVS) are compartments surrounding cerebral blood vessels that become visible on MRI when enlarged. Enlarged PVS (EPVS) are commonly seen in patients with cerebral small vessel disease (CSVD) and have been suggested to reflect dysfunctional perivascular clearance of soluble waste products from the brain. In this study, we investigated histopathological correlates of EPVS and how they relate to vascular amyloid-β (Aβ) in cerebral amyloid angiopathy (CAA), a form of CSVD that commonly co-exists with Alzheimer's disease (AD) pathology. We used ex vivo MRI, semi-automatic segmentation and validated deep-learning-based models to quantify EPVS and associated histopathological abnormalities. Severity of MRI-visible PVS during life was significantly associated with severity of MRI-visible PVS on ex vivo MRI in formalin fixed intact hemispheres and corresponded with PVS enlargement on histopathology in the same areas. EPVS were located mainly around the white matter portion of perforating cortical arterioles and their burden was associated with CAA severity in the overlying cortex. Furthermore, we observed markedly reduced smooth muscle cells and increased vascular Aβ accumulation, extending into the WM, in individually affected vessels with an EPVS. Overall, these findings are consistent with the notion that EPVS reflect impaired outward flow along arterioles and have implications for our understanding of perivascular clearance mechanisms, which play an important role in the pathophysiology of CAA and AD.
Collapse
Affiliation(s)
- Valentina Perosa
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA.
- Department of Neurology, Otto-Von-Guericke University, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Jan Oltmer
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Leon P Munting
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Whitney M Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, The Netherlands
| | - Corinne A Auger
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
| | - Ashley A Scherlek
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Andre J van der Kouwe
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Juan Eugenio Iglesias
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Centre for Medical Image Computing, University College London, London, UK
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alessia Atzeni
- Centre for Medical Image Computing, University College London, London, UK
| | - Brian J Bacskai
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
| | - Matthew P Frosch
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
167
|
Lee DA, Park BS, Ko J, Park SH, Park JH, Kim IH, Lee YJ, Park KM. Glymphatic system function in patients with newly diagnosed focal epilepsy. Brain Behav 2022; 12:e2504. [PMID: 35107879 PMCID: PMC8933756 DOI: 10.1002/brb3.2504] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 01/01/2022] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The aim of this study was to analyze the glymphatic system function and its relationship with clinical characteristics, global diffusion tensor imaging (DTI) parameters, and global structural connectivity in treatment-naïve patients with newly diagnosed focal epilepsy. METHODS This retrospective single-center study investigated patients with focal epilepsy and healthy controls. All participants underwent routine brain magnetic resonance imaging and DTI. DTI analysis along the perivascular space (DTI-ALPS) was used to evaluate glymphatic system function. We also calculated the measures of global DTI parameters, including whole-brain fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD), and performed a graph theoretical network analysis to measure global structural connectivity. RESULTS A total of 109 patients with focal epilepsy and 88 healthy controls were analyzed. There were no significant differences in the DTI-ALPS index (1.67 vs. 1.68, p = 0.861) between the groups. However, statistically significant associations were found between the DTI-ALPS index and age (r = -0.242, p = 0.01), FA (r = 0.257, p = 0.007), MD (r = -0.469, p < 0.001), AD (r = -0.303, p = 0.001), RD (r = -0.434, p < 0.001), and the assortative coefficient (r = 0.230, p = 0.016) in patients with focal epilepsy. CONCLUSION The main finding of this study is that DTI-ALPS index is significantly correlated with global DTI parameters and structural connectivity measures of the brain in patients with focal epilepsy. In addition, DTI-ALPS index decreases with age in these patients. We conclude that the DTI-ALPS index can be used to investigate glymphatic system function in patients with focal epilepsy.
Collapse
Affiliation(s)
- Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Bong Soo Park
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Junghae Ko
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Si Hyung Park
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Jin-Han Park
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Il Hwan Kim
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Yoo Jin Lee
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
168
|
Tsutsumi S, Ono H, Ishii H. Hyperintense areas in the cisternal segments of the cranial nerves: a magnetic resonance imaging study. Surg Radiol Anat 2022; 44:503-509. [PMID: 35195771 DOI: 10.1007/s00276-022-02902-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE The study aimed to explore hyperintense areas in the cisternal segments of the cranial nerves using magnetic resonance imaging (MRI). METHODS Seventy outpatients underwent thin-sliced, coronal constructive interference steady-state (CISS) sequence and sagittal T2-weighted MRI following conventional MRI examination. RESULTS With the coronal CISS sequence, hyperintense areas were located in the central parts of the olfactory bulbs in 65.7% of patients. For the intracranial optic nerve and optic chiasm, hyperintense areas were detected in 98.6% of the CISS sequences and 100% of the T2-weighted images. In the optic tract, hyperintense areas were detected in 51.4% of cases. In 35% of the patients who underwent the CISS sequence, the intracranial optic nerves were considerably compressed by the internal carotid and anterior cerebral arteries, with hyperintense areas similar to those in patients without vascular compression. Hyperintense areas of the cisternal segments of the oculomotor nerve and trigeminal root were identified in 52.9% and 87.1% of the patients, respectively. CONCLUSIONS The hyperintense areas found within the cisternal segments of the cranial nerves delineated on the coronal CISS sequence and sagittal T2-weighted imaging may indicate the intracranial part of the glymphatic pathway through the cranial nerves. The cranial nerves may function as part of the glymphatic pathway.
Collapse
Affiliation(s)
- Satoshi Tsutsumi
- Department of Neurological Surgery, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu, Chiba, 279-0021, Japan.
| | - Hideo Ono
- Division of Radiological Technology, Medical Satellite Yaesu Clinic, Tokyo, Japan
| | - Hisato Ishii
- Department of Neurological Surgery, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu, Chiba, 279-0021, Japan
| |
Collapse
|
169
|
Wardlaw JM, Benveniste H, Williams A. Cerebral Vascular Dysfunctions Detected in Human Small Vessel Disease and Implications for Preclinical Studies. Annu Rev Physiol 2022; 84:409-434. [PMID: 34699267 DOI: 10.1146/annurev-physiol-060821-014521] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral small vessel disease (SVD) is highly prevalent and a common cause of ischemic and hemorrhagic stroke and dementia, yet the pathophysiology is poorly understood. Its clinical expression is highly varied, and prognostic implications are frequently overlooked in clinics; thus, treatment is currently confined to vascular risk factor management. Traditionally, SVD is considered the small vessel equivalent of large artery stroke (occlusion, rupture), but data emerging from human neuroimaging and genetic studies refute this, instead showing microvessel endothelial dysfunction impacting on cell-cell interactions and leading to brain damage. These dysfunctions reflect defects that appear to be inherited and secondary to environmental exposures, including vascular risk factors. Interrogation in preclinical models shows consistent and converging molecular and cellular interactions across the endothelial-glial-neural unit that increasingly explain the human macroscopic observations and identify common patterns of pathology despite different triggers. Importantly, these insights may offer new targets for therapeutic intervention focused on restoring endothelial-glial physiology.
Collapse
Affiliation(s)
- Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences; UK Dementia Research Institute; and Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom;
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
170
|
Zhang W, Bai J, Hang K, Xu J, Zhou C, Li L, Wang Z, Wang Y, Wang K, Xue D. Role of Lysosomal Acidification Dysfunction in Mesenchymal Stem Cell Senescence. Front Cell Dev Biol 2022; 10:817877. [PMID: 35198560 PMCID: PMC8858834 DOI: 10.3389/fcell.2022.817877] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been widely used as a potential treatment for a variety of diseases. However, the contradiction between the low survival rate of transplanted cells and the beneficial therapeutic effects has affected its clinical use. Lysosomes as organelles at the center of cellular recycling and metabolic signaling, play essential roles in MSC homeostasis. In the first part of this review, we summarize the role of lysosomal acidification dysfunction in MSC senescence. In the second part, we summarize some of the potential strategies targeting lysosomal proteins to enhance the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Weijun Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinwu Bai
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Hang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianxiang Xu
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengwei Zhou
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lijun Li
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongxiang Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yibo Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kanbin Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Deting Xue,
| |
Collapse
|
171
|
Liu J, Guo Y, Zhang C, Zeng Y, Luo Y, Wang G. Clearance Systems in the Brain, From Structure to Function. Front Cell Neurosci 2022; 15:729706. [PMID: 35173581 PMCID: PMC8841422 DOI: 10.3389/fncel.2021.729706] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
As the most metabolically active organ in the body, there is a recognized need for pathways that remove waste proteins and neurotoxins from the brain. Previous research has indicated potential associations between the clearance system in the brain and the pathological conditions of the central nervous system (CNS), due to its importance, which has attracted considerable attention recently. In the last decade, studies of the clearance system have been restricted to the glymphatic system. However, removal of toxic and catabolic waste by-products cannot be completed independently by the glymphatic system, while no known research or article has focused on a comprehensive overview of the structure and function of the clearance system. This thesis addresses a neglected aspect of linkage between the structural composition and main components as well as the role of neural cells throughout the clearance system, which found evidence that the components of CNS including the glymphatic system and the meningeal lymphatic system interact with a neural cell, such as astrocytes and microglia, to carry out vital clearance functions. As a result of this evidence that can contribute to a better understanding of the clearance system, suggestions were identified for further clinical intervention development of severe conditions caused by the accumulation of metabolic waste products and neurotoxins in the brain, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Yunzhi Guo
- Xiangya Medical College of Central South University, Changsha, China
| | - Chengyue Zhang
- Xiangya Medical College of Central South University, Changsha, China
| | - Yang Zeng
- Xiangya Medical College of Central South University, Changsha, China
| | - Yongqi Luo
- Xiangya Medical College of Central South University, Changsha, China
| | - Gaiqing Wang
- Shanxi Medical University, Taiyuan, China
- Department of Neurology, Affiliated Sanya Central Hospital of Hainan Medical University, Sanya, China
- *Correspondence: Gaiqing Wang, ,
| |
Collapse
|
172
|
Deffner F, Gleiser C, Mattheus U, Wagner A, Neckel PH, Fallier-Becker P, Hirt B, Mack AF. Aquaporin-4 expression in the human choroid plexus. Cell Mol Life Sci 2022; 79:90. [PMID: 35072772 PMCID: PMC8785037 DOI: 10.1007/s00018-022-04136-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 01/15/2023]
Abstract
The choroid plexus (CP) consists of specialized ependymal cells and underlying blood vessels and stroma producing the bulk of the cerebrospinal fluid (CSF). CP epithelial cells are considered the site of the internal blood-cerebrospinal fluid barrier, show epithelial characteristics (basal lamina, tight junctions), and express aquaporin-1 (AQP1) apically. In this study, we analyzed the expression of aquaporins in the human CP using immunofluorescence and qPCR. As previously reported, AQP1 was expressed apically in CP epithelial cells. Surprisingly, and previously unknown, many cells in the CP epithelium were also positive for aquaporin-4 (AQP4), normally restricted to ventricle-lining ependymal cells and astrocytes in the brain. Expression of AQP1 and AQP4 was found in the CP of all eight body donors investigated (3 males, 5 females; age 74–91). These results were confirmed by qPCR, and by electron microscopy detecting orthogonal arrays of particles. To find out whether AQP4 expression correlated with the expression pattern of relevant transport-related proteins we also investigated expression of NKCC1, and Na/K-ATPase. Immunostaining with NKCC1 was similar to AQP1 and revealed no particular pattern related to AQP4. Co-staining of AQP4 and Na/K-ATPase indicated a trend for an inverse correlation of their expression. We hypothesized that AQP4 expression in the CP was caused by age-related changes. To address this, we investigated mouse brains from young (2 months), adult (12 months) and old (30 months) mice. We found a significant increase of AQP4 on the mRNA level in old mice compared to young and adult animals. Taken together, we provide evidence for AQP4 expression in the CP of the aging brain which likely contributes to the water flow through the CP epithelium and CSF production. In two alternative hypotheses, we discuss this as a beneficial compensatory, or a detrimental mechanism influencing the previously observed CSF changes during aging.
Collapse
Affiliation(s)
- Felix Deffner
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Corinna Gleiser
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Ulrich Mattheus
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Andreas Wagner
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Peter H Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Petra Fallier-Becker
- Institute of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Bernhard Hirt
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany
| | - Andreas F Mack
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstr. 3, 72074, Tübingen, Germany.
| |
Collapse
|
173
|
Albayram MS, Smith G, Tufan F, Tuna IS, Bostancıklıoğlu M, Zile M, Albayram O. Non-invasive MR imaging of human brain lymphatic networks with connections to cervical lymph nodes. Nat Commun 2022; 13:203. [PMID: 35017525 PMCID: PMC8752739 DOI: 10.1038/s41467-021-27887-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 12/21/2021] [Indexed: 12/15/2022] Open
Abstract
Meningeal lymphatic vessels have been described in animal studies, but limited comparable data is available in human studies. Here we show dural lymphatic structures along the dural venous sinuses in dorsal regions and along cranial nerves in the ventral regions in the human brain. 3D T2-Fluid Attenuated Inversion Recovery magnetic resonance imaging relies on internal signals of protein rich lymphatic fluid rather than contrast media and is used in the present study to visualize the major human dural lymphatic structures. Moreover we detect direct connections between lymphatic fluid channels along the cranial nerves and vascular structures and the cervical lymph nodes. We also identify age-related cervical lymph node atrophy and thickening of lymphatics channels in both dorsal and ventral regions, findings which reflect the reduced lymphatic output of the aged brain.
Collapse
Affiliation(s)
- Mehmet Sait Albayram
- Department of Radiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
| | - Garrett Smith
- Department of Radiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Fatih Tufan
- Geriatrician (PP), Silivrikapi Mh. Hisaralti Cd, Istanbul, 34093, Turkey
| | - Ibrahim Sacit Tuna
- Department of Radiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | | | - Michael Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Division of Cardiology, Department of Medicine, Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC, 29425, USA
| | - Onder Albayram
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
174
|
Yu N, Sinclair B, Posada LMG, Chen Z, Di Q, Lin X, Kolbe S, Hlauschek G, Kwan P, Law M. Asymmetric distribution of enlarged perivascular spaces in centrum semiovale may be associated with epilepsy after acute ischemic stroke. CNS Neurosci Ther 2022; 28:343-353. [PMID: 34981639 PMCID: PMC8841310 DOI: 10.1111/cns.13786] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/12/2023] Open
Abstract
Objective To investigate the factors influencing enlarged perivascular space (EPVS) characteristics at the onset of acute ischemic stroke (AIS), and whether the PVS characteristics can predict later post‐stroke epilepsy (PSE). Methods A total of 312 patients with AIS were identified, of whom 58/312 (18.6%) developed PSE. Twenty healthy participants were included as the control group. The number of PVS in the basal ganglia (BG), centrum semiovale (CS), and midbrain (MB) was manually calculated on T2‐weighted MRI. The scores and asymmetry index (AI) of EPVS in each region were compared among the enrolled participants. Other potential risk factors for PSE were also analyzed, including NIHSS at admission and stroke etiologies. Results The EPVS scores were significantly higher in the bilateral BG and CS of AIS patients compared to those of the control group (both p < 0.01). No statistical differences in EPVS scores in BG, CS, and MB were obtained between the PSE group and the nonepilepsy AIS group (all p > 0.01). However, markedly different AI scores in CS were found between the PSE group and the nonepilepsy AIS group (p = 0.004). Multivariable analysis showed that high asymmetry index of EPVS (AI≥0.2) in CS was an independent predictor for PSE (OR = 3.7, 95% confidence interval 1.5–9.1, p = 0.004). Conclusions Asymmetric distribution of EPVS in CS may be an independent risk factor and a novel imaging biomarker for the development of PSE. Further studies to understand the mechanisms of this association and confirmation with larger patient populations are warranted.
Collapse
Affiliation(s)
- Nian Yu
- Department of Neurology, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurology, Royal Melbourne Hospital, Melbourne, Vic., Australia.,Department of Radiology, Alfred Hospital, Melbourne, Vic., Australia
| | - Benjamin Sinclair
- Department of Neuroscience, Monash University, Melbourne, Vic., Australia.,Department of Neurology, Alfred Hospital, Melbourne, Vic., Australia
| | | | - Zhibin Chen
- Department of Neuroscience, Monash University, Melbourne, Vic., Australia
| | - Qing Di
- Department of Neurology, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xingjian Lin
- Department of Neurology, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Scott Kolbe
- Department of Neuroscience, Monash University, Melbourne, Vic., Australia
| | - Gernot Hlauschek
- National Centre for Epilepsy, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | - Patrick Kwan
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Vic., Australia.,Department of Neuroscience, Monash University, Melbourne, Vic., Australia.,Department of Neurology, Alfred Hospital, Melbourne, Vic., Australia.,Department of Medicine, University of Melbourne, Melbourne, Vic., Australia
| | - Meng Law
- Department of Radiology, Alfred Hospital, Melbourne, Vic., Australia.,Department of Neuroscience, Monash University, Melbourne, Vic., Australia.,Department of Neurological Surgery, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
175
|
Neuroimmune contributions to Alzheimer's disease: a focus on human data. Mol Psychiatry 2022; 27:3164-3181. [PMID: 35668160 PMCID: PMC9168642 DOI: 10.1038/s41380-022-01637-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
The past decade has seen the convergence of a series of new insights that arose from genetic and systems analyses of Alzheimer's disease (AD) with a wealth of epidemiological data from a variety of fields; this resulted in renewed interest in immune responses as important, potentially causal components of AD. Here, we focus primarily on a review of human data which has recently yielded a set of robust, reproducible results that exist in a much larger universe of conflicting reports stemming from small studies with important limitations in their study design. Thus, we are at an important crossroads in efforts to first understand at which step of the long, multiphasic course of AD a given immune response may play a causal role and then modulate this response to slow or block the pathophysiology of AD. We have a wealth of new experimental tools, analysis methods, and capacity to sample human participants at large scale longitudinally; these resources, when coupled to a foundation of reproducible results and novel study designs, will enable us to monitor human immune function in the CNS at the level of complexity that is required while simultaneously capturing the state of the peripheral immune system. This integration of peripheral and central perturbations in immune responses results in pathologic responses in the central nervous system parenchyma where specialized cellular microenvironments composed of multiple cell subtypes respond to these immune perturbations as well as to environmental exposures, comorbidities and the impact of the advancing life course. Here, we offer an overview that seeks to illustrate the large number of interconnecting factors that ultimately yield the neuroimmune component of AD.
Collapse
|
176
|
Mehramiz M, Porter T, Laws SM, Rainey-Smith SR. Sleep, Sirtuin 1 and Alzheimer's disease: A review. AGING BRAIN 2022; 2:100050. [PMID: 36908890 PMCID: PMC9997138 DOI: 10.1016/j.nbas.2022.100050] [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: 09/09/2021] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Sleep plays a major role in brain health, and cognition. Disrupted sleep is a well-described symptom of Alzheimer's disease (AD). However, accumulating evidence suggests suboptimal sleep also increases AD risk. The deacetylase Sirtuin 1 (Sirt 1), encoded by the SIRT1 gene, impacts sleep via its relationship to wake-sleep neurotransmitters and somnogens. Evidence from animal and human studies supports a significant and complex relationship between sleep, Sirt 1/ SIRT1 and AD. Numerous hypotheses attempt to explain the critical impact of Sirt 1/ SIRT1 on wake- and sleep- promoting neurons, their related mechanisms and neurotransmitters. However, there is a paucity of studies assessing the interaction between sleep and Sirt 1/ SIRT1, as a principal component of sleep regulation, on AD pathology. In this review, we explore the potential association between Sirt 1/ SIRT1, sleep, and AD aetiology. Given sleep is a likely modifiable risk factor for AD, and recent studies suggest Sirt 1/ SIRT1 activation can be modulated by lifestyle or dietary approaches, further research in this area is required to explore its potential as a target for AD prevention and treatment.
Collapse
Affiliation(s)
- Mehrane Mehramiz
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
177
|
Tran AA, De Smet M, Grant GD, Khoo TK, Pountney DL. Investigating the Convergent Mechanisms between Major Depressive Disorder and Parkinson's Disease. Complex Psychiatry 2021; 6:47-61. [PMID: 34883500 DOI: 10.1159/000512657] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/23/2020] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder (MDD) affects more than cognition, having a temporal relationship with neuroinflammatory pathways of Parkinson's disease (PD). Although this association is supported by epidemiological and clinical studies, the underlying mechanisms are unclear. Microglia and astrocytes play crucial roles in the pathophysiology of both MDD and PD. In PD, these cells can be activated by misfolded forms of the protein α-synuclein to release cytokines that can interact with multiple different physiological processes to produce depressive symptoms, including monoamine transport and availability, the hypothalamus-pituitary axis, and neurogenesis. In MDD, glial cell activation can be induced by peripheral inflammatory agents that cross the blood-brain barrier and/or c-Fos signalling from neurons. The resulting neuroinflammation can cause neurodegeneration due to oxidative stress and glutamate excitotoxicity, contributing to PD pathology. Astrocytes are another major link due to their recognized role in the glymphatic clearance mechanism. Research suggesting that MDD causes astrocytic destruction or structural atrophy highlights the possibility that accumulation of α-synuclein in the brain is facilitated as the brain cannot adequately clear the protein aggregates. This review examines research into the overlapping pathophysiology of MDD and PD with particular focus on the roles of glial cells and neuroinflammation.
Collapse
Affiliation(s)
- Angela A Tran
- School of Medical Science, Griffith University, Southport, Queensland, Australia.,School of Medicine, Griffith University, Southport, Queensland, Australia
| | - Myra De Smet
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Gary D Grant
- School of Pharmacy and Pharmacology, Griffith University, Southport, Queensland, Australia
| | - Tien K Khoo
- School of Medicine, Griffith University, Southport, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - Dean L Pountney
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
178
|
Ma X, Li S, Li C, Wang R, Chen M, Chen H, Su W. Diffusion Tensor Imaging Along the Perivascular Space Index in Different Stages of Parkinson's Disease. Front Aging Neurosci 2021; 13:773951. [PMID: 34867300 PMCID: PMC8634754 DOI: 10.3389/fnagi.2021.773951] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/14/2021] [Indexed: 01/11/2023] Open
Abstract
Background: The aim of this study was to evaluate the glymphatic system activity in patients with Parkinson’s disease (PD) using the diffusion tensor image analysis along the perivascular space (DTI-ALPS) methods. Methods: In total, 71 patients with idiopathic PD and 36 age- and sex-matched normal controls (NCs) were involved. Patients with PD were divided into early (n = 35) and late (n = 36) subgroups, based on Hoehn and Yahr (HY) stages. We calculated the diffusivity along the perivascular spaces (ALPS), as well as projection fibers and association fibers separately, to acquire the ALPS index. Enlarged perivascular spaces (EPVS) and periventricular white matter hyperintensities were also rated. Differences in ALPS index between the PD group and NCs and between two PD subgroups and NCs were compared. In addition, a multivariate logistic regression analysis was conducted to investigate the association between ALPS index and clinical variables. Results: Patients with PD revealed lower ALPS index than NCs (p = 0.010). The late PD group exhibited significantly lower ALPS index than NCs (p = 0.006). However, there were no marked differences noticed in ALPS index between NCs and early PD group and between the two PD subgroups. In the early PD group, there was a significantly positive correlation between ALPS index and Mini-Mental State Examination (MMSE) score (β = 0.021, p = 0.029) and a negative correlation between ALPS index and EPVS score (β = −0.050, p = 0.034), after controlling for multiple variables. In the late PD group, ALPS index was inversely associated with age (β = −0.012, p = 0.004). Conclusion: Impairment of the glymphatic system is involved in PD. DTI-ALPS index could be a promising biomarker of glymphatic system in PD.
Collapse
Affiliation(s)
- Xinxin Ma
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuhua Li
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunmei Li
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Wang
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Chen
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Haibo Chen
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wen Su
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
179
|
Ray LA, Pike M, Simon M, Iliff JJ, Heys JJ. Quantitative analysis of macroscopic solute transport in the murine brain. Fluids Barriers CNS 2021; 18:55. [PMID: 34876169 PMCID: PMC8650464 DOI: 10.1186/s12987-021-00290-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Understanding molecular transport in the brain is critical to care and prevention of neurological disease and injury. A key question is whether transport occurs primarily by diffusion, or also by convection or dispersion. Dynamic contrast-enhanced (DCE-MRI) experiments have long reported solute transport in the brain that appears to be faster than diffusion alone, but this transport rate has not been quantified to a physically relevant value that can be compared to known diffusive rates of tracers. METHODS In this work, DCE-MRI experimental data is analyzed using subject-specific finite-element models to quantify transport in different anatomical regions across the whole mouse brain. The set of regional effective diffusivities ([Formula: see text]), a transport parameter combining all mechanisms of transport, that best represent the experimental data are determined and compared to apparent diffusivity ([Formula: see text]), the known rate of diffusion through brain tissue, to draw conclusions about dominant transport mechanisms in each region. RESULTS In the perivascular regions of major arteries, [Formula: see text] for gadoteridol (550 Da) was over 10,000 times greater than [Formula: see text]. In the brain tissue, constituting interstitial space and the perivascular space of smaller blood vessels, [Formula: see text] was 10-25 times greater than [Formula: see text]. CONCLUSIONS The analysis concludes that convection is present throughout the brain. Convection is dominant in the perivascular space of major surface and branching arteries (Pe > 1000) and significant to large molecules (> 1 kDa) in the combined interstitial space and perivascular space of smaller vessels (not resolved by DCE-MRI). Importantly, this work supports perivascular convection along penetrating blood vessels.
Collapse
Affiliation(s)
- Lori A Ray
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA
| | - Martin Pike
- Advanced Imaging Research Center, Oregon Health and Sciences University, Portland, USA
| | - Matthew Simon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, USA
- Neuroscience Graduate Program, Oregon Health and Science University, Portland, USA
- Denali Therapeutics, San Francisco, USA
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, USA
| | - Jeffrey J Heys
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA.
| |
Collapse
|
180
|
Gireud-Goss M, Mack AF, McCullough LD, Urayama A. Cerebral Amyloid Angiopathy and Blood-Brain Barrier Dysfunction. Neuroscientist 2021; 27:668-684. [PMID: 33238806 PMCID: PMC9853919 DOI: 10.1177/1073858420954811] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cerebral hemorrhage, a devastating subtype of stroke, is often caused by hypertension and cerebral amyloid angiopathy (CAA). Pathological evidence of CAA is detected in approximately half of all individuals over the age of 70 and is associated with cortical microinfarcts and cognitive impairment. The underlying pathophysiology of CAA is characterized by accumulation of pathogenic amyloid β (Aβ) fragments of amyloid precursor protein in the cerebral vasculature. Vascular deposition of Aβ damages the vessel wall, results in blood-brain barrier (BBB) leakiness, vessel occlusion or rupture, and leads to hemorrhages and decreased cerebral blood flow that negatively affects vessel integrity and cognitive function. Currently, the main hypothesis surrounding the mechanism of CAA pathogenesis is that there is an impaired clearance of Aβ peptides, which includes compromised perivascular drainage as well as dysfunction of BBB transport. Also, the immune response in CAA pathogenesis plays an important role. Therefore, the mechanism by which Aβ vascular deposition occurs is crucial for our understanding of CAA pathogenesis and for the development of potential therapeutic options.
Collapse
Affiliation(s)
- Monica Gireud-Goss
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexis F. Mack
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
181
|
Liu X, Naomi SSM, Sharon WL, Russell EJ. The Applications of Focused Ultrasound (FUS) in Alzheimer's Disease Treatment: A Systematic Review on Both Animal and Human Studies. Aging Dis 2021; 12:1977-2002. [PMID: 34881081 PMCID: PMC8612615 DOI: 10.14336/ad.2021.0510] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) affects the basic ability to function and has imposed an immense burden on the community and health care system. Focused ultrasound (FUS) has recently been proposed as a novel noninvasive therapeutic approach for AD. However, systematic reviews on the FUS application in AD treatment have not been forthcoming. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria to summarize the techniques associated with safety and efficacy, as well as possible underlying mechanisms of FUS effects on AD in animal and human studies. Animal studies demonstrated FUS with microbubbles (FUS-MB) induced blood-brain-barrier (BBB) opening that could facilitate various therapeutic agents entering the brain. Repeated FUS-MB and FUS stimulation can relieve AD pathology and improve cognitive and memory function. Human studies showed repeated FUS-MB are well tolerated with few adverse events and FUS stimulation could enhance local perfusion and neural function, which correlated with cognitive improvement. We conclude that FUS is a feasible and safe therapeutic and drug delivery strategy for AD. However, FUS treatment on humans is still in the early stages and requires further optimization and standardization.
Collapse
Affiliation(s)
- Xiaodan Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - S. Sta Maria Naomi
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Wu Lin Sharon
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - E. Jacobs Russell
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
182
|
Falter A, Van Den Bossche MJA. How non-rapid eye movement sleep and Alzheimer pathology are linked. World J Psychiatry 2021; 11:1027-1038. [PMID: 34888171 PMCID: PMC8613758 DOI: 10.5498/wjp.v11.i11.1027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder characterized by the presence of senile plaques and neurofibrillary tangles. Research attempts to identify characteristic factors that are associated with the presence of the AD pathology on the one hand and that increase the risk of developing AD on the other. Changes in non-rapid eye movement (NREM) sleep may meet both requirements for various reasons. First, NREM-sleep is important for optimal memory function. In addition, studies report that the presence of AD pathology is associated with NREM-sleep changes. Finally, more and more results appear to suggest that sleep problems are not only a symptom of AD but can also increase the risk of AD. Several of these studies suggest that it is primarily a lack of NREM-sleep that is responsible for this increased risk. However, the majority investigated sleep only through subjective reporting, as a result of which NREM-sleep could not be analyzed separately. The aim of this literature study is therefore to present the results of the studies that relate the AD pathology and NREM-sleep (registered by electroencephalography). Furthermore, we try to evaluate whether NREM-sleep analysis could be used to support the diagnosis of AD and whether NREM-sleep deficiency could be a causal factor in the development of AD.
Collapse
Affiliation(s)
- Annelies Falter
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven 3000, Belgium
| | - Maarten J A Van Den Bossche
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven 3000, Belgium
- Center for Neuropsychiatry, Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
183
|
Zheng R, Huang YM, Zhou Q. Xueshuantong Improves Functions of Lymphatic Ducts and Modulates Inflammatory Responses in Alzheimer's Disease Mice. Front Pharmacol 2021; 12:605814. [PMID: 34650426 PMCID: PMC8505705 DOI: 10.3389/fphar.2021.605814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
Recent studies have revealed significant contributions of lymphatic vessels (LVs) to vital functions of the brain, especially related to clearance of waste from the brain and immune responses in the brain. These studies collectively indicate that enhancing the functions of LVs may improve brain functions during brain aging and in Alzheimer’s disease (AD) where LV functions are impaired. However, it is currently unknown whether this enhancement can be achieved using small molecules. We have previously shown that a widely used Chinese herbal medicine Xueshuantong (XST) significantly improves functions and reduces pathology in AD transgenic mice associated with elevated cerebral blood flow (CBF). Here, we show that XST partially rescues deficits in lymphatic structures, improves clearance of amyloid-β (Aβ) from the brain, and reduces the inflammatory responses in the serum and brains of transgenic AD mice. In addition, we showed that this improvement in the lymphatic system occurs independently of elevated CBF, suggesting independent modulation and limited interaction between blood circulation and lymphatic systems. Moreover, XST treatment leads to a significant increase in GLT-1 level and a significantly lower level of MMP-9 and restores AQP4 polarity in APP/PS1 mice. These results provide the basis for further exploration of XST to enhance or restore LV functions, which may be beneficial to treat neurodegenerative diseases or promote healthy aging.
Collapse
Affiliation(s)
- Rui Zheng
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yang-Mei Huang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
184
|
Gama Sosa MA, De Gasperi R, Pryor D, Perez Garcia GS, Perez GM, Abutarboush R, Kawoos U, Hogg S, Ache B, Janssen WG, Sowa A, Tetreault T, Cook DG, Tappan SJ, Gandy S, Hof PR, Ahlers ST, Elder GA. Low-level blast exposure induces chronic vascular remodeling, perivascular astrocytic degeneration and vascular-associated neuroinflammation. Acta Neuropathol Commun 2021; 9:167. [PMID: 34654480 PMCID: PMC8518227 DOI: 10.1186/s40478-021-01269-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023] Open
Abstract
Cerebral vascular injury as a consequence of blast-induced traumatic brain injury is primarily the result of blast wave-induced mechanical disruptions within the neurovascular unit. In rodent models of blast-induced traumatic brain injury, chronic vascular degenerative processes are associated with the development of an age-dependent post-traumatic stress disorder-like phenotype. To investigate the evolution of blast-induced chronic vascular degenerative changes, Long-Evans rats were blast-exposed (3 × 74.5 kPa) and their brains analyzed at different times post-exposure by X-ray microcomputed tomography, immunohistochemistry and electron microscopy. On microcomputed tomography scans, regional cerebral vascular attenuation or occlusion was observed as early as 48 h post-blast, and cerebral vascular disorganization was visible at 6 weeks and more accentuated at 13 months post-blast. Progression of the late-onset pathology was characterized by detachment of the endothelial and smooth muscle cellular elements from the neuropil due to degeneration and loss of arteriolar perivascular astrocytes. Development of this pathology was associated with vascular remodeling and neuroinflammation as increased levels of matrix metalloproteinases (MMP-2 and MMP-9), collagen type IV loss, and microglial activation were observed in the affected vasculature. Blast-induced chronic alterations within the neurovascular unit should affect cerebral blood circulation, glymphatic flow and intramural periarterial drainage, all of which may contribute to development of the blast-induced behavioral phenotype. Our results also identify astrocytic degeneration as a potential target for the development of therapies to treat blast-induced brain injury.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Georgina S Perez Garcia
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
| | - Gissel M Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, MD, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, MD, USA
| | - Seth Hogg
- Micro Photonics, Inc, 1550 Pond Road, Suite 110, Allentown, PA, 18104, USA
| | - Benjamin Ache
- Micro Photonics, Inc, 1550 Pond Road, Suite 110, Allentown, PA, 18104, USA
| | - William G Janssen
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Allison Sowa
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - David G Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Susan J Tappan
- MBF Bioscience LLC, 185 Allen Brook Lane, Williston, VT, 05495, USA
| | - Sam Gandy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Patrick R Hof
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Gregory A Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| |
Collapse
|
185
|
Hiraldo-González L, Trillo-Contreras JL, García-Miranda P, Pineda-Sánchez R, Ramírez-Lorca R, Rodrigo-Herrero S, Blanco MO, Oliver M, Bernal M, Franco-Macías E, Villadiego J, Echevarría M. Evaluation of aquaporins in the cerebrospinal fluid in patients with idiopathic normal pressure hydrocephalus. PLoS One 2021; 16:e0258165. [PMID: 34597351 PMCID: PMC8486078 DOI: 10.1371/journal.pone.0258165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022] Open
Abstract
Brain aquaporin 1 (AQP1) and AQP4 are involved in cerebrospinal fluid (CSF) homeostasis and might participate in the origin of hydrocephalus. Studies have shown alterations of perivascular AQP4 expression in idiopathic normal pressure hydrocephalus (iNPH) and Alzheimer's disease (AD). Due to the overlapping of clinical signs between iNPH and certain neurological conditions, mainly AD, specific biomarkers might improve the diagnostic accuracy for iNPH. The goal of the present study was to analyze and quantify the presence of AQP1 and AQP4 in the CSF of patients with iNPH and AD to determine whether these proteins can be used as biomarkers of iNPH. We examined AQP1 and AQP4 protein levels in the CSF of 179 participants (88 women) classified into 5 groups: possible iNPH (81 participants), hydrocephalus associated with other neurological disorders (13 participants), AD (41 participants), non-AD dementia (32 participants) and healthy controls (12 participants). We recorded each participant's demographic and clinical variables and indicated, when available in the clinical history, the record of cardiovascular and respiratory complications. An ELISA showed virtually no AQP content in the CSF. Information on the vascular risk factors (available for 61 patients) confirmed some type of vascular risk factor in 86% of the patients with possible iNPH and 58% of the patients with AD. In conclusion, the ELISA analysis showed insufficient sensitivity to detect the presence of AQP1 and AQP4 in CSF, ruling out the possible use of these proteins as biomarkers for diagnosing iNPH.
Collapse
Affiliation(s)
- Laura Hiraldo-González
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - José Luis Trillo-Contreras
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - Pablo García-Miranda
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - Rocío Pineda-Sánchez
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - Reposo Ramírez-Lorca
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Department of Physiology and Biophysics, University of Seville, Seville, Spain
| | - Silvia Rodrigo-Herrero
- Clinical Neuroscience Management Unit, Neurology Service, University Hospital Virgen del Rocío, Seville, Spain
| | - Magdalena Olivares Blanco
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Clinical Neuroscience Management Unit, Neurosurgery Service, University Hospital Virgen del Rocío, Seville, Spain
| | - María Oliver
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Clinical Neuroscience Management Unit, Neurosurgery Service, University Hospital Virgen del Rocío, Seville, Spain
| | - Maria Bernal
- Clinical Neuroscience Management Unit, Neurology Service, University Hospital Virgen del Rocío, Seville, Spain
| | - Emilio Franco-Macías
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Clinical Neuroscience Management Unit, Neurology Service, University Hospital Virgen del Rocío, Seville, Spain
| | - Javier Villadiego
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Department of Physiology and Biophysics, University of Seville, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Seville, Spain
| | - Miriam Echevarría
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Department of Physiology and Biophysics, University of Seville, Seville, Spain
- * E-mail:
| |
Collapse
|
186
|
Jiaerken Y, Lian C, Huang P, Yu X, Zhang R, Wang S, Hong H, Luo X, Yap PT, Shen D, Zhang M. Dilated perivascular space is related to reduced free-water in surrounding white matter among healthy adults and elderlies but not in patients with severe cerebral small vessel disease. J Cereb Blood Flow Metab 2021; 41:2561-2570. [PMID: 33818186 PMCID: PMC8504939 DOI: 10.1177/0271678x211005875] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Perivascular space facilitates cerebral interstitial water clearance. However, it is unclear how dilated perivascular space (dPVS) affects the interstitial water of surrounding white matter. We aimed to determine the presence and extent of changes in normal-appearing white matter water components around dPVS in different populations. Twenty healthy elderly subjects and 15 elderly subjects with severe cerebral small vessel disease (CSVD, with lacunar infarction 6 months before the scan) were included in our study. And other 28 healthy adult subjects were enrolled under a different scanning parameter to see if the results are comparable. The normal-appearing white matter around dPVS was categorized into 10 layers (1 mm thickness each) based on their distance to dPVS. We evaluated the mean isotropic-diffusing water volume fraction in each layer. We discovered a significantly reduced free-water content in the layers closely adjacent to the dPVS in the healthy elderlies. however, this reduction around dPVS was weaker in the CSVD subjects. We also discovered an elevated free-water content within dPVS. DPVS played different roles in healthy subjects or CSVD subjects. The reduced water content around dPVS in healthy subjects suggests these MR-visible PVSs are not always related to the stagnation of fluid.
Collapse
Affiliation(s)
- Yeerfan Jiaerken
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China.,Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chunfeng Lian
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peiyu Huang
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Xinfeng Yu
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Ruiting Zhang
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Shuyue Wang
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Hui Hong
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Xiao Luo
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Pew-Thian Yap
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dinggang Shen
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China.,Shanghai United Imaging Intelligence Co., Ltd, Shanghai, China.,Department of Artificial Intelligence, Korea University, Seoul, Republic of Korea
| | - Minming Zhang
- Department of Radiology, School of Medicine, Second Affiliated Hospital of Zhejiang University, Zhejiang, China
| |
Collapse
|
187
|
Mahan VL. Effects of lactate and carbon monoxide interactions on neuroprotection and neuropreservation. Med Gas Res 2021; 11:158-173. [PMID: 34213499 PMCID: PMC8374456 DOI: 10.4103/2045-9912.318862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/21/2020] [Accepted: 10/23/2020] [Indexed: 11/04/2022] Open
Abstract
Lactate, historically considered a waste product of anerobic metabolism, is a metabolite in whole-body metabolism needed for normal central nervous system (CNS) functions and a potent signaling molecule and hormone in the CNS. Neuronal activity signals normally induce its formation primarily in astrocytes and production is dependent on anerobic and aerobic metabolisms. Functions are dependent on normal dynamic, expansive, and evolving CNS functions. Levels can change under normal physiologic conditions and with CNS pathology. A readily combusted fuel that is sshuttled throughout the body, lactate is used as an energy source and is needed for CNS hemostasis, plasticity, memory, and excitability. Diffusion beyond the neuron active zone impacts activity of neurons and astrocytes in other areas of the brain. Barriergenesis, function of the blood-brain barrier, and buffering between oxidative metabolism and glycolysis and brain metabolism are affected by lactate. Important to neuroprotection, presence or absence is associated with L-lactate and heme oxygenase/carbon monoxide (a gasotransmitter) neuroprotective systems. Effects of carbon monoxide on L-lactate affect neuroprotection - interactions of the gasotransmitter with L-lactate are important to CNS stability, which will be reviewed in this article.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
188
|
Mijnders LSP, Steup FWR, Lindhout M, van der Kleij PA, Brink WM, van der Molen AJ. Optimal sequences and sequence parameters for GBCA-enhanced MRI of the glymphatic system: a systematic literature review. Acta Radiol 2021; 62:1324-1332. [PMID: 33153270 DOI: 10.1177/0284185120969950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The glymphatic system (GS) is a recently discovered waste clearance system in the brain. PURPOSE To evaluate the most promising magnetic resonance imaging (MRI) sequence(s) and the most optimal sequence parameters for glymphatic MRI (gMRI) 4-24 h after administration of gadolinium-based contrast agent (GBCA). MATERIAL AND METHODS Multiple literature databases were systematically searched for articles regarding gMRI or MRI of the perilymph in the inner ear until 11 May 2020. All relevant MRI sequence parameters were tabulated for qualitative analysis. Their potential was assessed based on detection of low dose GBCA, primarily measured as signal intensity (SI) ratio. RESULTS Thirty articles were included in the analysis. Three-dimensional fluid attenuated inversion recovery (3D-FLAIR), 3D Real Inversion Recovery (3D-Real IR), and multiple 3D T1-weighted gradient echo sequences were used. In perilymph, 3D-FLAIR with a TE of at least 400 ms yielded the highest SIRs. In the qualitative analysis of inner ear studies using 3D-FLAIR, TR was in the range of 4400-10,000 ms, TI 1500-2600 ms, refocusing flip angle (rFA) (range 120°-180°), and echo train length (ETL) 23-173. In the gMRI studies, quantitative analysis was not possible. In the qualitative analysis, 3D-FLAIR was used in the majority (8/12) of the studies, usually with TR 4800-9000 ms, TI 1650-2500 ms, TE 311-561 ms, rFA 90°-120°, and ETL 167-278. CONCLUSION Long TE 3D-FLAIR is the most promising sequence for detection of low-dose GBCA in the GS. Clinical and/or phantom studies on other MRI parameters are needed for further optimization of gMRI.
Collapse
Affiliation(s)
- Liesje SP Mijnders
- Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, The Netherlands
| | - Feline WR Steup
- Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, The Netherlands
| | - Mette Lindhout
- Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, The Netherlands
| | - Paul A van der Kleij
- Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, The Netherlands
| | - Wyger M Brink
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aart J van der Molen
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
189
|
Potential of Multiscale Astrocyte Imaging for Revealing Mechanisms Underlying Neurodevelopmental Disorders. Int J Mol Sci 2021; 22:ijms221910312. [PMID: 34638653 PMCID: PMC8508625 DOI: 10.3390/ijms221910312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/18/2023] Open
Abstract
Astrocytes provide trophic and metabolic support to neurons and modulate circuit formation during development. In addition, astrocytes help maintain neuronal homeostasis through neurovascular coupling, blood-brain barrier maintenance, clearance of metabolites and nonfunctional proteins via the glymphatic system, extracellular potassium buffering, and regulation of synaptic activity. Thus, astrocyte dysfunction may contribute to a myriad of neurological disorders. Indeed, astrocyte dysfunction during development has been implicated in Rett disease, Alexander's disease, epilepsy, and autism, among other disorders. Numerous disease model mice have been established to investigate these diseases, but important preclinical findings on etiology and pathophysiology have not translated into clinical interventions. A multidisciplinary approach is required to elucidate the mechanism of these diseases because astrocyte dysfunction can result in altered neuronal connectivity, morphology, and activity. Recent progress in neuroimaging techniques has enabled noninvasive investigations of brain structure and function at multiple spatiotemporal scales, and these technologies are expected to facilitate the translation of preclinical findings to clinical studies and ultimately to clinical trials. Here, we review recent progress on astrocyte contributions to neurodevelopmental and neuropsychiatric disorders revealed using novel imaging techniques, from microscopy scale to mesoscopic scale.
Collapse
|
190
|
Lee HJ, Lee DA, Shin KJ, Park KM. Glymphatic system dysfunction in patients with juvenile myoclonic epilepsy. J Neurol 2021; 269:2133-2139. [PMID: 34510256 DOI: 10.1007/s00415-021-10799-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The glymphatic system is a glial cell-dependent waste clearance pathway in the brain that is essential for the maintenance of brain homeostasis. In this study, we evaluated glymphatic system function in patients with juvenile myoclonic epilepsy (JME) compared with healthy controls. METHODS Patients with JME and healthy controls were retrospectively enrolled in this study. All the participants underwent brain diffusion tensor imaging (DTI). The "DTI-analysis along the perivascular space (ALPS)"-index was calculated to evaluate the glymphatic system function of the participants. The ALPS-indices of the patients with JME were compared with those of the healthy controls. In addition, the correlations between ALPS-index and the clinical characteristics of the patients with JME were analyzed to validate changes in glymphatic system function. RESULTS A total of 39 patients with JME and 38 healthy controls were enrolled in this study. The mean ALPS- index of the patients with JME was significantly lower than that of the healthy controls (1.541 vs. 1.653, p = 0.041). ALPS-index was negatively correlated with age in patients with JME (r = -0.375, p = 0.018). However, ALPS-index was not correlated with age at onset, duration of epilepsy, or anti-seizure medication load in patients with JME. CONCLUSION This study is the first in which the ALPS method was used to demonstrate that patients with JME have significant glymphatic system dysfunction. The results also show that glymphatic system index is negatively correlated with age in patients with JME, a finding which demonstrates that the glymphatic system function of patients with JME gradually declines with age. The ALPS-index might be a potential biomarker for monitoring glymphatic system function in patients with epilepsy.
Collapse
Affiliation(s)
- Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Republic of Korea
| | - Kyong Jin Shin
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Republic of Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Republic of Korea.
| |
Collapse
|
191
|
Huang P, Zhang R, Jiaerken Y, Wang S, Yu W, Hong H, Lian C, Li K, Zeng Q, Luo X, Yu X, Xu X, Wu X, Zhang M. Deep white matter hyperintensity is associated with the dilation of perivascular space. J Cereb Blood Flow Metab 2021; 41:2370-2380. [PMID: 33757317 PMCID: PMC8393291 DOI: 10.1177/0271678x211002279] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Understanding the pathophysiology of white matter hyperintensity (WMH) is necessary to reduce its harmfulness. Dilated perivascular space (PVS) had been found related to WMH. In the present study, we aimed to examine the topological connections between WMH and PVS, and to investigate whether increased interstitial fluid mediates the correlation between PVS and WMH volumes. One hundred and thirty-six healthy elder subjects were retrospectively included from a prospectively collected community cohort. Sub-millimeter T2 weighted and FLAIR images were acquired for assessing the association between PVS and WMH. Diffusion tensor imaging and free-water (FW) analytical methods were used to quantify white matter free water content, and to explore whether it mediates the PVS-WMH association. We found that most (89%) of the deep WMH lesions were spatially connected with PVS, exhibiting several interesting topological types. PVS and WMH volumes were also significantly correlated (r = 0.222, p < 0.001). FW mediated this association in the whole sample (β = 0.069, p = 0.037) and in subjects with relatively high WMH load (β = 0.118, p = 0.006). These findings suggest a tight association between PVS dilation and WMH formation, which might be linked by the impaired glymphatic drainage function and accumulated local interstitial fluid.
Collapse
Affiliation(s)
- Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiting Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yeerfan Jiaerken
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenke Yu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunfeng Lian
- Department of Radiology and BRIC, University of North Carolina, Chapel Hill, NC, USA
| | - Kaicheng Li
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinfeng Yu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopei Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
192
|
Zebhauser PT, Berthele A, Franz MS, Goldhardt O, Diehl-Schmid J, Priller J, Ortner M, Grimmer T. Age-Dependency of Total Tau in the Cerebrospinal Fluid Is Corrected by Amyloid-β 1-40: A Correlational Study in Healthy Adults. J Alzheimers Dis 2021; 83:155-162. [PMID: 34250938 DOI: 10.3233/jad-210286] [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: 11/15/2022]
Abstract
BACKGROUND Tau proteins are established biomarkers of neuroaxonal damage in a wide range of neurodegenerative conditions. Although measurement of total-Tau in the cerebrospinal fluid is widely used in research and clinical settings, the relationship between age and total-Tau in the cerebrospinal fluid is yet to be fully understood. While past studies reported a correlation between age and total-Tau in the cerebrospinal fluid of healthy adults, in clinical practice the same cut-off value is used independently of patient's age. OBJECTIVE To further explore the relationship between age and total-Tau and to disentangle neurodegenerative from drainage-dependent effects. METHODS We analyzed cerebrospinal fluid samples of 76 carefully selected cognitively healthy adults and included amyloid-β 1-40 as a potential marker of drainage from the brain's interstitial system. RESULTS We found a significant correlation of total-Tau and age, which was no longer present when correcting total-Tau for amyloid-β 1-40 concentrations. These findings were replicated under varied inclusion criteria. CONCLUSION Results call into question the association of age and total-Tau in the cerebrospinal fluid. Furthermore, they suggest diagnostic utility of amyloid-β 1-40 as a possible proxy for drainage-mechanisms into the cerebrospinal fluid when interpreting biomarker concentrations for neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul Theo Zebhauser
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Achim Berthele
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Marie-Sophie Franz
- Department of Anesthesiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Charité-Universitätsmedizin Berlin and DZNE, Berlin, Germany.,University of Edinburgh and UK Dementia Research Institute, Edinburgh, UK
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| |
Collapse
|
193
|
Mitchell D, Shireman J, Sierra Potchanant EA, Lara-Velazquez M, Dey M. Neuroinflammation in Autoimmune Disease and Primary Brain Tumors: The Quest for Striking the Right Balance. Front Cell Neurosci 2021; 15:716947. [PMID: 34483843 PMCID: PMC8414998 DOI: 10.3389/fncel.2021.716947] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
According to classical dogma, the central nervous system (CNS) is defined as an immune privileged space. The basis of this theory was rooted in an incomplete understanding of the CNS microenvironment, however, recent advances such as the identification of resident dendritic cells (DC) in the brain and the presence of CNS lymphatics have deepened our understanding of the neuro-immune axis and revolutionized the field of neuroimmunology. It is now understood that many pathological conditions induce an immune response in the CNS, and that in many ways, the CNS is an immunologically distinct organ. Hyperactivity of neuro-immune axis can lead to primary neuroinflammatory diseases such as multiple sclerosis and antibody-mediated encephalitis, whereas immunosuppressive mechanisms promote the development and survival of primary brain tumors. On the therapeutic front, attempts are being made to target CNS pathologies using various forms of immunotherapy. One of the most actively investigated areas of CNS immunotherapy is for the treatment of glioblastoma (GBM), the most common primary brain tumor in adults. In this review, we provide an up to date overview of the neuro-immune axis in steady state and discuss the mechanisms underlying neuroinflammation in autoimmune neuroinflammatory disease as well as in the development and progression of brain tumors. In addition, we detail the current understanding of the interactions that characterize the primary brain tumor microenvironment and the implications of the neuro-immune axis on the development of successful therapeutic strategies for the treatment of CNS malignancies.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jack Shireman
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | | | - Montserrat Lara-Velazquez
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mahua Dey
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
194
|
Che Mohd Nassir CMN, Damodaran T, Yusof SR, Norazit A, Chilla G, Huen I, K. N. BP, Mohamed Ibrahim N, Mustapha M. Aberrant Neurogliovascular Unit Dynamics in Cerebral Small Vessel Disease: A Rheological Clue to Vascular Parkinsonism. Pharmaceutics 2021; 13:1207. [PMID: 34452169 PMCID: PMC8398765 DOI: 10.3390/pharmaceutics13081207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/26/2022] Open
Abstract
The distinctive anatomical assemble and functionally discrete multicellular cerebrovasculature dynamics confer varying rheological and blood-brain barrier permeabilities to preserve the integrity of cerebral white matter and its neural microenvironment. This homeostasis intricately involves the glymphatic system that manages the flow of interstitial solutes, metabolic waste, and clearance through the venous circulation. As a physiologically integrated neurogliovascular unit (NGVU) serving a particularly vulnerable cerebral white matter (from hypoxia, metabolic insults, infection, and inflammation), a likely insidious process over a lifetime could inflict microenvironment damages that may lead to pathological conditions. Two such conditions, cerebral small vessel disease (CSVD) and vascular parkinsonism (VaP), with poorly understood pathomechanisms, are frequently linked to this brain-wide NGVU. VaP is widely regarded as an atypical parkinsonism, described by cardinal motor manifestations and the presence of cerebrovascular disease, particularly white matter hyperintensities (WMHs) in the basal ganglia and subcortical region. WMHs, in turn, are a recognised imaging spectrum of CSVD manifestations, and in relation to disrupted NGVU, also include enlarged perivascular spaces. Here, in this narrative review, we present and discuss on recent findings that argue for plausible clues between CSVD and VaP by focusing on aberrant multicellular dynamics of a unique integrated NGVU-a crossroad of the immune-vascular-nervous system-which may also extend fresher insights into the elusive interplay between cerebral microvasculature and neurodegeneration, and the potential therapeutic targets.
Collapse
Affiliation(s)
- Che Mohd Nasril Che Mohd Nassir
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Thenmoly Damodaran
- Centre for Drug Research, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia; (T.D.); (S.R.Y.)
| | - Siti R. Yusof
- Centre for Drug Research, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia; (T.D.); (S.R.Y.)
| | - Anwar Norazit
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Selangor, Malaysia;
| | - Geetha Chilla
- A*STAR Institute of Bioengineering and Bioimaging, Helios, 11 Biopolis Way, Singapore 138667, Singapore; (G.C.); (I.H.); (B.P.K.N.)
| | - Isaac Huen
- A*STAR Institute of Bioengineering and Bioimaging, Helios, 11 Biopolis Way, Singapore 138667, Singapore; (G.C.); (I.H.); (B.P.K.N.)
| | - Bhanu Prakash K. N.
- A*STAR Institute of Bioengineering and Bioimaging, Helios, 11 Biopolis Way, Singapore 138667, Singapore; (G.C.); (I.H.); (B.P.K.N.)
| | - Norlinah Mohamed Ibrahim
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Selangor, Malaysia;
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
- Hospital Universiti Sains Malaysia, Jalan Raja Perempuan Zainab II, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
195
|
Yamaguchi Y, Wada M, Kimihira L, Nagasawa H. Cognitive impairment due to widespread enlarged perivascular spaces. Radiol Case Rep 2021; 16:2640-2645. [PMID: 34345324 PMCID: PMC8319478 DOI: 10.1016/j.radcr.2021.06.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 11/28/2022] Open
Abstract
Perivascular spaces, also known as Virchow-Robin spaces, are usually considered as a normal, asymptomatic finding. However, this finding can occasionally demonstrate an atypical appearance and can be symptomatic. We report herein a rare case of cognitive impairment associated with extremely enlarged perivascular spaces. A 68-year-old Japanese woman visited our hospital with a 1-year history of progressive memory impairment. In addition to temporal disorientation and short-term memory impairment, neuropsychological testing showed frontal lobe-related symptoms such as slowed thinking processes, reduced verbal fluency, attention deficit, and reduced working memory. Magnetic resonance imaging of the brain showed widespread enlarged perivascular spaces almost symmetrically in the subcortical white matter of bilateral hemispheres, prominently in bilateral insulas, and frontal opercula. On 99mTc-ethyl cysteinate dimer single photon emission computed tomography, hypoperfusion was apparent in bilateral insulas and frontal opercula where enlarged periventricular spaces were prominent, whereas cerebral perfusion was preserved in areas where enlargement of perivascular spaces was mild or absent. Because symptoms were consistent with the distribution of the enlarged perivascular spaces and hypoperfusion in the brain, cognitive impairment due to enlarged perivascular spaces was diagnosed. Clinicians should note enlarged perivascular spaces as a potential cause of neurological deficits including cognitive impairment.
Collapse
Affiliation(s)
- Yoshitaka Yamaguchi
- Department of Neurology, Yamagata Prefectural Central Hospital, 1800 Aoyagi, Yamagata 990-2292, Japan
| | - Manabu Wada
- Department of Neurology, Yamagata Prefectural Central Hospital, 1800 Aoyagi, Yamagata 990-2292, Japan
| | - Luna Kimihira
- Department of Neurology, Yamagata Prefectural Central Hospital, 1800 Aoyagi, Yamagata 990-2292, Japan
| | - Hikaru Nagasawa
- Department of Neurology, Yamagata Prefectural Central Hospital, 1800 Aoyagi, Yamagata 990-2292, Japan
| |
Collapse
|
196
|
Thompson TB, Meisl G, Knowles TPJ, Goriely A. The role of clearance mechanisms in the kinetics of pathological protein aggregation involved in neurodegenerative diseases. J Chem Phys 2021; 154:125101. [PMID: 33810689 DOI: 10.1063/5.0031650] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The deposition of pathological protein aggregates in the brain plays a central role in cognitive decline and structural damage associated with neurodegenerative diseases. In Alzheimer's disease, the formation of amyloid-β plaques and neurofibrillary tangles of the tau protein is associated with the appearance of symptoms and pathology. Detailed models for the specific mechanisms of aggregate formation, such as nucleation and elongation, exist for aggregation in vitro where the total protein mass is conserved. However, in vivo, an additional class of mechanisms that clear pathological species is present and is believed to play an essential role in limiting the formation of aggregates and preventing or delaying the emergence of disease. A key unanswered question in the field of neuro-degeneration is how these clearance mechanisms can be modeled and how alterations in the processes of clearance or aggregation affect the stability of the system toward aggregation. Here, we generalize classical models of protein aggregation to take into account both production of monomers and the clearance of protein aggregates. We show that, depending on the specifics of the clearance process, a critical clearance value emerges above which accumulation of aggregates does not take place. Our results show that a sudden switch from a healthy to a disease state can be caused by small variations in the efficiency of the clearance process and provide a mathematical framework to explore the detailed effects of different mechanisms of clearance on the accumulation of aggregates.
Collapse
Affiliation(s)
- T B Thompson
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - G Meisl
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - T P J Knowles
- Department of Chemistry and Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - A Goriely
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
197
|
Mogensen FLH, Delle C, Nedergaard M. The Glymphatic System (En)during Inflammation. Int J Mol Sci 2021; 22:7491. [PMID: 34299111 PMCID: PMC8305763 DOI: 10.3390/ijms22147491] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 01/15/2023] Open
Abstract
The glymphatic system is a fluid-transport system that accesses all regions of the brain. It facilitates the exchange of cerebrospinal fluid and interstitial fluid and clears waste from the metabolically active brain. Astrocytic endfeet and their dense expression of the aquaporin-4 water channels promote fluid exchange between the perivascular spaces and the neuropil. Cerebrospinal and interstitial fluids are together transported back to the vascular compartment by meningeal and cervical lymphatic vessels. Multiple lines of work show that neurological diseases in general impair glymphatic fluid transport. Insofar as the glymphatic system plays a pseudo-lymphatic role in the central nervous system, it is poised to play a role in neuroinflammation. In this review, we discuss how the association of the glymphatic system with the meningeal lymphatic vessel calls for a renewal of established concepts on the CNS as an immune-privileged site. We also discuss potential approaches to target the glymphatic system to combat neuroinflammation.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (F.L.-H.M.); (C.D.)
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (F.L.-H.M.); (C.D.)
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (F.L.-H.M.); (C.D.)
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
198
|
Lv T, Zhao B, Hu Q, Zhang X. The Glymphatic System: A Novel Therapeutic Target for Stroke Treatment. Front Aging Neurosci 2021; 13:689098. [PMID: 34305569 PMCID: PMC8297504 DOI: 10.3389/fnagi.2021.689098] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
The glymphatic system (GS) is a novel defined brain-wide perivascular transit network between cerebrospinal fluid (CSF) and interstitial solutes that facilitates the clearance of brain metabolic wastes. The complicated network of the GS consists of the periarterial CSF influx pathway, astrocytes-mediated convective transport of fluid and solutes supported by AQP4 water channels, and perivenous efflux pathway. Recent researches indicate that the GS dysfunction is associated with various neurological disorders, including traumatic brain injury, hydrocephalus, epilepsy, migraine, and Alzheimer’s disease (AD). Meanwhile, the GS also plays a pivotal role in the pathophysiological process of stroke, including brain edema, blood–brain barrier (BBB) disruption, immune cell infiltration, neuroinflammation, and neuronal apoptosis. In this review, we illustrated the key anatomical structures of the GS, the relationship between the GS and the meningeal lymphatic system, the interaction between the GS and the BBB, and the crosstalk between astrocytes and other GS cellular components. In addition, we contributed to the current knowledge about the role of the GS in the pathology of stroke and the role of AQP4 in stroke. We further discussed the potential use of the GS in early risk assessment, diagnostics, prognostics, and therapeutics of stroke.
Collapse
Affiliation(s)
- Tao Lv
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Zhao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Hu
- Central Laboratory, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
199
|
Kaur J, Fahmy LM, Davoodi-Bojd E, Zhang L, Ding G, Hu J, Zhang Z, Chopp M, Jiang Q. Waste Clearance in the Brain. Front Neuroanat 2021; 15:665803. [PMID: 34305538 PMCID: PMC8292771 DOI: 10.3389/fnana.2021.665803] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
Waste clearance (WC) is an essential process for brain homeostasis, which is required for the proper and healthy functioning of all cerebrovascular and parenchymal brain cells. This review features our current understanding of brain WC, both within and external to the brain parenchyma. We describe the interplay of the blood-brain barrier (BBB), interstitial fluid (ISF), and perivascular spaces within the brain parenchyma for brain WC directly into the blood and/or cerebrospinal fluid (CSF). We also discuss the relevant role of the CSF and its exit routes in mediating WC. Recent discoveries of the glymphatic system and meningeal lymphatic vessels, and their relevance to brain WC are highlighted. Controversies related to brain WC research and potential future directions are presented.
Collapse
Affiliation(s)
- Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Lara M. Fahmy
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, United States
| | - Esmaeil Davoodi-Bojd
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Radiology, Henry Ford Health System, Detroit, MI, United States
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI, United States
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
200
|
Fatty Acid Binding Protein 3 (FABP3) and Apolipoprotein E4 (ApoE4) as Lipid Metabolism-Related Biomarkers of Alzheimer's Disease. J Clin Med 2021; 10:jcm10143009. [PMID: 34300173 PMCID: PMC8303862 DOI: 10.3390/jcm10143009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Lipid metabolism-related biomarkers gain increasing researchers interest in the field of neurodegenerative disorders. Mounting evidence have indicated the role of fatty acid-binding proteins and pathology lipid metabolism in Alzheimer’s Disease (AD). The imbalance of fatty acids (FA) and lipids may negatively affect brain functions related to neurodegenerative disorders. The ApoE4 and FABP3 proteins may reflect processes leading to neurodegeneration. This study aimed to evaluate the relationship between the CSF levels of FABP3 and ApoE4 proteins and cognitive decline as well as the diagnostic performance of these candidate biomarkers in AD and mild cognitive impairment (MCI). Methods: A total of 70 subjects, including patients with AD, MCI, and non-demented controls, were enrolled in the study. CSF concentrations of FABP3 and ApoE4 were measured using immunoassay technology. Results: Significantly higher CSF concentrations of FABP3 and ApoE4 were observed in AD patients compared to MCI subjects and individuals without cognitive impairment. Both proteins were inversely associated with Aβ42/40 ratio: ApoE4 (rho = −0.472, p < 0.001), and FABP3 (rho = −0.488, p < 0.001) in the whole study group, respectively. Additionally, FABP3 was negatively correlated with Mini-Mental State Examination score in the whole study cohort (rho = −0.585 p < 0.001). Conclusion: Presented results indicate the pivotal role of FABP3 and ApoE4 in AD pathology as lipid-related biomarkers, but studies on larger cohorts are needed.
Collapse
|