101
|
Kylkilahti TM, Berends E, Ramos M, Shanbhag NC, Töger J, Markenroth Bloch K, Lundgaard I. Achieving brain clearance and preventing neurodegenerative diseases-A glymphatic perspective. J Cereb Blood Flow Metab 2021; 41:2137-2149. [PMID: 33461408 PMCID: PMC8392766 DOI: 10.1177/0271678x20982388] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/28/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
Age-related neurodegenerative diseases are a growing burden to society, and many are sporadic, meaning that the environment, diet and lifestyle play significant roles. Cerebrospinal fluid (CSF)-mediated clearing of brain waste products via perivascular pathways, named the glymphatic system, is receiving increasing interest, as it offers unexplored perspectives on understanding neurodegenerative diseases. The glymphatic system is involved in clearance of metabolic by-products such as amyloid-β from the brain, and its function is believed to lower the risk of developing some of the most common neurodegenerative diseases. Here, we present magnetic resonance imaging (MRI) data on the heart cycle's control of CSF flow in humans which corroborates findings from animal studies. We also review the importance of sleep, diet, vascular health for glymphatic clearance and find that these factors are also known players in brain longevity.
Collapse
Affiliation(s)
- Tekla Maria Kylkilahti
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Eline Berends
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Marta Ramos
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Nagesh C Shanbhag
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Johannes Töger
- Diagnostic Radiology, Department of Clinical Sciences, Lund University and Skane University Hospital Lund, Lund, Sweden
| | | | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
102
|
Benveniste H, Nedergaard M. Cerebral small vessel disease: A glymphopathy? Curr Opin Neurobiol 2021; 72:15-21. [PMID: 34407477 DOI: 10.1016/j.conb.2021.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/23/2022]
Abstract
Small vessel disease (SVD) is a common instigator of dementia in the aging population. The hallmarks of SVD are enlargement of the perivascular spaces and white matter hyperintensities. The latter represents local fluid accumulation in white matter that either subsides or develops into lacunar infarcts. We here propose that failure of brain fluid transport-via the glymphatic system-plays a key role in initiation and progression of SVD. Our major case for this concept is that perivascular spaces are utilized as waterways for influx of cerebrospinal fluid. Stagnation of glymphatic transport may drive loss of brain fluid homeostasis leading to transient white matter edema, perivascular dilation, and ultimately demyelination. This review will discuss how glymphatic rodent studies of hypertension and diabetes have provided new insight into the pathogenesis of SVD.
Collapse
Affiliation(s)
- Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
103
|
Wafford KA. Aberrant waste disposal in neurodegeneration: why improved sleep could be the solution. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100025. [PMID: 36324713 PMCID: PMC9616228 DOI: 10.1016/j.cccb.2021.100025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 06/16/2023]
Abstract
Sleep takes up a large percentage of our lives and the full functions of this state are still not understood. However, over the last 10 years a new and important function has emerged as a mediator of brain clearance. Removal of toxic metabolites and proteins from the brain parenchyma generated during waking activity and high levels of synaptic processing is critical to normal brain function and only enabled during deep sleep. Understanding of this process is revealing how impaired sleep contributes an important and likely causative role in the accumulation and aggregation of aberrant proteins such as β-amyloid and phosphorylated tau, as well as inflammation and neuronal damage. We are also beginning to understand how brain slow-wave activity interacts with vascular function allowing the flow of CSF and interstitial fluid to drain into the body's lymphatic system. New methodology is enabling visualization of this process in both animals and humans and is revealing how these processes break down during ageing and disease. With this understanding we can begin to envisage novel therapeutic approaches to the treatment of neurodegeneration, and how reversing sleep impairment in the correct manner may provide a way to slow these processes and improve brain function.
Collapse
Key Words
- AQP4, aquaporin-4
- Alzheimer's disease
- Amyloid
- Aquaporin-4
- Astrocyte
- Aβ, beta amyloid
- BOLD, blood-oxygen level dependent imaging
- CAA, cerebral amyloid angiopathy
- CSF, Cerebrospinal fluid
- Clearance
- EEG, electroencephalography
- EMG, electromyography
- Glymphatic
- ISF, interstitial fluid
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- NOS, nitric oxide synthase
- NREM, non-rapid eye movement
- OSA, obstructive sleep apnea
- PET, positron emission tomography
- REM, rapid-eye movement
- SWA, slow wave activity
- SWS, slow-wave sleep
- Slow-wave sleep
- iNPH, idiopathic normal pressure hydrocephalus
Collapse
|
104
|
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
|
105
|
Ludwig HC, Dreha-Kulaczewski S, Bock HC. Neurofluids-Deep inspiration, cilia and preloading of the astrocytic network. J Neurosci Res 2021; 99:2804-2821. [PMID: 34323313 DOI: 10.1002/jnr.24935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/15/2021] [Accepted: 07/13/2021] [Indexed: 01/20/2023]
Abstract
With the advent of real-time MRI, the motion and passage of cerebrospinal fluid can be visualized without gating and exclusion of low-frequency waves. This imaging modality gives insights into low-volume, rapidly oscillating cardiac-driven movement as well as sustained, high-volume, slowly oscillating inspiration-driven movement. Inspiration means a spontaneous or artificial increase in the intrathoracic dimensions independent of body position. Alterations in thoracic diameter enable the thoracic and spinal epidural venous compartments to be emptied and filled, producing an upward surge of cerebrospinal fluid inside the spine during inspiration; this surge counterbalances the downward pooling of venous blood toward the heart. Real-time MRI, as a macroscale in vivo observation method, could expand our knowledge of neurofluid dynamics, including how astrocytic fluid preloading is adjusted and how brain buoyancy and turgor are maintained in different postures and zero gravity. Along with these macroscale findings, new microscale insights into aquaporin-mediated fluid transfer, its sensing by cilia, and its tuning by nitric oxide will be reviewed. By incorporating clinical knowledge spanning several disciplines, certain disorders-congenital hydrocephalus with Chiari malformation, idiopathic intracranial hypertension, and adult idiopathic hydrocephalus-are interpreted and reviewed according to current concepts, from the basics of the interrelated systems to their pathology.
Collapse
Affiliation(s)
- Hans C Ludwig
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Steffi Dreha-Kulaczewski
- Division of Pediatric Neurology, Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Hans C Bock
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
106
|
Lee KP, Chang AYW, Sung PS. Association between Blood Pressure, Blood Pressure Variability, and Post-Stroke Cognitive Impairment. Biomedicines 2021; 9:773. [PMID: 34356837 PMCID: PMC8301473 DOI: 10.3390/biomedicines9070773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
After stroke, dynamic changes take place from necrotic-apoptotic continuum, inflammatory response to poststroke neurogenesis, and remodeling of the network. These changes and baseline brain pathology such as small vessel disease (SVD) and amyloid burden may be associated with the occurrence of early or late poststroke cognitive impairment (PSCI) or dementia (PSD), which affect not only stroke victims but also their families and even society. We reviewed the current concepts and understanding of the pathophysiology for PSCI/PSD and identified useful tools for the diagnosis and the prediction of PSCI in serological, CSF, and image characteristics. Then, we untangled their relationships with blood pressure (BP) and blood pressure variability (BPV), important but often overlooked risk factors for PSCI/PSD. Finally, we provided evidence for the modifying effects of BP and BPV on PSCI as well as pharmacological and non-pharmacological interventions and life style modification for PSCI/PSD prevention and treatment.
Collapse
Affiliation(s)
- Kang-Po Lee
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Department of Neurology, E-DA Hospital, Kaohsiung 824, Taiwan
| | - Alice Y. W. Chang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Pi-Shan Sung
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| |
Collapse
|
107
|
Abstract
Cerebrospinal fluid flows around and into the brain, driven by intricate mechanisms, with profound implications for human health. According to the glymphatic hypothesis, in physiological conditions, cerebrospinal fluid flows primarily during sleep and serves to remove metabolic wastes like the amyloid-beta and tau proteins whose accumulation is believed to cause Alzheimer's disease. This paper reviews one research team's recent in vivo experiments and theoretical studies to better understand the fluid dynamics of brain cerebrospinal fluid flow. Driving mechanisms are considered, particularly arterial pulsation. Flow correlates closely with artery motion and changes when artery motion is manipulated. Though there are discrepancies between in vivo observations and predictions from simulations and theoretical studies of the mechanism, realistic boundary conditions bring closer agreement. Vessel shapes are considered, and have elongation that minimizes their hydraulic resistance, perhaps through evolutionary optimization. The pathological condition of stroke is considered. Much tissue damage after stroke is caused by swelling, and there is now strong evidence that early swelling is caused not by fluid from blood, as is commonly thought, but by cerebrospinal fluid. Finally, drug delivery is considered, and demonstrations show the glymphatic system could quickly deliver drugs across the blood-brain barrier. The paper closes with a discussion of future opportunities in the fast-changing field of brain fluid dynamics.
Collapse
Affiliation(s)
- Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, New York 14627, USA
| |
Collapse
|
108
|
Wang C, Wang X, Tan C, Wang Y, Tang Z, Zhang Z, Liu J, Xiao G. Novel therapeutics for hydrocephalus: Insights from animal models. CNS Neurosci Ther 2021; 27:1012-1022. [PMID: 34151523 PMCID: PMC8339528 DOI: 10.1111/cns.13695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/09/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Hydrocephalus is a cerebrospinal fluid physiological disorder that causes ventricular dilation with normal or high intracranial pressure. The current regular treatment for hydrocephalus is cerebrospinal fluid shunting, which is frequently related to failure and complications. Meanwhile, considering that the current nonsurgical treatments of hydrocephalus can only relieve the symptoms but cannot eliminate this complication caused by primary brain injuries, the exploration of more effective therapies has become the focus for many researchers. In this article, the current research status and progress of nonsurgical treatment in animal models of hydrocephalus are reviewed to provide new orientations for animal research and clinical practice.
Collapse
Affiliation(s)
- Chuansen Wang
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaoqiang Wang
- Department of Pediatric NeurosurgeryXinhua HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Changwu Tan
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yuchang Wang
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zhi Tang
- Department of NeurosurgeryHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Zhiping Zhang
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Jingping Liu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Gelei Xiao
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center for HydrocephalusXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
109
|
Glymphatic clearance function in patients with cerebral small vessel disease. Neuroimage 2021; 238:118257. [PMID: 34118396 DOI: 10.1016/j.neuroimage.2021.118257] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 02/08/2023] Open
Abstract
Few studies have focused on the connection between glymphatic dysfunction and cerebral small vessel disease (CSVD), partially due to the lack of non-invasive methods to measure glymphatic function. We established modified index for diffusion tensor image analysis along the perivascular space (mALPS-index), which was calculated on diffusion tensor image (DTI), compared it with the classical detection of glymphatic clearance function calculated on Glymphatic MRI after intrathecal administration of gadolinium (study 1), and analyzed the relationship between CSVD imaging markers and mALPS-index in CSVD patients from the CIRCLE study (ClinicalTrials.gov ID: NCT03542734) (study 2). Among 39 patients included in study 1, mALPS-index were significantly related to glymphatic clearance function calculated on Glymphatic MRI ( r = -0.772~-0.844, p < 0.001). A total of 330 CSVD patients were included in study 2. Severer periventricular and deep white matter hyperintensities (β = -0.332, p < 0.001; β = -0.293, p < 0.001), number of lacunas (β = -0.215, p < 0.001), number of microbleeds (β = -0.152, p = 0.005), and severer enlarged perivascular spaces in basal ganglia (β = -0.223, p < 0.001) were related to mALPS-index. Our results indicated that non-invasive mALPS-index might represent glymphatic clearance function, which could be applied in clinic in future. Glymphatic clearance function might play a role in the development of CSVD.
Collapse
|
110
|
Ren X, Liu S, Lian C, Li H, Li K, Li L, Zhao G. Dysfunction of the Glymphatic System as a Potential Mechanism of Perioperative Neurocognitive Disorders. Front Aging Neurosci 2021; 13:659457. [PMID: 34163349 PMCID: PMC8215113 DOI: 10.3389/fnagi.2021.659457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
Perioperative neurocognitive disorder (PND) frequently occurs in the elderly as a severe postoperative complication and is characterized by a decline in cognitive function that impairs memory, attention, and other cognitive domains. Currently, the exact pathogenic mechanism of PND is multifaceted and remains unclear. The glymphatic system is a newly discovered glial-dependent perivascular network that subserves a pseudo-lymphatic function in the brain. Recent studies have highlighted the significant role of the glymphatic system in the removal of harmful metabolites in the brain. Dysfunction of the glymphatic system can reduce metabolic waste removal, leading to neuroinflammation and neurological disorders. We speculate that there is a causal relationship between the glymphatic system and symptomatic progression in PND. This paper reviews the current literature on the glymphatic system and some perioperative factors to discuss the role of the glymphatic system in PND.
Collapse
Affiliation(s)
- Xuli Ren
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shan Liu
- Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Chuang Lian
- Department of Anaesthesiology, Jilin City People's Hospital, Jilin, China
| | - Haixia Li
- Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Kai Li
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Longyun Li
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Guoqing Zhao
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China.,Jilin University, Changchun, China
| |
Collapse
|
111
|
Liu X, Xie Y, Wan X, Wu J, Fan Z, Yang L. Protective Effects of Aquaporin-4 Deficiency on Longer-term Neurological Outcomes in a Mouse Model. Neurochem Res 2021; 46:1380-1389. [PMID: 33651262 DOI: 10.1007/s11064-021-03272-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Traumatic brain injury (TBI) has been a crucial health problem, with more than 50 million patients worldwide each year. Glymphatic system is a fluid exchange system that relies on the polarized water channel aquaporin-4 (AQP4) at the astrocytes, accounting for the clearance of abnormal proteins and metabolites from brain tissues. However, the dysfunction of glymphatic system and alteration of AQP4 polarization during the progression of TBI remain unclear. AQP4-/- and Wild Type (WT) mice were used to establish the TBI mouse model respectively. Brain edema and Evans blue extravasation were conducted 24 h post-injury to evaluate the acute TBI. Morris water maze (MWM) was used to establish the long-term cognitive functions of AQP4-/- and WT mice post TBI. Western-blot and qRT-PCR assays were performed to demonstrate protective effects of AQP4 deficiency to blood-brain barrier (BBB) integrity and amyloid-β clearance. The inflammation of cerebral tissues post TBI was estimated by ELISA assay. AQP4 deficiency alleviated the brain edema and neurological deficit in TBI mice. AQP4-knockout led to improved cognitive outcomes in mice post TBI. The BBB integrity and cerebral amyloid-β clearance were protected by AQP4 deficiency in TBI mice. AQP4 deficiency ameliorated the TBI-induced inflammation. AQP4 deficiency improved longer-term neurological outcomes in a mouse model of TBI.
Collapse
Affiliation(s)
- Xiaosong Liu
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Yingxin Xie
- Department of Doppler Ultrasound, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Xiangdong Wan
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Jianliang Wu
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Zhenzeng Fan
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China
| | - Lijun Yang
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215, Heping Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
112
|
Abstract
The glymphatic system hypothesis is associated with the circulation of cerebrospinal fluid (CSF) in the skull and interstitial fluid (ISF) in the brain. There are several imaging techniques to visualize the dynamics of CSF and ISF. Magnetic resonance imaging (MRI) is one of the promising modalities for glymphatic imaging and diffusion MRI is expected imaging tool. Several disorders are associated with glymphatic dysfunction or impairment in the dynamics of CSF or ISF. The Central Nervous System interstitial fluidopathy concept has been proposed to encompass diseases with pathologies that are predominantly associated with abnormal ISF/CSF dynamics.
Collapse
|
113
|
Gottlieb E, Khlif MS, Bird L, Werden E, Churchward T, Pase MP, Egorova N, Howard ME, Brodtmann A. Sleep architectural dysfunction and undiagnosed obstructive sleep apnea after chronic ischemic stroke. Sleep Med 2021; 83:45-53. [PMID: 33991892 DOI: 10.1016/j.sleep.2021.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE/BACKGROUND Sleep-wake dysfunction is bidirectionally associated with the incidence and evolution of acute stroke. It remains unclear whether sleep disturbances are transient post-stroke or are potentially enduring sequelae in chronic stroke. Here, we characterize sleep architectural dysfunction, sleep-respiratory parameters, and hemispheric sleep in ischemic stroke patients in the chronic recovery phase compared to healthy controls. PATIENTS/METHODS Radiologically confirmed ischemic stroke patients (n = 28) and matched control participants (n = 16) were tested with ambulatory polysomnography, bi-hemispheric sleep EEG, and demographic, stroke-severity, mood, and sleep-circadian questionnaires. RESULTS Twenty-eight stroke patients (22 men; mean age = 69.61 ± 7.4 years) were cross-sectionally evaluated 4.1 ± 0.9 years after mild-moderate ischemic stroke (baseline NIHSS: 3.0 ± 2.0). Fifty-seven percent of stroke patients (n = 16) exhibited undiagnosed moderate-to-severe obstructive sleep apnea (apnea-hypopnea index >15). Despite no difference in total sleep or wake after sleep onset, stroke patients had reduced slow-wave sleep time (66.25 min vs 99.26 min, p = 0.02), increased time in non-rapid-eye-movement (NREM) stages 1-2 (NREM-1: 48.43 vs 28.95, p = 0.03; NREM-2: 142.61 vs 115.87, p = 0.02), and a higher arousal index (21.46 vs 14.43, p = 0.03) when compared to controls. Controlling for sleep apnea severity did not attenuate the magnitude of sleep architectural differences between groups (NREM 1-3=ηp2 >0.07). We observed no differences in ipsilesionally versus contralesionally scored sleep architecture. CONCLUSIONS Fifty-seven percent of chronic stroke patients had undiagnosed moderate-severe obstructive sleep apnea and reduced slow-wave sleep with potentially compensatory increases in NREM 1-2 sleep relative to controls. Formal sleep studies are warranted after stroke, even in the absence of self-reported history of sleep-wake pathology.
Collapse
Affiliation(s)
- Elie Gottlieb
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia.
| | - Mohamed S Khlif
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Laura Bird
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Thomas Churchward
- Institute for Breathing and Sleep, Melbourne, VIC, Australia; Austin Health, Heidelberg, VIC, Australia
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, VIC, Australia; Harvard T.H. Chan School of Public Health, Harvard University, MA, USA
| | - Natalia Egorova
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia; Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Mark E Howard
- University of Melbourne, Melbourne, VIC, Australia; Institute for Breathing and Sleep, Melbourne, VIC, Australia; Austin Health, Heidelberg, VIC, Australia
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
114
|
Raghunandan A, Ladron-de-Guevara A, Tithof J, Mestre H, Du T, Nedergaard M, Thomas JH, Kelley DH. Bulk flow of cerebrospinal fluid observed in periarterial spaces is not an artifact of injection. eLife 2021; 10:65958. [PMID: 33687330 PMCID: PMC7979157 DOI: 10.7554/elife.65958] [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: 12/20/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Cerebrospinal fluid (CSF) flowing through periarterial spaces is integral to the brain’s mechanism for clearing metabolic waste products. Experiments that track tracer particles injected into the cisterna magna (CM) of mouse brains have shown evidence of pulsatile CSF flow in perivascular spaces surrounding pial arteries, with a bulk flow in the same direction as blood flow. However, the driving mechanism remains elusive. Several studies have suggested that the bulk flow might be an artifact, driven by the injection itself. Here, we address this hypothesis with new in vivo experiments where tracer particles are injected into the CM using a dual-syringe system, with simultaneous injection and withdrawal of equal amounts of fluid. This method produces no net increase in CSF volume and no significant increase in intracranial pressure. Yet, particle-tracking reveals flows that are consistent in all respects with the flows observed in earlier experiments with single-syringe injection.
Collapse
Affiliation(s)
- Aditya Raghunandan
- Department of Mechanical Engineering, University of Rochester, Rochester, United States
| | - Antonio Ladron-de-Guevara
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, United States
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Rochester, Rochester, United States.,Department of Mechanical Engineering, University of Minnesota, Minneapolis, United States
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, United States
| | - Ting Du
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, United States
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, United States.,Center for Translational Neuromedicine, University of Copenhagen, Rochester, United States
| | - John H Thomas
- Department of Mechanical Engineering, University of Rochester, Rochester, United States
| | - Douglas H Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, United States
| |
Collapse
|
115
|
Primary empty sella: The risk factors and associations with the cerebral small vessel diseases-An observational study. Clin Neurol Neurosurg 2021; 203:106586. [PMID: 33730618 DOI: 10.1016/j.clineuro.2021.106586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/11/2021] [Accepted: 02/27/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the risk factors of primary empty sella (PES) and its associations with cerebral small vessel diseases (CSVD). METHODS A total of 132 consecutive patients were recruited from Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University from December 2018 to January 2020, including 69 cases of PES, and age, gender-matched 63 subjects without PES. Demographics and clinical characteristics were recorded. Enlarged perivascular spaces (PVS) and white matter hyperintensities (WMH), which are image markers for CSVD, were assessed. Univariate logistic regression models and multivariate logistic regression models were performed to predict the independent risk factors of PES. RESULTS There was a significant difference in baseline characteristics in terms of hypertension (p < 0.001) and pregnancy (p = 0.019) between PES and the control group; among markers of CSVD, whole WMH (p = 0.030) and periventricular hyperintensities (PVH) (p = 0.027) were significantly different; however, no significant differences concerning deep WMH, total PVS, basilar ganglia-PVS and centrum semiovale-PVS (p > 0.05). After adjusting relevant potential confounders, multivariate logistic regression revealed hypertension (OR=3.158, 95 %CI: 1.452∼6.865, p = 0.004) and pregnancy (OR=2.236, 95 %CI: 1.036-4.826, p = 0.040) were independent risk factors for PES. CONCLUSION Hypertension and pregnancy are independent risk factors of PES. There is a possible correlation between PES and WMH, especially PVH, however, further studies are required to confirm these findings.
Collapse
|
116
|
Schellhorn T, Aamodt EB, Lydersen S, Aam S, Wyller TB, Saltvedt I, Beyer MK. Clinically accessible neuroimaging predictors of post-stroke neurocognitive disorder: a prospective observational study. BMC Neurol 2021; 21:89. [PMID: 33632149 PMCID: PMC7905565 DOI: 10.1186/s12883-021-02117-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/19/2021] [Indexed: 01/21/2023] Open
Abstract
Background Neurocognitive disorder (NCD) is common in stroke survivors. We aimed to identify clinically accessible imaging markers of stroke and chronic pathology that are associated with early post-stroke NCD. Methods We included 231 stroke survivors from the “Norwegian Cognitive Impairment after Stroke (Nor-COAST)” study who underwent a standardized cognitive assessment 3 months after the stroke. Any NCD (mild cognitive impairment and dementia) and major NCD (dementia) were diagnosed according to “Diagnostic and Statistical Manual of Mental Disorders (DSM-5)” criteria. Clinically accessible imaging findings were analyzed on study-specific brain MRIs in the early phase after stroke. Stroke lesion volumes were semi automatically quantified and strategic stroke locations were determined by an atlas based coregistration. White matter hyperintensities (WMH) and medial temporal lobe atrophy (MTA) were visually scored. Logistic regression was used to identify neuroimaging findings associated with major NCD and any NCD. Results Mean age was 71.8 years (SD 11.1), 101 (43.7%) were females, mean time from stroke to imaging was 8 (SD 16) days. At 3 months 63 (27.3%) had mild NCD and 65 (28.1%) had major NCD. Any NCD was significantly associated with WMH pathology (odds ratio (OR) = 2.73 [1.56 to 4.77], p = 0.001), MTA pathology (OR = 1.95 [1.12 to 3.41], p = 0.019), and left hemispheric stroke (OR = 1.8 [1.05 to 3.09], p = 0.032). Major NCD was significantly associated with WMH pathology (OR = 2.54 [1.33 to 4.84], p = 0.005) and stroke lesion volume (OR (per ml) =1.04 [1.01 to 1.06], p = 0.001). Conclusion WMH pathology, MTA pathology and left hemispheric stroke were associated with the development of any NCD. Stroke lesion volume and WMH pathology were associated with the development of major NCD 3 months after stroke. These imaging findings may be used in the routine clinical setting to identify patients at risk for early post-stroke NCD. Trial registration ClinicalTrials.gov, NCT02650531, Registered 8 January 2016 – Retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02117-8.
Collapse
Affiliation(s)
- Till Schellhorn
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Eva Birgitte Aamodt
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stian Lydersen
- Regional Centre for Child and Youth Mental Health and Child Welfare, Department of Mental Health, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Stina Aam
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Geriatric Medicine, Clinic of Medicine St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Torgeir Bruun Wyller
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Geriatric Medicine, Clinic of Medicine St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Mona Kristiansen Beyer
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
117
|
Yankova G, Bogomyakova O, Tulupov A. The glymphatic system and meningeal lymphatics of the brain: new understanding of brain clearance. Rev Neurosci 2021; 32:693-705. [PMID: 33618444 DOI: 10.1515/revneuro-2020-0106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/31/2021] [Indexed: 12/25/2022]
Abstract
The glymphatic system and meningeal lymphatics have recently been characterized. Glymphatic system is a glia-dependent system of perivascular channels, and it plays an important role in the removal of interstitial metabolic waste products. The meningeal lymphatics may be a key drainage route for cerebrospinal fluid into the peripheral blood, may contribute to inflammatory reaction and central nervous system (CNS) immune surveillance. Breakdowns and dysfunction of the glymphatic system and meningeal lymphatics play a crucial role in age-related brain changes, the pathogenesis of neurovascular and neurodegenerative diseases, as well as in brain injuries and tumors. This review discusses the relationship recently characterized meningeal lymphatic vessels with the glymphatic system, which provides perfusion of the CNS with cerebrospinal and interstitial fluids. The review also presents the results of human studies concerning both the presence of meningeal lymphatics and the glymphatic system. A new understanding of how aging, medications, sleep and wake cycles, genetic predisposition, and even body posture affect the brain drainage system has not only changed the idea of brain fluid circulation but has also contributed to an understanding of the pathology and mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Galina Yankova
- Lavrentyev Institute of Hydrodynamics, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia.,Novosibirsk State University, Novosibirsk630090,Russia
| | - Olga Bogomyakova
- International Tomography Center, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia
| | - Andrey Tulupov
- Novosibirsk State University, Novosibirsk630090,Russia.,International Tomography Center, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia.,Meshalkin National Medical Research Center, Ministry of Health of Russian Federation, Novosibirsk 630055, Russia
| |
Collapse
|
118
|
Manouchehrian O, Ramos M, Bachiller S, Lundgaard I, Deierborg T. Acute systemic LPS-exposure impairs perivascular CSF distribution in mice. J Neuroinflammation 2021; 18:34. [PMID: 33514389 PMCID: PMC7844902 DOI: 10.1186/s12974-021-02082-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background The exchange of cerebrospinal (CSF) and interstitial fluid is believed to be vital for waste clearance in the brain. The sleep-dependent glymphatic system, which is comprised of perivascular flow of CSF and is largely dependent on arterial pulsatility and astrocytic aquaporin-4 (AQP4) expression, facilitates much of this brain clearance. During the last decade, several observations have indicated that impaired glymphatic function goes hand in hand with neurodegenerative diseases. Since pathologies of the brain carry inflammatory components, we wanted to know how acute inflammation, e.g., with lipopolysaccharide (LPS) injections, would affect the glymphatic system. In this study, we aim to measure the effect of LPS on perivascular CSF distribution as a measure of glymphatic function. Methods Three hours after injection of LPS (1 mg/kg i.p.), C57bl/6 mice were (1) imaged for two CSF tracers, injected into cisterna magna, (2) transcardially perfused with buffer, or (3) used for physiological readouts. Tracer flow was imaged using a low magnification microscope on fixed brains, as well as using vibratome-cut slices for measuring tracer penetration in the brain. Cytokines, glial, and BBB-permeability markers were measured with ELISAs, Western blots, and immunohistochemistry. Cerebral blood flow was approximated using laser Doppler flowmetry, respiration and heart rate with a surgical monitor, and AQP4-polarization was quantified using confocal microscopy of immunolabeled brain sections. Results LPS-injections significantly lowered perivascular CSF tracer flow and penetration into the parenchyma. No differences in AQP4 polarization, cytokines, astroglial and BBB markers, cerebral blood flow, or respiration were detected in LPS-injected mice, although LPS did elevate cortical Iba1+ area and heart rate. Conclusions This study reports another physiological response after acute exposure to the bacterial endotoxin LPS, namely the statistically significant decrease in perivascular distribution of CSF. These observations may benefit our understanding of the role of systemic inflammation in brain clearance.
Collapse
Affiliation(s)
- Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.
| | - Marta Ramos
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| |
Collapse
|
119
|
Kambara A, Kajimoto Y, Yagi R, Ikeda N, Furuse M, Nonoguchi N, Kawabata S, Kuroiwa T, Kuroda K, Tsuji S, Saura R, Wanibuchi M. Long-Term Prognosis of Cognitive Function in Patients With Idiopathic Normal Pressure Hydrocephalus After Shunt Surgery. Front Aging Neurosci 2021; 12:617150. [PMID: 33551791 PMCID: PMC7854532 DOI: 10.3389/fnagi.2020.617150] [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: 10/14/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022] Open
Abstract
The long-term prognosis of cognitive function in patients with idiopathic normal pressure hydrocephalus (iNPH) remains unclear. This study aimed to determine the long-term prognosis of cognitive function in patients with iNPH, as well as the factors related to it. It included 48 patients with iNPH who were treated with cerebrospinal fluid shunting between January 2015 and December 2017 at Osaka Medical College Hospital, with follow-up evaluation of their cognitive function for >2 years. Cognitive function was measured using the Mini-Mental State Examination (MMSE) preoperatively and at 3 months, 1 and 2 years post-operatively. The mean MMSE score (22.4 ± 5.4 preoperatively) improved at 3 months [23.8 ± 5.0 (p = 0.0002)] and 1 year [23.7 ± 4.8 (p = 0.004)] post-operatively. At 2 years post-operatively, they were able to maintain their preoperative level (22.6 ± 5.3). The patients were classified in to the cognitive decline group [11 (23%) patients; a decrease in the MMSE score by ≥ 2 points 2 years after surgery] and the maintenance/improvement group [37 (77%) patients]. Univariate and receiver operating characteristic analyses were performed for the two groups to identify factors associated with cognitive prognosis. In both groups, the patients who were younger (p = 0.009) or had milder symptoms (p = 0.035) had a better long-term prognosis of cognitive function. The cutoffs for age and disease severity (idiopathic normal-pressure hydrocephalus grading scale; INPHGS) were 78 years (area under the curve = 0.77) and 5 points (area under the curve = 0.71), respectively. In conclusion, most patients (77%) were able to improve and maintain cognitive function for at least 2 years after surgery. The fact that disease severity and age are associated with cognitive prognosis suggests that early iNPH intervention is desirable to improve cognitive prognosis.
Collapse
Affiliation(s)
- Akihiro Kambara
- Department of Neurosurgery, Osaka Medical College, Takatsuki, Japan
| | | | - Ryokichi Yagi
- Department of Neurosurgery, Osaka Medical College, Takatsuki, Japan
| | - Naokado Ikeda
- Department of Neurosurgery, Osaka Medical College, Takatsuki, Japan
| | - Motomasa Furuse
- Department of Neurosurgery, Osaka Medical College, Takatsuki, Japan
| | | | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical College, Takatsuki, Japan
| | - Toshihiko Kuroiwa
- Department of Neurosurgery, Tesseikai Neurosurgery Hospital, Shijonawate, Japan
| | - Kenji Kuroda
- Clinical Department of Rehabilitation, Osaka Medical College, Takatsuki, Japan
| | - Shohei Tsuji
- Clinical Department of Rehabilitation, Osaka Medical College, Takatsuki, Japan
| | - Ryuichi Saura
- Department of Physical and Rehabilitation Medicine, Osaka Medical College, Takatsuki, Japan
| | | |
Collapse
|
120
|
Silva I, Silva J, Ferreira R, Trigo D. Glymphatic system, AQP4, and their implications in Alzheimer's disease. Neurol Res Pract 2021; 3:5. [PMID: 33499944 PMCID: PMC7816372 DOI: 10.1186/s42466-021-00102-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Lacking conventional lymphatic system, the central nervous system requires alternative clearance systems, such as the glymphatic system, which promotes clearance of interstitial solutes. Aquaporin-4 water channels (AQP4) are an integral part of this system and related to neuropathologies, such as Alzheimer's disease (AD). The clearance of Alzheimer's associated proteins amyloid β and tau is diminished by glymphatic system impairment, due to lack of AQP4. Even though AQP4 mislocalisation (which affects its activity) is a phenotype of AD, it remains a controversial topic, as it is still unclear if it is a phenotype-promoting factor or a consequence of this pathology. This review provides important and updated knowledge about glymphatic system, AQP4 itself, and their link with Alzheimer's disease. Finally, AQP4 as a therapeutic target is proposed to ameliorate Alzheimer's Disease and other neuropathologies AQP4-related.
Collapse
Affiliation(s)
- Inês Silva
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jéssica Silva
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rita Ferreira
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal.
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
121
|
Stanton EH, Persson NDÅ, Gomolka RS, Lilius T, Sigurðsson B, Lee H, Xavier ALR, Benveniste H, Nedergaard M, Mori Y. Mapping of CSF transport using high spatiotemporal resolution dynamic contrast-enhanced MRI in mice: Effect of anesthesia. Magn Reson Med 2021; 85:3326-3342. [PMID: 33426699 DOI: 10.1002/mrm.28645] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Dynamic contrast-enhanced MRI (DCE-MRI) represents the only available approach for glymphatic cerebrospinal fluid (CSF) flow 3D mapping in the brain of living animals and humans. The purpose of this study was to develop a novel DCE-MRI protocol for mapping of the glymphatic system transport with improved spatiotemporal resolution, and to validate the new protocol by comparing the transport in mice anesthetized with either isoflurane or ketamine/xylazine. METHODS The contrast agent, gadobutrol, was administered into the CSF of the cisterna magna and its transport visualized continuously on a 9.4T preclinical scanner using 3D fast-imaging with a steady-state free-precession sequence (3D-FISP), which has a spatial resolution of 0.001 mm3 and a temporal resolution of 30 s. The MR signals were measured dynamically for 60 min in multiple volumes of interest covering the entire CSF space and brain parenchyma. RESULTS The results confirm earlier findings that glymphatic CSF influx is higher under ketamine/xylazine than with isoflurane anesthesia. This was extended to account for new details about the distinct CSF efflux pathways under the two anesthetic regimens. Dynamic contrast MR shows that CSF clearance occurs mainly along the vagus nerve near the jugular vein under isoflurane and via the olfactory bulb under ketamine/xylazine. CONCLUSION The improved spatial and temporal sampling rates afforded by 3D-FISP shed new light on the pharmacological modulation of CSF efflux paths. The present observations may have the potential to set a new standard for future experimental DCE-MRI studies of the glymphatic system.
Collapse
Affiliation(s)
- Evan Hunter Stanton
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas Daniel Åke Persson
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ryszard Stefan Gomolka
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas Lilius
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Björn Sigurðsson
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anna Lenice Ribeiro Xavier
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
122
|
Ozturk BO, Monte B, Koundal S, Dai F, Benveniste H, Lee H. Disparate volumetric fluid shifts across cerebral tissue compartments with two different anesthetics. Fluids Barriers CNS 2021; 18:1. [PMID: 33407650 PMCID: PMC7788828 DOI: 10.1186/s12987-020-00236-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
Background Large differences in glymphatic system transport—similar in magnitude to those of the sleep/wake cycle—have been observed during anesthesia with dexmedetomidine supplemented with low dose isoflurane (DEXM-I) in comparison to isoflurane (ISO). However, the biophysical and bioenergetic tissue status underlying glymphatic transport differences between anesthetics remains undefined. To further understand biophysical characteristics underlying these differences we investigated volume status across cerebral tissue compartments, water diffusivity, and T2* values in rats anesthetized with DEXM-I in comparison to ISO. Methods Using a crossover study design, a group of 12 Sprague Dawley female rats underwent repetitive magnetic resonance imaging (MRI) under ISO and DEXM-I. Physiological parameters were continuously measured. MRI included a proton density weighted (PDW) scan to investigate cerebrospinal fluid (CSF) and parenchymal volumetric changes, a multigradient echo scan (MGE) to calculate T2* maps as a measure of ‘bioenergetics’, and a diffusion scan to quantify the apparent diffusion coefficient (ADC). Results The heart rate was lower with DEXM-I in comparison to ISO, but all other physiological variables were similar across scans and groups. The PDW images revealed a 1% parenchymal volume increase with ISO compared to DEXM-I comprising multiple focal tissue areas scattered across the forebrain. In contrast, with DEXM-I the CSF compartment was enlarged by ~ 6% in comparison to ISO at the level of the basal cisterns and peri-arterial conduits which are main CSF influx routes for glymphatic transport. The T2* maps showed brain-wide increases in T2* in ISO compared to DEXM-I rats. Diffusion-weighted images yielded no significant differences in ADCs across the two anesthesia groups. Conclusions We demonstrated CSF volume expansion with DEXM-I (in comparison to ISO) and parenchymal (GM) expansion with ISO (in comparison to DEXM-I), which may explain the differences in glymphatic transport. The T2* changes in ISO are suggestive of an increased bioenergetic state associated with excess cellular firing/bursting when compared to DEXM-I.
Collapse
Affiliation(s)
- Burhan O Ozturk
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT, USA
| | - Brittany Monte
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT, USA
| | - Feng Dai
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT, USA. .,Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA.
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT, USA
| |
Collapse
|
123
|
Taoka T, Naganawa S. Imaging for central nervous system (CNS) interstitial fluidopathy: disorders with impaired interstitial fluid dynamics. Jpn J Radiol 2021; 39:1-14. [PMID: 32653987 PMCID: PMC7813706 DOI: 10.1007/s11604-020-01017-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
After the introduction of the glymphatic system hypothesis, an increasing number of studies on cerebrospinal fluid and interstitial fluid dynamics within the brain have been investigated and reported. A series of diseases are known which develop due to abnormality of the glymphatic system including Alzheimer's disease, traumatic brain injury, stroke, or other disorders. These diseases or disorders share the characteristics of the glymphatic system dysfunction or other mechanisms related to the interstitial fluid dynamics. In this review article, we propose "Central Nervous System (CNS) Interstitial Fluidopathy" as a new concept encompassing diseases whose pathologies are majorly associated with abnormal interstitial fluid dynamics. Categorizing these diseases or disorders as "CNS interstitial fluidopathies," will promote the understanding of their mechanisms and the development of potential imaging methods for the evaluation of the disease as well as clinical methods for disease treatment or prevention. In other words, having a viewpoint of the dynamics of interstitial fluid appears relevant for understanding CNS diseases or disorders, and it would be possible to develop novel common treatment methods or medications for "CNS interstitial fluidopathies."
Collapse
Affiliation(s)
- Toshiaki Taoka
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan. .,Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
124
|
Benveniste H, Lee H, Ozturk B, Chen X, Koundal S, Vaska P, Tannenbaum A, Volkow ND. Glymphatic Cerebrospinal Fluid and Solute Transport Quantified by MRI and PET Imaging. Neuroscience 2020; 474:63-79. [PMID: 33248153 DOI: 10.1016/j.neuroscience.2020.11.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 12/13/2022]
Abstract
Over the past decade there has been an enormous progress in our understanding of fluid and solute transport in the central nervous system (CNS). This is due to a number of factors, including important developments in whole brain imaging technology and computational fluid dynamics analysis employed for the elucidation of glymphatic transport function in the live animal and human brain. In this paper, we review the technical aspects of dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) in combination with administration of Gd-based tracers into the cerebrospinal fluid (CSF) for tracking glymphatic solute and fluid transport in the CNS as well as lymphatic drainage. Used in conjunction with advanced computational processing methods including optimal mass transport analysis, one gains new insights into the biophysical forces governing solute transport in the CNS which leads to intriguing new research directions. Considering drainage pathways, we review the novel T1 mapping technique for quantifying glymphatic transport and cervical lymph node drainage concurrently in the same subject. We provide an overview of knowledge gleaned from DCE-MRI studies of glymphatic transport and meningeal lymphatic drainage. Finally, we introduce positron emission tomography (PET) and CSF administration of radiotracers as an alternative method to explore other pharmacokinetic aspects of CSF transport into brain parenchyma as well as efflux pathways.
Collapse
Affiliation(s)
- Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States; Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, United States.
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Burhan Ozturk
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Xinan Chen
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Paul Vaska
- Department of Radiology and Biomedical Engineering, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Nora D Volkow
- Laboratory for Neuroimaging, NIAAA, Bethesda, MD, United States
| |
Collapse
|
125
|
Howe MD, McCullough LD, Urayama A. The Role of Basement Membranes in Cerebral Amyloid Angiopathy. Front Physiol 2020; 11:601320. [PMID: 33329053 PMCID: PMC7732667 DOI: 10.3389/fphys.2020.601320] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Dementia is a neuropsychiatric syndrome characterized by cognitive decline in multiple domains, often leading to functional impairment in activities of daily living, disability, and death. The most common causes of age-related progressive dementia include Alzheimer's disease (AD) and vascular cognitive impairment (VCI), however, mixed disease pathologies commonly occur, as epitomized by a type of small vessel pathology called cerebral amyloid angiopathy (CAA). In CAA patients, the small vessels of the brain become hardened and vulnerable to rupture, leading to impaired neurovascular coupling, multiple microhemorrhage, microinfarction, neurological emergencies, and cognitive decline across multiple functional domains. While the pathogenesis of CAA is not well understood, it has long been thought to be initiated in thickened basement membrane (BM) segments, which contain abnormal protein deposits and amyloid-β (Aβ). Recent advances in our understanding of CAA pathogenesis link BM remodeling to functional impairment of perivascular transport pathways that are key to removing Aβ from the brain. Dysregulation of this process may drive CAA pathogenesis and provides an important link between vascular risk factors and disease phenotype. The present review summarizes how the structure and composition of the BM allows for perivascular transport pathways to operate in the healthy brain, and then outlines multiple mechanisms by which specific dementia risk factors may promote dysfunction of perivascular transport pathways and increase Aβ deposition during CAA pathogenesis. A better understanding of how BM remodeling alters perivascular transport could lead to novel diagnostic and therapeutic strategies for CAA patients.
Collapse
Affiliation(s)
| | | | - Akihiko Urayama
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
126
|
Bryniarski MA, Ren T, Rizvi AR, Snyder AM, Morris ME. Targeting the Choroid Plexuses for Protein Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12100963. [PMID: 33066423 PMCID: PMC7602164 DOI: 10.3390/pharmaceutics12100963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
Delivery of therapeutic agents to the central nervous system is challenged by the barriers in place to regulate brain homeostasis. This is especially true for protein therapeutics. Targeting the barrier formed by the choroid plexuses at the interfaces of the systemic circulation and ventricular system may be a surrogate brain delivery strategy to circumvent the blood-brain barrier. Heterogenous cell populations located at the choroid plexuses provide diverse functions in regulating the exchange of material within the ventricular space. Receptor-mediated transcytosis may be a promising mechanism to deliver protein therapeutics across the tight junctions formed by choroid plexus epithelial cells. However, cerebrospinal fluid flow and other barriers formed by ependymal cells and perivascular spaces should also be considered for evaluation of protein therapeutic disposition. Various preclinical methods have been applied to delineate protein transport across the choroid plexuses, including imaging strategies, ventriculocisternal perfusions, and primary choroid plexus epithelial cell models. When used in combination with simultaneous measures of cerebrospinal fluid dynamics, they can yield important insight into pharmacokinetic properties within the brain. This review aims to provide an overview of the choroid plexuses and ventricular system to address their function as a barrier to pharmaceutical interventions and relevance for central nervous system drug delivery of protein therapeutics. Protein therapeutics targeting the ventricular system may provide new approaches in treating central nervous system diseases.
Collapse
|
127
|
Abstract
Sleep is evolutionarily conserved across all species, and impaired sleep is a common trait of the diseased brain. Sleep quality decreases as we age, and disruption of the regular sleep architecture is a frequent antecedent to the onset of dementia in neurodegenerative diseases. The glymphatic system, which clears the brain of protein waste products, is mostly active during sleep. Yet the glymphatic system degrades with age, suggesting a causal relationship between sleep disturbance and symptomatic progression in the neurodegenerative dementias. The ties that bind sleep, aging, glymphatic clearance, and protein aggregation have shed new light on the pathogenesis of a broad range of neurodegenerative diseases, for which glymphatic failure may constitute a therapeutically targetable final common pathway.
Collapse
Affiliation(s)
- Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
128
|
Benveniste H, Elkin R, Heerdt PM, Koundal S, Xue Y, Lee H, Wardlaw J, Tannenbaum A. The glymphatic system and its role in cerebral homeostasis. J Appl Physiol (1985) 2020; 129:1330-1340. [PMID: 33002383 DOI: 10.1152/japplphysiol.00852.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The brain's high bioenergetic state is paralleled by high metabolic waste production. Authentic lymphatic vasculature is lacking in brain parenchyma. Cerebrospinal fluid (CSF) flow has long been thought to facilitate central nervous system detoxification in place of lymphatics, but the exact processes involved in toxic waste clearance from the brain remain incompletely understood. Over the past 8 yr, novel data in animals and humans have begun to shed new light on these processes in the form of the "glymphatic system," a brain-wide perivascular transit passageway dedicated to CSF transport and interstitial fluid exchange that facilitates metabolic waste drainage from the brain. Here we will discuss glymphatic system anatomy and methods to visualize and quantify glymphatic system (GS) transport in the brain and also discuss physiological drivers of its function in normal brain and in neurodegeneration.
Collapse
Affiliation(s)
- Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Rena Elkin
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York
| | - Paul M Heerdt
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Joanna Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York
| |
Collapse
|
129
|
Presa JL, Saravia F, Bagi Z, Filosa JA. Vasculo-Neuronal Coupling and Neurovascular Coupling at the Neurovascular Unit: Impact of Hypertension. Front Physiol 2020; 11:584135. [PMID: 33101063 PMCID: PMC7546852 DOI: 10.3389/fphys.2020.584135] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Components of the neurovascular unit (NVU) establish dynamic crosstalk that regulates cerebral blood flow and maintain brain homeostasis. Here, we describe accumulating evidence for cellular elements of the NVU contributing to critical physiological processes such as cerebral autoregulation, neurovascular coupling, and vasculo-neuronal coupling. We discuss how alterations in the cellular mechanisms governing NVU homeostasis can lead to pathological changes in which vascular endothelial and smooth muscle cell, pericyte and astrocyte function may play a key role. Because hypertension is a modifiable risk factor for stroke and accelerated cognitive decline in aging, we focus on hypertension-associated changes on cerebral arteriole function and structure, and the molecular mechanisms through which these may contribute to cognitive decline. We gather recent emerging evidence concerning cognitive loss in hypertension and the link with vascular dementia and Alzheimer’s disease. Collectively, we summarize how vascular dysfunction, chronic hypoperfusion, oxidative stress, and inflammatory processes can uncouple communication at the NVU impairing cerebral perfusion and contributing to neurodegeneration.
Collapse
Affiliation(s)
- Jessica L Presa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
130
|
Xue Y, Liu X, Koundal S, Constantinou S, Dai F, Santambrogio L, Lee H, Benveniste H. In vivo T1 mapping for quantifying glymphatic system transport and cervical lymph node drainage. Sci Rep 2020; 10:14592. [PMID: 32884041 PMCID: PMC7471332 DOI: 10.1038/s41598-020-71582-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022] Open
Abstract
Dynamic contrast-enhanced magnetic resonance imaging (MRI) for tracking glymphatic system transport with paramagnetic contrast such as gadoteric acid (Gd-DOTA) administration into cerebrospinal fluid (CSF) requires pre-contrast data for proper quantification. Here we introduce an alternative approach for glymphatic system quantification in the mouse brain via T1 mapping which also captures drainage of Gd-DOTA to the cervical lymph nodes. The Gd-DOTA injection into CSF was performed on the bench after which the mice underwent T1 mapping using a 3D spoiled gradient echo sequence on a 9.4 T MRI. In Ketamine/Xylazine (KX) anesthetized mice, glymphatic transport and drainage of Gd-DOTA to submandibular and deep cervical lymph nodes was demonstrated as 25–50% T1 reductions in comparison to control mice receiving CSF saline. To further validate the T1 mapping approach we also verified increased glymphatic transport of Gd-DOTA transport in mice anesthetized with KX in comparison with ISO. The novel T1 mapping method allows for quantification of glymphatic transport as well as drainage to the deep and superficial cervical lymph nodes. The ability to measure glymphatic transport and cervical lymph node drainage in the same animal longitudinally is advantageous and time efficient and the coupling between the two systems can be studied and translated to human studies.
Collapse
Affiliation(s)
- Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA.,Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA
| | - Stefan Constantinou
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA
| | - Feng Dai
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Laura Santambrogio
- Englander Institute of Precision Medicine, Department of Radiation Oncology, Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, 330 Cedar St, TMP 3, New Haven, CT, 06520, USA.
| |
Collapse
|
131
|
Ng Kee Kwong KC, Mehta AR, Nedergaard M, Chandran S. Defining novel functions for cerebrospinal fluid in ALS pathophysiology. Acta Neuropathol Commun 2020; 8:140. [PMID: 32819425 PMCID: PMC7439665 DOI: 10.1186/s40478-020-01018-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Despite the considerable progress made towards understanding ALS pathophysiology, several key features of ALS remain unexplained, from its aetiology to its epidemiological aspects. The glymphatic system, which has recently been recognised as a major clearance pathway for the brain, has received considerable attention in several neurological conditions, particularly Alzheimer's disease. Its significance in ALS has, however, been little addressed. This perspective article therefore aims to assess the possibility of CSF contribution in ALS by considering various lines of evidence, including the abnormal composition of ALS-CSF, its toxicity and the evidence for impaired CSF dynamics in ALS patients. We also describe a potential role for CSF circulation in determining disease spread as well as the importance of CSF dynamics in ALS neurotherapeutics. We propose that a CSF model could potentially offer additional avenues to explore currently unexplained features of ALS, ultimately leading to new treatment options for people with ALS.
Collapse
Affiliation(s)
- Koy Chong Ng Kee Kwong
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh bioQuarter, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Arpan R Mehta
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh bioQuarter, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - 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, NY, USA
| | - Siddharthan Chandran
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK.
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh bioQuarter, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK.
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK.
- Centre for Brain Development and Repair, inStem, Bangalore, India.
| |
Collapse
|
132
|
Zhang Y, Song J, He XZ, Xiong J, Xue R, Ge JH, Lu SY, Hu D, Zhang GX, Xu GY, Wang LH. Quantitative Determination of Glymphatic Flow Using Spectrophotofluorometry. Neurosci Bull 2020; 36:1524-1537. [PMID: 32710307 DOI: 10.1007/s12264-020-00548-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022] Open
Abstract
Following intrathecal injection of fluorescent tracers, ex vivo imaging of brain vibratome slices has been widely used to study the glymphatic system in the rodent brain. Tracer penetration into the brain is usually quantified by image-processing, even though this approach requires much time and manual operation. Here, we illustrate a simple protocol for the quantitative determination of glymphatic activity using spectrophotofluorometry. At specific time-points following intracisternal or intrastriatal injection of fluorescent tracers, certain brain regions and the spinal cord were harvested and tracers were extracted from the tissue. The intensity of tracers was analyzed spectrophotometrically and their concentrations were quantified from standard curves. Using this approach, the regional and dynamic delivery of subarachnoid CSF tracers into the brain parenchyma was assessed, and the clearance of tracers from the brain was also determined. Furthermore, the impairment of glymphatic influx in the brains of old mice was confirmed using our approach. Our method is more accurate and efficient than the imaging approach in terms of the quantitative determination of glymphatic activity, and this will be useful in preclinical studies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Jian Song
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Xu-Zhong He
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Jian Xiong
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, 215009, China
| | - Rong Xue
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Jia-Hao Ge
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Shi-Yu Lu
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Die Hu
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Guo-Xing Zhang
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China
| | - Guang-Yin Xu
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Lin-Hui Wang
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
133
|
Naessens DMP, Coolen BF, de Vos J, VanBavel E, Strijkers GJ, Bakker ENTP. Altered brain fluid management in a rat model of arterial hypertension. Fluids Barriers CNS 2020; 17:41. [PMID: 32590994 PMCID: PMC7318739 DOI: 10.1186/s12987-020-00203-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/18/2020] [Indexed: 12/22/2022] Open
Abstract
Background Proper neuronal function is directly dependent on the composition, turnover, and amount of interstitial fluid that bathes the cells. Most of the interstitial fluid is likely to be derived from ion and water transport across the brain capillary endothelium, a process that may be altered in hypertension due to vascular pathologies as endothelial dysfunction and arterial remodelling. In the current study, we investigated the effects of hypertension on the brain for differences in the water homeostasis. Methods Magnetic resonance imaging (MRI) was performed on a 7T small animal MRI system on male spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto rats (WKY) of 10 months of age. The MRI protocol consisted of T2-weighted scans followed by quantitative apparent diffusion coefficient (ADC) mapping to measure volumes of different anatomical structures and water diffusion respectively. After MRI, we assessed the spatial distribution of aquaporin 4 expression around blood vessels. Results MRI analysis revealed a significant reduction in overall brain volume and remarkably higher cerebroventricular volume in SHR compared to WKY. Whole brain ADC, as well as ADC values of a number of specific anatomical structures, were significantly lower in hypertensive animals. Additionally, SHR exhibited higher brain parenchymal water content. Immunohistochemical analysis showed a profound expression of aquaporin 4 around blood vessels in both groups, with a significantly larger area of influence around arterioles. Evaluation of specific brain regions revealed a decrease in aquaporin 4 expression around capillaries in the corpus callosum of SHR. Conclusion These results indicate a shift in the brain water homeostasis of adult hypertensive rats.
Collapse
Affiliation(s)
- Daphne M P Naessens
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Judith de Vos
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ed VanBavel
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Gustav J Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
134
|
Kananen J, Helakari H, Korhonen V, Huotari N, Järvelä M, Raitamaa L, Raatikainen V, Rajna Z, Tuovinen T, Nedergaard M, Jacobs J, LeVan P, Ansakorpi H, Kiviniemi V. Respiratory-related brain pulsations are increased in epilepsy-a two-centre functional MRI study. Brain Commun 2020; 2:fcaa076. [PMID: 32954328 PMCID: PMC7472909 DOI: 10.1093/braincomms/fcaa076] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 01/03/2023] Open
Abstract
Resting-state functional MRI has shown potential for detecting changes in cerebral blood oxygen level-dependent signal in patients with epilepsy, even in the absence of epileptiform activity. Furthermore, it has been suggested that coefficient of variation mapping of fast functional MRI signal may provide a powerful tool for the identification of intrinsic brain pulsations in neurological diseases such as dementia, stroke and epilepsy. In this study, we used fast functional MRI sequence (magnetic resonance encephalography) to acquire ten whole-brain images per second. We used the functional MRI data to compare physiological brain pulsations between healthy controls (n = 102) and patients with epilepsy (n = 33) and furthermore to drug-naive seizure patients (n = 9). Analyses were performed by calculating coefficient of variation and spectral power in full band and filtered sub-bands. Brain pulsations in the respiratory-related frequency sub-band (0.11-0.51 Hz) were significantly (P < 0.05) increased in patients with epilepsy, with an increase in both signal variance and power. At the individual level, over 80% of medicated and drug-naive seizure patients exhibited areas of abnormal brain signal power that correlated well with the known clinical diagnosis, while none of the controls showed signs of abnormality with the same threshold. The differences were most apparent in the basal brain structures, respiratory centres of brain stem, midbrain and temporal lobes. Notably, full-band, very low frequency (0.01-0.1 Hz) and cardiovascular (0.8-1.76 Hz) brain pulses showed no differences between groups. This study extends and confirms our previous results of abnormal fast functional MRI signal variance in epilepsy patients. Only respiratory-related brain pulsations were clearly increased with no changes in either physiological cardiorespiratory rates or head motion between the subjects. The regional alterations in brain pulsations suggest that mechanisms driving the cerebrospinal fluid homeostasis may be altered in epilepsy. Magnetic resonance encephalography has both increased sensitivity and high specificity for detecting the increased brain pulsations, particularly in times when other tools for locating epileptogenic areas remain inconclusive.
Collapse
Affiliation(s)
- Janne Kananen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Heta Helakari
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Vesa Korhonen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Niko Huotari
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Matti Järvelä
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Lauri Raitamaa
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Ville Raatikainen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Zalan Rajna
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Center for Machine Vision and Signal Analysis (CMVS), University of Oulu, Oulu 90014, Finland
| | - Timo Tuovinen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Julia Jacobs
- Department of Pediatric Neurology and Muscular Disease, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Pierre LeVan
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hanna Ansakorpi
- Medical Research Center (MRC), Oulu 90220, Finland
- Research Unit of Neuroscience, Neurology, University of Oulu, Oulu 90220, Finland
- Department of Neurology, Oulu University Hospital, Oulu 90029, Finland
| | - Vesa Kiviniemi
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| |
Collapse
|
135
|
Lin L, Hao X, Li C, Sun C, Wang X, Yin L, Zhang X, Tian J, Yang Y. Impaired glymphatic system in secondary degeneration areas after ischemic stroke in rats. J Stroke Cerebrovasc Dis 2020; 29:104828. [PMID: 32404284 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104828] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/16/2020] [Accepted: 03/22/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pathomechanism of secondary degeneration in remote regions after ischemic stroke has not been totally clarified. Contrast-enhanced MRI with injecting Gd-DTPA in cisterna magna (CM) is regarded as an efficient method to measure glymphatic system function in brain. Our research aimed at evaluating glymphatic system changes in secondary degeneration areas by contrast-enhanced MRI. METHODS Ischemic stroke was induced by left middle cerebral artery occlusion (MCAO) model. A total of 12 Sprague-Dawley rats were randomly divided into three groups: control group with sham operations (n=4), the group of acute phase (1 day after MCAO) (n=4), and the group of subacute phase (7 days after MCAO) (n=4). Contrast-enhanced MRI was performed in 1days or 7days after operations respectively. All rats received an intrathecal injection of Gd-DTPA (2μl/min, totally 20μl) and high-resolution 3D T1-weighted MRI for 6 h. The time course of the signal-to-noise ratio (SNR) in substantia Nigra (SN) and ventral thalamic nucleus (VTN) was evaluated between two hemispheres in all rats. RESULTS In control group without ischemia, time-to-peak of SNR in SN was earlier than that in VTN. There were no differences of SNR between two hemispheres after intrathecal Gd-DTPA administration. In the group of acute phase, MRI revealed similar time course and time-to-peak of SNR between ipsilateral and contralateral VTN, while a tendency of higher SNR in ipsilateral SN than contralateral SN at 4h, 5h, 6h after Gd-DTPA injection. And time-to-peak of SNR was similar in bilateral SN. In the group of subacute phase, time-to-peak of SNR was similar in bilateral VTN, while longer in ipsilateral SN compared with contralateral side. In addition, SNR in T1WI in ipsilateral was significantly higher than SNR in contralateral SN and VTN at 5h (VTN, P= 0.003; SN, P=0.004) and 6h (VTN, P=0.015; SN, P=0.006) after Gd-DTPA injection. CONCLUSION Glymphatic system was impaired in ipsilateral SN and VTN after ischemic stroke, which may contribute to neural degeneration.
Collapse
Affiliation(s)
- Luyi Lin
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China
| | - Xiaozhu Hao
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China
| | - Chanchan Li
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China
| | - Chengfeng Sun
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China
| | - Xiaohong Wang
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China
| | - Lekang Yin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoxue Zhang
- Department of Radiotherapy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jiaqi Tian
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanmei Yang
- Department of Radiology, Huashan Hospital, Fudan University, No. 12 Middle Urumqi Road, Shanghai 200040, China.
| |
Collapse
|
136
|
Xue Y, Liu N, Zhang M, Ren X, Tang J, Fu J. Concomitant enlargement of perivascular spaces and decrease in glymphatic transport in an animal model of cerebral small vessel disease. Brain Res Bull 2020; 161:78-83. [PMID: 32353396 DOI: 10.1016/j.brainresbull.2020.04.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/04/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVES To observe glymphatic transport and evaluate enlarged perivascular spaces (PVSs) in spontaneously hypertensive rats (SHRs). METHODS SHRs were used as an animal model of cerebral small vessel disease, and Wistar Kyoto (WKY) rats were used as the control group. Histopathology was used to evaluate the enlargement of PVSs. A fluorescent tracer was infused into the cisterna magna of rats, and the proportion of the brain parenchyma area exposed to the fluorescent tracer was later quantified to evaluate the influx and efflux function of the glymphatic system. The global and polarized expression of aquaporin protein 4 (AQP4) was analyzed by immunofluorescence. RESULTS Compared with WKY rats, SHRs exhibited obviously enlarged PVSs and significantly decreased influx and efflux function of the glymphatic system. The results showed a significant decrease in AQP4 polarity in SHRs, but a difference in global AQP4 expression was not observed between SHRs and WKY rats. CONCLUSIONS Impaired glymphatic transport may be involved in the pathogenesis of arteriolosclerotic cerebral small vessel disease, and enlarged PVSs may be a manifestation of the impaired glymphatic system.
Collapse
Affiliation(s)
- Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Na Liu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Miaoyi Zhang
- Department of Neurology, North of Huashan Hospital, Fudan University, Shanghai, China.
| | - Xue Ren
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
137
|
Keep RF, Jones HC, Drewes LR. This was the year that was: brain barriers and brain fluid research in 2019. Fluids Barriers CNS 2020; 17:20. [PMID: 32138786 PMCID: PMC7059280 DOI: 10.1186/s12987-020-00181-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research published in 2019, as well as addressing current controversies and pressing needs. Topics include recent advances related to: the cerebral endothelium and the neurovascular unit; the choroid plexus, arachnoid membrane; cerebrospinal fluid and the glymphatic hypothesis; the impact of disease states on brain barriers and brain fluids; drug delivery to the brain; and translation of preclinical data to the clinic. This editorial also mourns the loss of two important figures in the field, Malcolm B. Segal and Edward G. Stopa.
Collapse
Affiliation(s)
- Richard F. Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | | | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812 USA
| |
Collapse
|
138
|
Koundal S, Elkin R, Nadeem S, Xue Y, Constantinou S, Sanggaard S, Liu X, Monte B, Xu F, Van Nostrand W, Nedergaard M, Lee H, Wardlaw J, Benveniste H, Tannenbaum A. Optimal Mass Transport with Lagrangian Workflow Reveals Advective and Diffusion Driven Solute Transport in the Glymphatic System. Sci Rep 2020; 10:1990. [PMID: 32029859 PMCID: PMC7004986 DOI: 10.1038/s41598-020-59045-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/22/2020] [Indexed: 02/05/2023] Open
Abstract
The glymphatic system (GS) hypothesis states that advective driven cerebrospinal fluid (CSF) influx from the perivascular spaces into the interstitial fluid space rapidly transport solutes and clear waste from brain. However, the presence of advection in neuropil is contested and solutes are claimed to be transported by diffusion only. To address this controversy, we implemented a regularized version of the optimal mass transport (rOMT) problem, wherein the advection/diffusion equation is the only a priori assumption required. rOMT analysis with a Lagrangian perspective of GS transport revealed that solute speed was faster in CSF compared to grey and white matter. Further, rOMT analysis also demonstrated 2-fold differences in regional solute speed within the brain. Collectively, these results imply that advective transport dominates in CSF while diffusion and advection both contribute to GS transport in parenchyma. In a rat model of cerebral small vessel disease (cSVD), solute transport in the perivascular spaces (PVS) and PVS-to-tissue transfer was slower compared to normal rats. Thus, the analytical framework of rOMT provides novel insights in the local dynamics of GS transport that may have implications for neurodegenerative diseases. Future studies should apply the rOMT analysis approach to confirm GS transport reductions in humans with cSVD.
Collapse
Affiliation(s)
- Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Rena Elkin
- Departments of Computer Science/Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Saad Nadeem
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, NY, NY, USA
| | - Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Simon Sanggaard
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Brittany Monte
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Rhode Island, RI, USA
| | - William Van Nostrand
- George & Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Rhode Island, RI, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Joanna Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Allen Tannenbaum
- Departments of Computer Science/Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|