201
|
Li G, Cao Y, Tang X, Huang J, Cai L, Zhou L. The meningeal lymphatic vessels and the glymphatic system: Potential therapeutic targets in neurological disorders. J Cereb Blood Flow Metab 2022; 42:1364-1382. [PMID: 35484910 PMCID: PMC9274866 DOI: 10.1177/0271678x221098145] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/03/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023]
Abstract
The recent discovery of the meningeal lymphatic vessels (mLVs) and glymphatic pathways has challenged the long-lasting dogma that the central nervous system (CNS) lacks a lymphatic system and therefore does not interact with peripheral immunity. This discovery has reshaped our understanding of mechanisms underlying CNS drainage. Under normal conditions, a close connection between mLVs and the glymphatic system enables metabolic waste removal, immune cell trafficking, and CNS immune surveillance. Dysfunction of the glymphatic-mLV system can lead to toxic protein accumulation in the brain, and it contributes to the development of a series of neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. The identification of precise cerebral transport routes is based mainly on indirect, invasive imaging of animals, and the results cannot always be applied to humans. Here we review the functions of the glymphatic-mLV system and evidence for its involvement in some CNS diseases. We focus on emerging noninvasive imaging techniques to evaluate the human glymphatic-mLV system and their potential for preclinical diagnosis and prevention of neurodegenerative diseases. Potential strategies that target the glymphatic-mLV system in order to treat and prevent neurological disorders are also discussed.
Collapse
Affiliation(s)
- Gaowei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Cao
- Department of Neurosurgery, Chengdu Second People's hospital, Chengdu, China
| | - Xin Tang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhan Huang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Linjun Cai
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
202
|
Virenque A, Koivisto H, Antila S, Zub E, Rooney EJ, Miszczuk D, Müller A, Stoka E, Marchi N, Alitalo K, Tanila H, Noe FM. Significance of developmental meningeal lymphatic dysfunction in experimental post-traumatic injury. Brain Behav Immun Health 2022; 23:100466. [PMID: 35694175 PMCID: PMC9184565 DOI: 10.1016/j.bbih.2022.100466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/01/2022] Open
Abstract
Understanding the pathological mechanisms unfolding after chronic traumatic brain injury (TBI) could reveal new therapeutic entry points. During the post-TBI sequel, the involvement of cerebrospinal fluid drainage through the meningeal lymphatic vessels was proposed. Here, we used K14-VEGFR3-Ig transgenic mice to analyze whether a developmental dysfunction of meningeal lymphatic vessels modifies post-TBI pathology. To this end, a moderate TBI was delivered by controlled cortical injury over the temporal lobe in male transgenic mice or their littermate controls. We performed MRI and a battery of behavioral tests over time to define the post-TBI trajectories. In vivo analyses were integrated by ex-vivo quantitative and morphometric examinations of the cortical lesion and glial cells. In post-TBI K14-VEGFR3-Ig mice, the recovery from motor deficits was protracted compared to littermates. This outcome is coherent with the observed slower hematoma clearance in transgenic mice during the first two weeks post-TBI. No other genotype-related behavioral differences were observed, and the volume of cortical lesions imaged by MRI in vivo, and confirmed by histology ex-vivo, were comparable in both groups. However, at the cellular level, post-TBI K14-VEGFR3-Ig mice exhibited an increased percentage of activated Iba1 microglia in the hippocampus and auditory cortex, areas that are proximal to the lesion. Although not impacting or modifying the structural brain damage and post-TBI behavior, a pre-existing dysfunction of meningeal lymphatic vessels is associated with morphological microglial activation over time, possibly representing a sub-clinical pathological imprint or a vulnerability factor. Our findings suggest that pre-existing mLV deficits could represent a possible risk factor for the overall outcome of TBI pathology. Developmental deficit in the meningeal lymphatic vessels contributes to sustain the chronic neuroinflammation and represent a susceptibility factor in TBI, despite the lack of a functional phenotype. Development and progression of TBI-related cortical lesion is not exacerbated by developmental deficit in meningeal lymphatics. Meningeal lymphatic developmental deficits result in increased neuroinflammation, suggesting a sub-clinical pathological imprint or a vulnerability factor. Congenital mLV deficit affects the interstitial fluid dynamics and the post-TBI hematoma resolution.
Collapse
Affiliation(s)
- Anaïs Virenque
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Hennariikka Koivisto
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Emma Zub
- Cerebrovascular and Glia Research, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Erin Jane Rooney
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Diana Miszczuk
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Adrian Müller
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Enija Stoka
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Heikki Tanila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Francesco Mattia Noe
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
- Corresponding author. HiLIFE, Neuroscience Center, Helsinki University, Helsinki, Finland.
| |
Collapse
|
203
|
Yoo SS, Kim HC, Kim J, Kim E, Kowsari K, Van Reet J, Yoon K. Enhancement of cerebrospinal fluid tracer movement by the application of pulsed transcranial focused ultrasound. Sci Rep 2022; 12:12940. [PMID: 35902724 PMCID: PMC9334279 DOI: 10.1038/s41598-022-17314-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022] Open
Abstract
Efficient transport of solutes in the cerebrospinal fluid (CSF) plays a critical role in their clearance from the brain. Convective bulk flow of solutes in the CSF in the perivascular space (PVS) is considered one of the important mechanisms behind solute movement in the brain, before their ultimate drainage to the systemic lymphatic system. Acoustic pressure waves can impose radiation force on a medium in its path, inducing localized and directional fluidic flow, known as acoustic streaming. We transcranially applied low-intensity focused ultrasound (FUS) to rats that received an intracisternal injection of fluorescent CSF tracers (dextran and ovalbumin, having two different molecular weights-Mw). The sonication pulsing parameter was determined on the set that propelled the aqueous solution of toluidine blue O dye into a porous media (melamine foam) at the highest level of infiltration. Fluorescence imaging of the brain showed that application of FUS increased the uptake of ovalbumin at the sonicated plane, particularly around the ventricles, whereas the uptake of high-Mw dextran was unaffected. Numerical simulation showed that the effects of sonication were non-thermal. Sonication did not alter the animals' behavior or disrupt the blood-brain barrier (BBB) while yielding normal brain histology. The results suggest that FUS may serve as a new non-invasive means to promote interstitial CSF solute transport in a region-specific manner without disrupting the BBB, providing potential for enhanced clearance of waste products from the brain.
Collapse
Affiliation(s)
- Seung-Schik Yoo
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Hyun-Chul Kim
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
- Department of Artificial Intelligence, Kyungpook National University, Daegu, Republic of Korea
| | - Jaeho Kim
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Evgenii Kim
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Kavin Kowsari
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Jared Van Reet
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Kyungho Yoon
- School of Mathematics and Computing (Computational Science and Engineering), Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
204
|
Standlee J, Malkani R. Sleep Dysfunction in Movement Disorders: a Window to the Disease Biology. Curr Neurol Neurosci Rep 2022; 22:565-576. [PMID: 35867306 DOI: 10.1007/s11910-022-01220-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To comprehensively summarize the sleep pathologies associated with movement disorders, focusing on neurodegenerative diseases. RECENT FINDINGS Mounting evidence has further implicated both sleep and circadian disruption in the pathophysiology of many movement disorders. In particular, recent data illuminate the mechanisms by which poor sleep quality and circadian dysfunction can exacerbate neurodegeneration. In addition, anti-IgLON5 disease is a recently described autoimmune disease with various symptoms that can feature prominent sleep disruption and parasomnia. Many movement disorders are associated with sleep and circadian rhythm disruption. Motor symptoms can cause sleep fragmentation, resulting in insomnia and excessive daytime sleepiness. Many neurodegenerative movement disorders involve brainstem pathology in regions close to or affecting nuclei that regulate sleep and wake. Further, commonly used movement medications may exacerbate sleep concerns. Providers should screen for and address these sleep symptoms to improve function and quality of life for patients and caregivers.
Collapse
Affiliation(s)
- Jordan Standlee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Roneil Malkani
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Jesse Brown Veterans Affairs Medical Center, Neurology Service, 820 S Damen Ave, Damen Building, 9th floor, Chicago, IL, 60612, USA.
| |
Collapse
|
205
|
Wang GQ, Wang FX, He YN, Lin JY. Plasticity of the spinal glymphatic system in male SD rats with painful diabetic neuropathy induced by type 2 diabetes mellitus. J Neurosci Res 2022; 100:1908-1920. [PMID: 35796387 PMCID: PMC9541551 DOI: 10.1002/jnr.25104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/06/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Abstract
The glymphatic system is a recently discovered glial‐dependent macroscopic interstitial waste clearance system that promotes the efficient elimination of soluble proteins and metabolites from the central nervous system. Its anatomic foundation is the astrocytes and aquaporin‐4 (AQP4) water channels on the endfeet of astrocytes. The aim of this study is to evaluate the plasticity of the spinal glymphatic system in male SD rats with painful diabetic neuropathy (PDN) induced by type 2 diabetes mellitus. PDN rats were modeled under a high‐fat and high‐glucose diet with a low dose of streptozotocin. MRI was applied to observe the infiltration and clearance of contrast to indicate the functional variability of the glymphatic system at the spinal cord level. The paw withdrawal threshold was used to represent mechanical allodynia. The numerical change of glial fibrillary acidic protein (GFAP) positive astrocytes was assessed and the polarity reversal of AQP4 protein was measured by immunofluorescence. As a result, deceased contrast infiltration and clearance, enhanced mechanical allodynia, increased number of GFAP positive astrocytes, and reversed polarity of AQP4 protein were found in the PDN rats. The above molecular level changes may contribute to the impairment of the spinal glymphatic system in PDN rats. This study revealed the molecular and functional variations of the spinal glymphatic system in PDN rats and for the first time indicated that there might be a correlation between the impaired spinal glymphatic system and PDN rats.
Collapse
Affiliation(s)
- Guo-Qiang Wang
- Department of Anesthesiology, the Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Pain Treatment, Physical and Mental Hospital of Nanchong City, Nanchong, China
| | - Fei-Xiang Wang
- Department of Anesthesiology, the Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yi-Na He
- Department of Anesthesiology, Nanchong Central Hospital, Nanchong, China
| | - Jing-Yan Lin
- Department of Anesthesiology, the Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
206
|
Lee MH, Lee SK, Kim S, Kim REY, Nam HR, Siddiquee AT, Thomas RJ, Hwang I, Yoon JE, Yun CH, Shin C. Association of Obstructive Sleep Apnea With White Matter Integrity and Cognitive Performance Over a 4-Year Period in Middle to Late Adulthood. JAMA Netw Open 2022; 5:e2222999. [PMID: 35857321 PMCID: PMC9301517 DOI: 10.1001/jamanetworkopen.2022.22999] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Obstructive sleep apnea (OSA) is associated with cognitive impairment and brain structural alterations, but longitudinal outcomes are understudied. OBJECTIVE To examine the associations of OSA with cognition and white matter (WM) integrity over a 4-year period. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study was conducted in a community-based adult population among participants who had both baseline (2011-2014) and 4-year follow-up (2015-2018) polysomnography, diffusion tensor imaging, and cognitive assessment data. Participants with neurological disorders, anomalous findings on brain magnetic resonance imaging, or inadequate quality of the evaluations were excluded. Data were analyzed from March to November 2021. EXPOSURES Participants were categorized depending on the presence vs absence of OSA at baseline and follow-up polysomnographic analysis. MAIN OUTCOMES AND MEASURES The main outcomes were proportional changes over a 4-year period in neuropsychological performance and WM integrity. The neuropsychological assessment battery included verbal and visual memory, verbal fluency, Digit Symbol-coding, Trail Making Test-A, and Stroop Test. WM integrity was assessed by fractional anisotropy, axial, and radial diffusivity. To examine interactions with age and sex, participants were subgrouped by age older than 60 years vs 60 years or younger and men vs women. RESULTS A total of 1998 individuals were assessed for eligibility, and 888 were excluded based on exclusion criteria, leaving 1110 participants (mean [SD] age, 58.0 [6.0] years; 517 [46.6%] men) for analysis, including 458 participants grouped as OSA-free, 72 participants with resolved OSA, 163 participants with incident OSA, and 417 participants with persistent OSA. Incident OSA was associated with altered WM integrity and with concomitant changes in sustained attention compared with participants without OSA (eg, change in Digit Symbol-coding test score, -3.2% [95% CI, -5.2% to -1.2%]). Participants with resolved OSA showed better visual recall at the follow-up (change in Visual Reproduction-immediate recall test, 17.5% [95% CI, 8.9% to 26.1%]; change in Visual Reproduction-delayed recall test, 33.1% [95% CI, 11.3% to 54.9%]), with concordant changes in diffusion parameters at the relevant anatomic areas. In the older group only (age >60 years), persistent OSA was associated with altered WM integrity and cognition (eg, Visual Reproduction-recognition test: β = -24.2 [95% CI, -40.7 to -7.7]). Sex also was associated with modifying the association of OSA with WM integrity of the left posterior internal capsule, the left genu of corpus callosum, and the right middle cerebellar peduncle only in men and with cognition only in women (eg, Visual Reproduction-immediate recall test: β = 33.4 [95% CI, 19.1 to 47.7]). CONCLUSIONS AND RELEVANCE These findings suggest that dynamic changes in OSA status were significantly associated with WM integrity and cognition, which varied by age and sex. It is possible that adequate interventions for OSA could better preserve brain health in middle to late adulthood.
Collapse
Affiliation(s)
- Min-Hee Lee
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Seung Ku Lee
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Soriul Kim
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Regina E. Y. Kim
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Hye Ryeong Nam
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Ali T. Siddiquee
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Robert J. Thomas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Inha Hwang
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jee-Eun Yoon
- Department of Neurology, Uijeongbu Eulji Medical Center, Uijeongbu, Republic of Korea
| | - Chang-Ho Yun
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Chol Shin
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
- Department of Pulmonary Sleep and Critical Care Medicine Disorder Center, College of Medicine, Korea University, Ansan, Republic of Korea
| |
Collapse
|
207
|
Keil SA, Braun M, O’Boyle R, Sevao M, Pedersen T, Agarwal S, Jansson D, Iliff JJ. Dynamic infrared imaging of cerebrospinal fluid tracer influx into the brain. NEUROPHOTONICS 2022; 9:031915. [PMID: 35602461 PMCID: PMC9113559 DOI: 10.1117/1.nph.9.3.031915] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/19/2022] [Indexed: 05/14/2023]
Abstract
Significance: The glymphatic system has been described recently as a series of perivascular channels that facilitate fluid exchange and solute clearance in the brain. Glymphatic dysfunction has been implicated in numerous pathological conditions, including Alzheimer's disease, traumatic brain injury, and stroke. Existing methods for assessing glymphatic function have been challenging: dynamic methods, such as two-photon microscopy and contrast-enhanced magnetic resonance imaging require expensive instrumentation and specific technical skills; slice-based fluorescent imaging is more readily implemented but lacks temporal resolution. Aim: To develop a straightforward and adaptable dynamic imaging approach for assessing glymphatic function in vivo in mice. Approach: Using a widely available small animal infrared (IR) imaging system (LICOR Pearl), visualization of IR cerebrospinal fluid tracer distribution over the cortical surface enables time-resolved measurement of the dynamics of glymphatic exchange. Using co-injection of IR and conventional fixable fluorescent tracers, dynamic imaging can be paired with whole-slice fluorescence imaging, permitting the quantification of glymphatic function throughout the brain as well as subsequent histological assessment. Results: These techniques were validated against one another, comparing differences between animals anesthetized with ketamine/xylazine and isoflurane. Conclusions: This technique permits sensitive dynamic imaging of glymphatic function, with the concurrent visualization of resolution of deeper structures.
Collapse
Affiliation(s)
- Samantha A. Keil
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
- University of Washington School of Medicine, Department of Psychiatry and Behavioral Sciences, Seattle, Washington, United States
| | - Molly Braun
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
- University of Washington School of Medicine, Department of Psychiatry and Behavioral Sciences, Seattle, Washington, United States
| | - Ryan O’Boyle
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
| | - Mathew Sevao
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
| | - Taylor Pedersen
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
| | - Sanjana Agarwal
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
| | - Deidre Jansson
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
- University of Washington School of Medicine, Department of Psychiatry and Behavioral Sciences, Seattle, Washington, United States
| | - Jeffrey J. Iliff
- VA Puget Sound Health Care System, VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), Seattle, Washington, United States
- University of Washington School of Medicine, Department of Psychiatry and Behavioral Sciences, Seattle, Washington, United States
- University of Washington School of Medicine, Department of Neurology, Seattle, Washington, United States
| |
Collapse
|
208
|
Quintin S, Barpujari A, Mehkri Y, Hernandez J, Lucke-Wold B. The glymphatic system and subarachnoid hemorrhage: disruption and recovery. EXPLORATION OF NEUROPROTECTIVE THERAPY 2022:118-130. [PMID: 35756328 PMCID: PMC9221287 DOI: 10.37349/ent.2022.00023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023]
Abstract
The glymphatic system, or glial-lymphatic system, is a waste clearance system composed of perivascular channels formed by astrocytes that mediate the clearance of proteins and metabolites from the brain. These channels facilitate the movement of cerebrospinal fluid throughout brain parenchyma and are critical for homeostasis. Disruption of the glymphatic system leads to an accumulation of these waste products as well as increased interstitial fluid in the brain. These phenomena are also seen during and after subarachnoid hemorrhages (SAH), contributing to the brain damage seen after rupture of a major blood vessel. Herein this review provides an overview of the glymphatic system, its disruption during SAH, and its function in recovery following SAH. The review also outlines drugs which target the glymphatic system and may have therapeutic applications following SAH.
Collapse
Affiliation(s)
- Stephan Quintin
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Arnav Barpujari
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Yusuf Mehkri
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Jairo Hernandez
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
209
|
Newell DW, Nedergaard M, Aaslid R. Physiological Mechanisms and Significance of Intracranial B Waves. Front Neurol 2022; 13:872701. [PMID: 35651339 PMCID: PMC9149212 DOI: 10.3389/fneur.2022.872701] [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: 02/09/2022] [Accepted: 04/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Recently published studies have described slow spontaneous cerebral blood flow (CBF) and cerebrospinal fluid (CSF) oscillations measured by magnetic resonance imaging (MRI) as potential drivers of brain glymphatic flow, with a similar frequency as intracranial B-waves. Aiming to establish the relationship between these waveforms, we performed additional analysis of frequency and waveform parameters, of our previously published transcranial Doppler (TCD) and intracranial pressure (ICP) recordings of intracranial B waves, to compare to published MRI frequency measurements of CBF and CSF slow oscillations. Patients and Methods We analyzed digital recordings of B waves in 29 patients with head injury, including middle cerebral artery (MCA) flow velocity (FV), ICP, end tidal CO2, and arterial blood pressure (ABP). A subset of these recordings demonstrated high B wave activity and was further analyzed for parameters including frequency, interaction, and waveform distribution curve features. These measures were compared to published similar measurements of spontaneous CBF and CSF fluctuations evaluated using MRI. Results In patients with at least 10% amplitude B wave activity, the MCA blood flow velocity oscillations comprising the B waves, had a maximum amplitude at 0.0245 Hz, and time derivative a maximum amplitude at 0.035 Hz. The frequency range of the B waves was between 0.6–2.3 cycles per min (0.011-0.038 Hz), which is in the same range as MRI measured CBF slow oscillations, reported in human volunteers. Waveform asymmetry in MCA velocity and ICP cycles during B waves, was also similar to published MRI measured CBF slow oscillations. Cross-correlation analysis showed equivalent time derivatives of FV vs. ICP in B waves, compared to MRI measured CBF slow oscillations vs. CSF flow fluctuations. Conclusions The TCD and ICP recordings of intracranial B waves show a similar frequency range as CBF and CSF flow oscillations measured using MRI, and share other unique morphological wave features. These findings strongly suggest a common physiological mechanism underlying the two classes of phenomena. The slow blood flow and volume oscillations causing intracranial B waves appear to be part of a cascade that may provide a significant driving force for compartmentalized CSF movement and facilitate glymphatic flow.
Collapse
Affiliation(s)
- David W Newell
- Department of Neurosurgery, Seattle Neuroscience Institute, Seattle, WA, United States
| | - Maiken Nedergaard
- Department of Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Translational Neuromedicine, University of Rochester Medical School, Rochester, NY, United States
| | - Rune Aaslid
- Department of Neurosurgery, University of Bern, Bern, Switzerland
| |
Collapse
|
210
|
Kim J, Lee DA, Lee HJ, Park BS, Ko J, Park SH, Lee YJ, Kim IH, Park JH, Park KM. Glymphatic system dysfunction in patients with cluster headache. Brain Behav 2022; 12:e2631. [PMID: 35582786 PMCID: PMC9226822 DOI: 10.1002/brb3.2631] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/31/2022] [Accepted: 05/03/2022] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION The aim of this study was to investigate alterations of the glymphatic system function in patients with cluster headache. METHODS We enrolled patients with cluster headache and healthy controls, and they underwent brain magnetic resonance imaging (MRI), including diffusion tensor imaging (DTI). We used the MRIcron and DSI studio programs for DTI preprocessing and DTI analysis with perivascular space (DTI-ALPS) index calculation. RESULTS Fourteen patients with cluster headache and 23 healthy controls were enrolled. The DTI-ALPS indexes of the groups were significantly different. The DTI-ALPS index for the patients with cluster headache was lower than that for the healthy controls (1.586 vs. 1.786, p = 0.044). There was a significant negative correlation between the DTI-ALPS index and age in the patients with cluster headache (r = -0.549, p = 0.042). However, the DTI-ALPS index was not associated with other clinical characteristics, including disease duration and headache intensity (r = -0.405, p = 0.150; r = -0.048, p = 0.869, respectively). CONCLUSION Patients with cluster headache had a lower DTI-ALPS index than the healthy controls; this might indicate glymphatic system dysfunction in the patients with cluster headache. Further research is required to determine whether glymphatic system dysfunction is related to the pathophysiology of cluster headache.
Collapse
Affiliation(s)
- Jinseung Kim
- Department of Family Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Bong Soo Park
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Junghae Ko
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Si Hyung Park
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Yoo Jin Lee
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Il Hwan Kim
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Jin Han Park
- Department of Internal medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
211
|
Boster KAS, Tithof J, Cook DD, Thomas JH, Kelley DH. Sensitivity analysis on a network model of glymphatic flow. J R Soc Interface 2022; 19:20220257. [PMID: 35642425 PMCID: PMC9156905 DOI: 10.1098/rsif.2022.0257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/05/2022] [Indexed: 12/24/2022] Open
Abstract
Intracranial cerebrospinal and interstitial fluid (ISF) flow and solute transport have important clinical implications, but limited in vivo access to the brain interior leaves gaping holes in human understanding of the nature of these neurophysiological phenomena. Models can address some gaps, but only insofar as model inputs are accurate. We perform a sensitivity analysis using a Monte Carlo approach on a lumped-parameter network model of cerebrospinal and ISF in perivascular and extracellular spaces in the murine brain. We place bounds on model predictions given the uncertainty in input parameters. Péclet numbers for transport in penetrating perivascular spaces (PVSs) and within the parenchyma are separated by at least two orders of magnitude. Low permeability in penetrating PVSs requires unrealistically large driving pressure and/or results in poor perfusion and are deemed unlikely. The model is most sensitive to the permeability of penetrating PVSs, a parameter whose value is largely unknown, highlighting an important direction for future experiments. Until the value of the permeability of penetrating PVSs is more accurately measured, the uncertainty of any model that includes flow in penetrating PVSs is so large that absolute numbers have little meaning and practical application is limited.
Collapse
Affiliation(s)
- Kimberly A. S. Boster
- Department of Mechanical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Douglas D. Cook
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602, USA
| | - John H. Thomas
- Department of Mechanical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
212
|
The Underlying Role of the Glymphatic System and Meningeal Lymphatic Vessels in Cerebral Small Vessel Disease. Biomolecules 2022; 12:biom12060748. [PMID: 35740873 PMCID: PMC9221030 DOI: 10.3390/biom12060748] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
There is a growing prevalence of vascular cognitive impairment (VCI) worldwide, and most research has suggested that cerebral small vessel disease (CSVD) is the main contributor to VCI. Several potential physiopathologic mechanisms have been proven to be involved in the process of CSVD, such as blood-brain barrier damage, small vessels stiffening, venous collagenosis, cerebral blood flow reduction, white matter rarefaction, chronic ischaemia, neuroinflammation, myelin damage, and subsequent neurodegeneration. However, there still is a limited overall understanding of the sequence and the relative importance of these mechanisms. The glymphatic system (GS) and meningeal lymphatic vessels (mLVs) are the analogs of the lymphatic system in the central nervous system (CNS). As such, these systems play critical roles in regulating cerebrospinal fluid (CSF) and interstitial fluid (ISF) transport, waste clearance, and, potentially, neuroinflammation. Accumulating evidence has suggested that the glymphatic and meningeal lymphatic vessels played vital roles in animal models of CSVD and patients with CSVD. Given the complexity of CSVD, it was significant to understand the underlying interaction between glymphatic and meningeal lymphatic transport with CSVD. Here, we provide a novel framework based on new advances in main four aspects, including vascular risk factors, potential mechanisms, clinical subtypes, and cognition, which aims to explain how the glymphatic system and meningeal lymphatic vessels contribute to the progression of CSVD and proposes a comprehensive insight into the novel therapeutic strategy of CSVD.
Collapse
|
213
|
Piirainen P, Kokki H, Kokki M. Epidural Oxycodone for Acute Pain. Pharmaceuticals (Basel) 2022; 15:643. [PMID: 35631469 PMCID: PMC9144954 DOI: 10.3390/ph15050643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Epidural analgesia is commonly used in labour analgesia and in postoperative pain after major surgery. It is highly effective in severe acute pain, has minimal effects on foetus and newborn, may reduce postoperative complications, and enhance patient satisfaction. In epidural analgesia, low concentrations of local anaesthetics are combined with opioids. Two opioids, morphine and sufentanil, have been approved for epidural use, but there is an interest in evaluating other opioids as well. Oxycodone is one of the most commonly used opioids in acute pain management. However, data on its use in epidural analgesia are sparse. In this narrative review, we describe the preclinical and clinical data on epidural oxycodone. Early data from the 1990s suggested that the epidural administration of oxycodone may not offer any meaningful benefits over intravenous administration, but more recent clinical data show that oxycodone has advantageous pharmacokinetics after epidural administration and that epidural administration is more efficacious than intravenous administration. Further studies are needed on the safety and efficacy of continuous epidural oxycodone administration and its use in epidural admixture.
Collapse
Affiliation(s)
- Panu Piirainen
- Department of Anesthesiology, Surgery and Intensive Care, Oulu University Hospital, 90220 Oulu, Finland;
| | - Hannu Kokki
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, Kuopio Campus, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Merja Kokki
- Department of Anaesthesiology and Intensive Care, Kuopio University Hospital, 70210 Kuopio, Finland
| |
Collapse
|
214
|
Tithof J, Boster KA, Bork PA, Nedergaard M, Thomas JH, Kelley DH. A network model of glymphatic flow under different experimentally-motivated parametric scenarios. iScience 2022; 25:104258. [PMID: 35521514 PMCID: PMC9062681 DOI: 10.1016/j.isci.2022.104258] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022] Open
Abstract
Flow of cerebrospinal fluid (CSF) through perivascular spaces (PVSs) in the brain delivers nutrients, clears metabolic waste, and causes edema formation. Brain-wide imaging cannot resolve PVSs, and high-resolution methods cannot access deep tissue. However, theoretical models provide valuable insight. We model the CSF pathway as a network of hydraulic resistances, using published parameter values. A few parameters (permeability of PVSs and the parenchyma, and dimensions of PVSs and astrocyte endfoot gaps) have wide uncertainties, so we focus on the limits of their ranges by analyzing different parametric scenarios. We identify low-resistance PVSs and high-resistance parenchyma as the only scenario that satisfies three essential criteria: that the flow be driven by a small pressure drop, exhibit good CSF perfusion throughout the cortex, and exhibit a substantial increase in flow during sleep. Our results point to the most important parameters, such as astrocyte endfoot gap dimensions, to be measured in future experiments. We model the CSF pathway as a network of hydraulic resistances Predictions are bracketed by analyzing parametric scenarios for unknown parameters Low-resistance PVSs and high-resistance parenchyma produce realistic flows Astrocyte endfoot gap size is among the important parameters to be measured
Collapse
Affiliation(s)
- Jeffrey Tithof
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis 55455, MN, USA
- Corresponding author
| | - Kimberly A.S. Boster
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| | - Peter A.R. Bork
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester 14642, NY, USA
| | - John H. Thomas
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, 235 Hopeman Building, Rochester 14627, NY, USA
| |
Collapse
|
215
|
Yi T, Gao P, Zhu T, Yin H, Jin S. Glymphatic System Dysfunction: A Novel Mediator of Sleep Disorders and Headaches. Front Neurol 2022; 13:885020. [PMID: 35665055 PMCID: PMC9160458 DOI: 10.3389/fneur.2022.885020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep contributes to the maintenance of overall health and well-being. There are a growing number of patients who have headache disorders that are significantly affected by poor sleep. This is a paradoxical relationship, whereby sleep deprivation or excess sleep leads to a worsening of headaches, yet sleep onset also alleviates ongoing headache pain. Currently, the mechanism of action remains controversial and poorly understood. The glymphatic system is a newly discovered perivascular network that encompasses the whole brain and is responsible for removing toxic proteins and waste metabolites from the brain as well as replenishing nutrition and energy. Recent studies have suggested that glymphatic dysfunction is a common underlying etiology of sleep disorders and headache pain. This study reviews the current literature on the relationship between the glymphatic system, sleep, and headaches, discusses their roles, and proposes acupuncture as a non-invasive way to focus on the glymphatic function to improve sleep quality and alleviate headache pain.
Collapse
Affiliation(s)
- Ting Yi
- Rehabilitation and Health Preservation School, Chengdu University of TCM, Chengdu, China
| | - Ping Gao
- Rehabilitation and Health Preservation School, Chengdu University of TCM, Chengdu, China
| | - Tianmin Zhu
- Rehabilitation and Health Preservation School, Chengdu University of TCM, Chengdu, China
- Tianmin Zhu
| | - Haiyan Yin
- School of Acupuncture and Tuina, Chengdu University of TCM, Chengdu, China
- *Correspondence: Haiyan Yin
| | - Shuoguo Jin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Shuoguo Jin
| |
Collapse
|
216
|
Kedarasetti RT, Drew PJ, Costanzo F. Arterial vasodilation drives convective fluid flow in the brain: a poroelastic model. Fluids Barriers CNS 2022; 19:34. [PMID: 35570287 PMCID: PMC9107702 DOI: 10.1186/s12987-022-00326-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/29/2022] [Indexed: 01/26/2023] Open
Abstract
The movement of fluid into, through, and out of the brain plays an important role in clearing metabolic waste. However, there is controversy regarding the mechanisms driving fluid movement in the fluid-filled paravascular spaces (PVS), and whether the movement of metabolic waste in the brain extracellular space (ECS) is primarily driven by diffusion or convection. The dilation of penetrating arterioles in the brain in response to increases in neural activity (neurovascular coupling) is an attractive candidate for driving fluid circulation, as it drives deformation of the brain tissue and of the PVS around arteries, resulting in fluid movement. We simulated the effects of vasodilation on fluid movement into and out of the brain ECS using a novel poroelastic model of brain tissue. We found that arteriolar dilations could drive convective flow through the ECS radially outward from the arteriole, and that this flow is sensitive to the dynamics of the dilation. Simulations of sleep-like conditions, with larger vasodilations and increased extracellular volume in the brain showed enhanced movement of fluid from the PVS into the ECS. Our simulations suggest that both sensory-evoked and sleep-related arteriolar dilations can drive convective flow of cerebrospinal fluid not just in the PVS, but also into the ECS through the PVS around arterioles.
Collapse
Affiliation(s)
- Ravi Teja Kedarasetti
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Patrick J. Drew
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Neurosurgery, Pennsylvania State University, University Park, PA USA
| | - Francesco Costanzo
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Mathematics, Pennsylvania State University, University Park, PA USA
| |
Collapse
|
217
|
Zhang D, Li X, Li B. Glymphatic System Dysfunction in Central Nervous System Diseases and Mood Disorders. Front Aging Neurosci 2022; 14:873697. [PMID: 35547631 PMCID: PMC9082304 DOI: 10.3389/fnagi.2022.873697] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
The glymphatic system, a recently discovered macroscopic waste removal system in the brain, has many unknown aspects, especially its driving forces and relationship with sleep, and thus further explorations of the relationship between the glymphatic system and a variety of possible related diseases are urgently needed. Here, we focus on the progress in current research on the role of the glymphatic system in several common central nervous system diseases and mood disorders, discuss the structural and functional abnormalities of the glymphatic system which may occur before or during the pathophysiological progress and the possible underlying mechanisms. We emphasize the relationship between sleep and the glymphatic system under pathological conditions and summarize the common imaging techniques for the glymphatic system currently available. The perfection of the glymphatic system hypothesis and the exploration of the effects of aging and endocrine factors on the central and peripheral regulatory pathways through the glymphatic system still require exploration in the future.
Collapse
Affiliation(s)
- Dianjun Zhang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinyu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
218
|
[64Cu]Cu-Albumin Clearance Imaging to Evaluate Lymphatic Efflux of Cerebrospinal Space Fluid in Mouse Model. Nucl Med Mol Imaging 2022; 56:137-146. [DOI: 10.1007/s13139-022-00746-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022] Open
|
219
|
Picchioni D, Özbay PS, Mandelkow H, de Zwart JA, Wang Y, van Gelderen P, Duyn JH. Autonomic arousals contribute to brain fluid pulsations during sleep. Neuroimage 2022; 249:118888. [PMID: 35017126 PMCID: PMC11395500 DOI: 10.1016/j.neuroimage.2022.118888] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/15/2021] [Accepted: 01/05/2022] [Indexed: 12/28/2022] Open
Abstract
During sleep, slow waves of neuro-electrical activity engulf the human brain and aid in the consolidation of memories. Recent research suggests that these slow waves may also promote brain health by facilitating the removal of metabolic waste, possibly by orchestrating the pulsatile flow of cerebrospinal fluid (CSF) through local neural control over vascular tone. To investigate the role of slow waves in the generation of CSF pulsations, we analyzed functional MRI data obtained across the full sleep-wake cycle and during a waking respiratory task. This revealed a novel generating mechanism that relies on the autonomic regulation of cerebral vascular tone without requiring slow electrocortical activity or even sleep. Therefore, the role of CSF pulsations in brain waste clearance may, in part, depend on proper autoregulatory control of cerebral blood flow.
Collapse
Affiliation(s)
- Dante Picchioni
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Pinar S Özbay
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Hendrik Mandelkow
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Jacco A de Zwart
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Yicun Wang
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Peter van Gelderen
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States
| | - Jeff H Duyn
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, United States.
| |
Collapse
|
220
|
Lee DA, Lee H, Park KM. Glymphatic dysfunction in isolated REM sleep behavior disorder. Acta Neurol Scand 2022; 145:464-470. [PMID: 34918348 DOI: 10.1111/ane.13573] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/26/2021] [Accepted: 12/08/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES This study aimed to evaluate glymphatic-system function in patients with isolated rapid eye movement sleep behavior disorder (iRBD) in comparison with healthy controls by using diffusion tensor imaging (DTI) along the perivascular space (DTI-ALPS) method. We hypothesized that patients with iRBD may show glymphatic-system dysfunction. METHODS We retrospectively enrolled 18 patients with iRBD and 18 age- and sex-matched healthy controls. All participants underwent DTI magnetic resonance imaging (MRI) using the same 3T MRI scanner, and the DTI-ALPS index was calculated using DTI data. We evaluated the differences in the DTI-ALPS index between patients with iRBD and healthy controls. In addition, we evaluated the correlation between the DTI-ALPS index and demographic and polysomnographic characteristics. RESULTS The DTI-ALPS index was significantly different between the groups; it was significantly lower in patients with iRBD than in healthy controls (1.5647 vs. 1.7612, p = .0157). The index did not correlate with demographic and polysomnographic characteristics, including age, Epworth Sleepiness Scale score, total sleep time, sleep efficiency, sleep stage N1 ratio, stage N2 ratio, stage N3 ratio, stage R ratio, and total apnea-hypopnea index. CONCLUSION The DTI-ALPS index was significantly lower in patients with iRBD than in healthy controls, indicating the presence of glymphatic-system dysfunction in patients with iRBD. Our study also suggests that the DTI-ALPS index could serve as a biomarker for evaluating glymphatic-system function in neurological disorders.
Collapse
Affiliation(s)
- Dong Ah Lee
- Department of Neurology Haeundae Paik Hospital Inje University College of Medicine Busan Korea
| | - Ho‐Joon Lee
- Department of Radiology Haeundae Paik Hospital Inje University College of Medicine Busan Korea
| | - Kang Min Park
- Department of Neurology Haeundae Paik Hospital Inje University College of Medicine Busan Korea
| |
Collapse
|
221
|
Hanke JM, Schindler KA, Seiler A. On the relationships between epilepsy, sleep, and Alzheimer's disease: A narrative review. Epilepsy Behav 2022; 129:108609. [PMID: 35176650 DOI: 10.1016/j.yebeh.2022.108609] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 12/21/2022]
Abstract
Epilepsy, sleep, and Alzheimer's disease (AD) are tightly and potentially causally interconnected. The aim of our review was to investigate current research directions on these relationships. Our hope is that they may indicate preventive measures and new treatment options for early neurodegeneration. We included articles that assessed all three topics and were published during the last ten years. We found that this literature corroborates connections on various pathophysiological levels, including sleep-stage-related epileptiform activity in AD, the negative consequences of different sleep disorders on epilepsy and cognition, common biochemical pathways as well as network dysfunctions. Here we provide a detailed overview of these topics and we discuss promising diagnostic and therapeutic consequences.
Collapse
Affiliation(s)
- Julie M Hanke
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland
| | - Kaspar A Schindler
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland
| | - Andrea Seiler
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland.
| |
Collapse
|
222
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
223
|
Salman MM, Kitchen P, Halsey A, Wang MX, Törnroth-Horsefield S, Conner AC, Badaut J, Iliff JJ, Bill RM. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2022; 145:64-75. [PMID: 34499128 PMCID: PMC9088512 DOI: 10.1093/brain/awab311] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/28/2021] [Accepted: 07/31/2021] [Indexed: 11/25/2022] Open
Abstract
Aquaporin channels facilitate bidirectional water flow in all cells and tissues. AQP4 is highly expressed in astrocytes. In the CNS, it is enriched in astrocyte endfeet, at synapses, and at the glia limitans, where it mediates water exchange across the blood-spinal cord and blood-brain barriers (BSCB/BBB), and controls cell volume, extracellular space volume, and astrocyte migration. Perivascular enrichment of AQP4 at the BSCB/BBB suggests a role in glymphatic function. Recently, we have demonstrated that AQP4 localization is also dynamically regulated at the subcellular level, affecting membrane water permeability. Ageing, cerebrovascular disease, traumatic CNS injury, and sleep disruption are established and emerging risk factors in developing neurodegeneration, and in animal models of each, impairment of glymphatic function is associated with changes in perivascular AQP4 localization. CNS oedema is caused by passive water influx through AQP4 in response to osmotic imbalances. We have demonstrated that reducing dynamic relocalization of AQP4 to the BSCB/BBB reduces CNS oedema and accelerates functional recovery in rodent models. Given the difficulties in developing pore-blocking AQP4 inhibitors, targeting AQP4 subcellular localization opens up new treatment avenues for CNS oedema, neurovascular and neurodegenerative diseases, and provides a framework to address fundamental questions about water homeostasis in health and disease.
Collapse
Affiliation(s)
- Mootaz M Salman
- Department of Physiology, Anatomy and Genetics,
University of Oxford, Oxford OX1 3PT, UK
| | - Philip Kitchen
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| | - Andrea Halsey
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Marie Xun Wang
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
| | | | - Alex C Conner
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Jerome Badaut
- CNRS-UMR 5536-Centre de Résonance
Magnétique des systèmes Biologiques, Université de
Bordeaux, 33076 Bordeaux, France
| | - Jeffrey J Iliff
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington
School of Medicine, Seattle, WA, USA
- VISN 20 Mental Illness Research, Education and
Clinical Center, VA Puget Sound Health Care System, Seattle, WA,
USA
| | - Roslyn M Bill
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| |
Collapse
|
224
|
Zamani A, Walker AK, Rollo B, Ayers KL, Farah R, O'Brien TJ, Wright DK. Impaired glymphatic function in the early stages of disease in a TDP-43 mouse model of amyotrophic lateral sclerosis. Transl Neurodegener 2022; 11:17. [PMID: 35287738 PMCID: PMC8922788 DOI: 10.1186/s40035-022-00291-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multiple lines of evidence suggest possible impairment of the glymphatic system in amyotrophic lateral sclerosis (ALS). To investigate this, we used in vivo magnetic resonance imaging (MRI) to assess glymphatic function early in the course of disease in a transgenic mouse with doxycycline (Dox)-controlled expression of cytoplasmic human TDP-43 (hTDP-43ΔNLS), mimicking the key pathology implicated in ALS. METHODS Adult TDP-43 transgenic and littermate monogenic control mice underwent longitudinal multimodal MRI one and three weeks after the cessation of Dox feed, together with weekly rotarod assessments of motor performance. Glymphatic function was assessed using dynamic contrast-enhanced MRI to track the clearance of an MR contrast agent injected into the cisterna magna. RESULTS Compared to their littermate controls, TDP-43 mice exhibited progressive neurodegeneration including that within the primary motor cortex, primary somatosensory cortex and corticospinal tract, significant weight loss including gastrocnemius atrophy, and shortened telomere length. Furthermore, in the presence of this ALS-like phenotype, these mice have significantly disrupted glymphatic function. CONCLUSIONS Although the relationship between glymphatic clearance and ALS disease progression remains to be elucidated, these changes occurred very early in the disease course. This provides initial evidence to suggest that the glymphatic system might be a potential therapeutic target in the treatment of ALS.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Adam K Walker
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Katie L Ayers
- The Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia.,Department of Pediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Raysha Farah
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
225
|
Blomqvist KJ, Skogster MOB, Kurkela MJ, Rosenholm MP, Ahlström FHG, Airavaara MT, Backman JT, Rauhala PV, Kalso EA, Lilius TO. Systemic hypertonic saline enhances glymphatic spinal cord delivery of lumbar intrathecal morphine. J Control Release 2022; 344:214-224. [PMID: 35301056 DOI: 10.1016/j.jconrel.2022.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier significantly limits effective drug delivery to central nervous system (CNS) targets. The recently characterized glymphatic system offers a perivascular highway for intrathecally (i.t.) administered drugs to reach deep brain structures. Although periarterial cerebrospinal fluid (CSF) influx and concomitant brain drug delivery can be enhanced by pharmacological or hyperosmotic interventions, their effects on drug delivery to the spinal cord, an important target for many drugs, have not been addressed. Hence, we studied in rats whether enhancement of periarterial flow by systemic hypertonic solution might be utilized to enhance spinal delivery and efficacy of i.t. morphine. We also studied whether the hyperosmolar intervention affects brain or cerebrospinal fluid drug concentrations after systemic administration. Periarterial CSF influx was enhanced by intraperitoneal injection of hypertonic saline (HTS, 5.8%, 20 ml/kg, 40 mOsm/kg). The antinociceptive effects of morphine were characterized, using tail flick, hot plate and paw pressure tests. Drug concentrations in serum, tissue and microdialysis samples were determined by liquid chromatography-tandem mass spectrometry. Compared with isotonic solution, HTS increased concentrations of spinal i.t. administered morphine by 240% at the administration level (T13-L1) at 60 min and increased the antinociceptive effect of morphine in tail flick, hot plate, and paw pressure tests. HTS also independently increased hot plate and paw pressure latencies but had no effect in the tail flick test. HTS transiently increased the penetration of intravenous morphine into the lateral ventricle, but not into the hippocampus. In conclusion, acute systemic hyperosmolality is a promising intervention for enhanced spinal delivery of i.t. administered morphine. The relevance of this intervention should be expanded to other i.t. drugs and brought to clinical trials.
Collapse
Affiliation(s)
- Kim J Blomqvist
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Moritz O B Skogster
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mika J Kurkela
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marko P Rosenholm
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik H G Ahlström
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko T Airavaara
- Faculty of Pharmacy and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Janne T Backman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pekka V Rauhala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eija A Kalso
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland; SleepWell Research Programme, Faculty of Medicine, University of Helsinki, Finland
| | - Tuomas O Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
226
|
Li L, Ding G, Zhang L, Davoodi-Bojd E, Chopp M, Li Q, Zhang ZG, Jiang Q. Aging-Related Alterations of Glymphatic Transport in Rat: In vivo Magnetic Resonance Imaging and Kinetic Study. Front Aging Neurosci 2022; 14:841798. [PMID: 35360203 PMCID: PMC8960847 DOI: 10.3389/fnagi.2022.841798] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Impaired glymphatic waste clearance function during brain aging leads to the accumulation of metabolic waste and neurotoxic proteins (e.g., amyloid-β, tau) which contribute to neurological disorders. However, how the age-related glymphatic dysfunction exerts its effects on different cerebral regions and affects brain waste clearance remain unclear. Methods We investigated alterations of glymphatic transport in the aged rat brain using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and advanced kinetic modeling. Healthy young (3-4 months) and aged (18-20 months) male rats (n = 12/group) underwent the identical MRI protocol, including T2-weighted imaging and 3D T1-weighted imaging with intracisternal administration of contrast agent (Gd-DTPA). Model-derived parameters of infusion rate and clearance rate, characterizing the kinetics of cerebrospinal fluid (CSF) tracer transport via the glymphatic system, were evaluated in multiple representative brain regions. Changes in the CSF-filled cerebral ventricles were measured using contrast-induced time signal curves (TSCs) in conjunction with structural imaging. Results Compared to the young brain, an overall impairment of glymphatic transport function was detected in the aged brain, evidenced by the decrease in both infusion and clearance rates throughout the brain. Enlarged ventricles in parallel with reduced efficiency in CSF transport through the ventricular regions were present in the aged brain. While the age-related glymphatic dysfunction was widespread, our kinetic quantification demonstrated that its impact differed considerably among cerebral regions with the most severe effect found in olfactory bulb, indicating the heterogeneous and regional preferential alterations of glymphatic function. Conclusion The robust suppression of glymphatic activity in the olfactory bulb, which serves as one of major efflux routes for brain waste clearance, may underlie, in part, age-related neurodegenerative diseases associated with neurotoxic substance accumulation. Our data provide new insight into the cerebral regional vulnerability to brain functional change with aging.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | | | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
227
|
Fang Y, Dai S, Jin C, Si X, Gu L, Song Z, Gao T, Chen Y, Yan Y, Yin X, Pu J, Zhang B. Aquaporin-4 Polymorphisms Are Associated With Cognitive Performance in Parkinson’s Disease. Front Aging Neurosci 2022; 13:740491. [PMID: 35356146 PMCID: PMC8959914 DOI: 10.3389/fnagi.2021.740491] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/04/2021] [Indexed: 12/29/2022] Open
Abstract
ObjectiveAquaporin-4 (AQP4) facilitates a sleep-enhanced interstitial brain waste clearance system. This study was conducted to determine the clinical implication of AQP4 polymorphisms in Parkinson’s disease (PD).MethodsThree-hundred and eighty-two patients with PD and 180 healthy controls with a mean follow-up time of 66.1 months from the Parkinson’s Progression Marker Initiative study were analyzed. We examined whether AQP4 SNPs were associated with an altered rate of motor or cognitive decline using linear mixed model and Cox regression. We then investigated whether AQP4 SNPs were associated with Aβ burden as measured by 18F Florbetapir standard uptake values. Furthermore, we examined if AQP4 SNPs moderated the association between REM sleep behavior disorder (RBD) and CSF biomarkers.ResultsIn patients with PD, AQP4 rs162009 (AA/AG vs. GG) was associated with slower dementia conversion, better performance in letter-number sequencing and symbol digit modalities, lower Aβ deposition in the putamen, anterior cingulum, and frontotemporal areas. In the subgroup of high RBD screening questionnaire score, rs162009 AA/AG had a higher CSF Aβ42 level. rs162009 AA/AG also had better performance in semantic fluency in healthy controls. Besides, rs68006382 (GG/GA vs. AA) was associated with faster progression to mild cognitive impairment, worse performance in letter-number sequencing, semantic fluency, and symbol digit modalities in patients with PD.InterpretationGenetic variations of AQP4 and subsequent alterations of glymphatic efficacy might contribute to an altered rate of cognitive decline in PD. AQP4 rs162009 is likely a novel genetic prognostic marker of glymphatic function and cognitive decline in PD.
Collapse
Affiliation(s)
- Yi Fang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shaobing Dai
- Department of Anesthesiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chongyao Jin
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoli Si
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luyan Gu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhe Song
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Gao
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yaping Yan
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinzhen Yin
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiali Pu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Baorong Zhang Jiali Pu
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Baorong Zhang Jiali Pu
| |
Collapse
|
228
|
von Gall C. The Effects of Light and the Circadian System on Rhythmic Brain Function. Int J Mol Sci 2022; 23:ijms23052778. [PMID: 35269920 PMCID: PMC8911243 DOI: 10.3390/ijms23052778] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Life on earth has evolved under the influence of regularly recurring changes in the environment, such as the 24 h light/dark cycle. Consequently, organisms have developed endogenous clocks, generating 24 h (circadian) rhythms that serve to anticipate these rhythmic changes. In addition to these circadian rhythms, which persist in constant conditions and can be entrained to environmental rhythms, light drives rhythmic behavior and brain function, especially in nocturnal laboratory rodents. In recent decades, research has made great advances in the elucidation of the molecular circadian clockwork and circadian light perception. This review summarizes the role of light and the circadian clock in rhythmic brain function, with a focus on the complex interaction between the different components of the mammalian circadian system. Furthermore, chronodisruption as a consequence of light at night, genetic manipulation, and neurodegenerative diseases is briefly discussed.
Collapse
Affiliation(s)
- Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, 40225 Dusseldorf, Germany
| |
Collapse
|
229
|
Choe KY, Bethlehem RAI, Safrin M, Dong H, Salman E, Li Y, Grinevich V, Golshani P, DeNardo LA, Peñagarikano O, Harris NG, Geschwind DH. Oxytocin normalizes altered circuit connectivity for social rescue of the Cntnap2 knockout mouse. Neuron 2022; 110:795-808.e6. [PMID: 34932941 PMCID: PMC8944915 DOI: 10.1016/j.neuron.2021.11.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/03/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
The neural basis of abnormal social behavior in autism spectrum disorders (ASDs) remains incompletely understood. Here we used two complementary but independent brain-wide mapping approaches, mouse resting-state fMRI and c-Fos-iDISCO+ imaging, to construct brain-wide activity and connectivity maps of the Cntnap2 knockout (KO) mouse model of ASD. At the macroscale level, we detected reduced functional coupling across social brain regions despite general patterns of hyperconnectivity across major brain structures. Oxytocin administration, which rescues social deficits in KO mice, strongly stimulated many brain areas and normalized connectivity patterns. Notably, chemogenetically triggered release of endogenous oxytocin strongly stimulated the nucleus accumbens (NAc), a forebrain nucleus implicated in social reward. Furthermore, NAc-targeted approaches to activate local oxytocin receptors sufficiently rescued their social deficits. Our findings establish circuit- and systems-level mechanisms of social deficits in Cntnap2 KO mice and reveal the NAc as a region that can be modulated by oxytocin to promote social interactions.
Collapse
Affiliation(s)
- Katrina Y Choe
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Martin Safrin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Hongmei Dong
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Elena Salman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Ying Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Valery Grinevich
- Department of Neuropeptide Research for Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Peyman Golshani
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Laura A DeNardo
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Vizcaya 48940, Spain
| | - Neil G Harris
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Daniel H Geschwind
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
230
|
The Bidirectional Link Between Sleep Disturbances and Traumatic Brain Injury Symptoms: A Role for Glymphatic Dysfunction? Biol Psychiatry 2022; 91:478-487. [PMID: 34481662 PMCID: PMC8758801 DOI: 10.1016/j.biopsych.2021.06.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022]
Abstract
Mild traumatic brain injury (mTBI), often referred to as concussion, is a major cause of morbidity and mortality worldwide. Sleep disturbances are common after mTBI. Moreover, subjects who develop subjective sleep complaints after mTBI also report more severe somatic, mental health, and cognitive impairment and take longer to recover from mTBI sequelae. Despite many previous studies addressing the role of sleep in post-mTBI morbidity, the mechanisms linking sleep to recovery after mTBI remain poorly understood. The glymphatic system is a brainwide network that supports fluid movement through the cerebral parenchyma and the clearance of interstitial solutes and wastes from the brain. Notably, the glymphatic system is active primarily during sleep. Clearance of cellular byproducts related to somatic, mental health, and neurodegenerative processes (e.g., amyloid-β and tau, among others) depends in part on intact glymphatic function, which becomes impaired after mTBI. In this viewpoint, we review the current knowledge regarding the association between sleep disturbances and post-mTBI symptoms. We also discuss the role of glymphatic dysfunction as a potential link between mTBI, sleep disruption, and posttraumatic morbidity. We outline a model where glymphatic dysfunction and sleep disruption caused by mTBI may have an additive effect on waste clearance, leading to cerebral dysfunction and impaired recovery. Finally, we review the novel techniques being developed to examine glymphatic function in humans and explore potential interventions to alter glymphatic exchange that may offer a novel therapeutic approach to those experiencing poor sleep and prolonged symptoms after mTBI.
Collapse
|
231
|
Chen X, Liu X, Koundal S, Elkin R, Zhu X, Monte B, Xu F, Dai F, Pedram M, Lee H, Kipnis J, Tannenbaum A, Van Nostrand WE, Benveniste H. Cerebral amyloid angiopathy is associated with glymphatic transport reduction and time-delayed solute drainage along the neck arteries. NATURE AGING 2022; 2:214-223. [PMID: 36199752 PMCID: PMC9531841 DOI: 10.1038/s43587-022-00181-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 01/19/2022] [Indexed: 01/27/2023]
Affiliation(s)
- Xinan Chen
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Rena Elkin
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaoyue Zhu
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Brittany Monte
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Feng Xu
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Feng Dai
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Maysam Pedram
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Allen Tannenbaum
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
- Department of Computer Science, Stony Brook University, Stony Brook, NY, USA
| | - William E Van Nostrand
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale School of Medicine New Haven, New Haven, CT, USA.
| |
Collapse
|
232
|
Siow TY, Toh CH, Hsu JL, Liu GH, Lee SH, Chen NH, Fu CJ, Castillo M, Fang JT. Association of Sleep, Neuropsychological Performance, and Gray Matter Volume With Glymphatic Function in Community-Dwelling Older Adults. Neurology 2022; 98:e829-e838. [PMID: 34906982 DOI: 10.1212/wnl.0000000000013215] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 12/02/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The glymphatic system, which is robustly enabled during some stages of sleep, is a fluid-transport pathway that clears cerebral waste products. Most contemporary knowledge regarding the glymphatic system is inferred from rodent experiments and human research is limited. Our objective is to explore the associations between human glymphatic function, sleep, neuropsychological performance, and cerebral gray matter volumes. METHODS This cross-sectional study included individuals 60 years or older who had participated in the Integrating Systemic Data of Geriatric Medicine to Explore the Solution for Health Aging study between September 2019 and October 2020. Community-dwelling older adults were enrolled at 2 different sites. Participants with dementia, major depressive disorders, and other major organ system abnormalities were excluded. Sleep profile was accessed using questionnaires and polysomnography. Administered neuropsychological test batteries included Everyday Cognition (ECog) and the Consortium to Establish a Registry for Alzheimer's Disease Neuropsychological Battery (CERAD-NB). Gray matter volumes were estimated based on MRI. Diffusion tensor imaging analysis along the perivascular space (DTI-ALPS) index was used as the MRI marker of glymphatic function. RESULTS A total of 84 participants (mean [SD] age 73.3 [7.1] years, 47 [56.0%] women) were analyzed. Multivariate linear regression model determined that age (unstandardized β, -0.0025 [SE 0.0001]; p = 0.02), N2 sleep duration (unstandardized β, 0.0002 [SE 0.0001]; p = 0.04), and the apnea-hypopnea index (unstandardized β, -0.0011 [SE 0.0005]; p = 0.03) were independently associated with DTI-ALPS. Higher DTI-ALPS was associated with better ECog language scores (unstandardized β, -0.59 [SE 0.28]; p = 0.04) and better CERAD-NB word list learning delayed recall subtest scores (unstandardized β, 6.17 [SE 2.31]; p = 0.009) after covarying for age and education. Higher DTI-ALPS was also associated with higher gray matter volume (unstandardized β, 107.00 [SE 43.65]; p = 0.02) after controlling for age, sex, and total intracranial volume. DISCUSSION Significant associations were identified between glymphatic function and sleep, stressing the importance of sleep for brain health. This study also revealed associations between DTI-ALPS, neuropsychological performance, and cerebral gray matter volumes, suggesting the potential of DTI-ALPS as a biomarker for cognitive disorders.
Collapse
Affiliation(s)
- Tiing Yee Siow
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Cheng Hong Toh
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill.
| | - Jung-Lung Hsu
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Geng-Hao Liu
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Shwu-Hua Lee
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Ning-Hung Chen
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Changjui James Fu
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Mauricio Castillo
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| | - Ji-Tseng Fang
- From the Department of Medical Imaging and Intervention (T.Y.S., C.H.T.) and Neuroscience Research Center, Department of Neurology, Medical Center and College of Medicine (J.-L.H.), Chang Gung University College of Medicine, and Division of Acupuncture and Moxibustion, Department of Traditional Chinese Medicine (G.-H.L.), Department of Psychiatry (S.-H.L.), Department of Pulmonary and Critical Care Medicine (N.-H.C.), Biomedical Informatics Unit, Clinical Trial Center (C.J.F.), and Department of Nephrology (J.-T.F.), Chang Gung Memorial Hospital at Linkou, Taoyuan; Graduate Institute of Mind, Brain, & Consciousness (J.-L.H.), Taipei Medical University; Brain & Consciousness Research Center (J.-L.H.), Shuang Ho Hospital, New Taipei City; School of Traditional Chinese Medicine (G.-H.L., N.-H.C.) and School of Medicine (J.-T.F.), College of Medicine (S.-H.L.), Chang Gung University; Sleep Center (G.-H.L., N.-H.C.), Chang Gung Memorial Hospital, Taoyuan, Taiwan; and Department of Radiology (M.C.), University of North Carolina School of Medicine, Chapel Hill
| |
Collapse
|
233
|
DiNuzzo M, Mangia S, Giove F. Manipulations of sleep‐like slow‐wave activity by noninvasive brain stimulation. J Neurosci Res 2022; 100:1218-1225. [DOI: 10.1002/jnr.25029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Mauro DiNuzzo
- Magnetic Resonance for Brain Investigation Laboratory Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Rome Italy
| | - Silvia Mangia
- Center for Magnetic Resonance Research, Department of Radiology University of Minnesota Minneapolis Minnesota USA
| | - Federico Giove
- Magnetic Resonance for Brain Investigation Laboratory Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Rome Italy
- Laboratory of Neurophysics and Neuroimaging Fondazione Santa Lucia IRCCS Rome Italy
| |
Collapse
|
234
|
Hladky SB, Barrand MA. The glymphatic hypothesis: the theory and the evidence. Fluids Barriers CNS 2022; 19:9. [PMID: 35115036 PMCID: PMC8815211 DOI: 10.1186/s12987-021-00282-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
The glymphatic hypothesis proposes a mechanism for extravascular transport into and out of the brain of hydrophilic solutes unable to cross the blood-brain barrier. It suggests that there is a circulation of fluid carrying solutes inwards via periarterial routes, through the interstitium and outwards via perivenous routes. This review critically analyses the evidence surrounding the mechanisms involved in each of these stages. There is good evidence that both influx and efflux of solutes occur along periarterial routes but no evidence that the principal route of outflow is perivenous. Furthermore, periarterial inflow of fluid is unlikely to be adequate to provide the outflow that would be needed to account for solute efflux. A tenet of the hypothesis is that flow sweeps solutes through the parenchyma. However, the velocity of any possible circulatory flow within the interstitium is too small compared to diffusion to provide effective solute movement. By comparison the earlier classical hypothesis describing extravascular transport proposed fluid entry into the parenchyma across the blood-brain barrier, solute movements within the parenchyma by diffusion, and solute efflux partly by diffusion near brain surfaces and partly carried by flow along "preferred routes" including perivascular spaces, white matter tracts and subependymal spaces. It did not suggest fluid entry via periarterial routes. Evidence is still incomplete concerning the routes and fate of solutes leaving the brain. A large proportion of the solutes eliminated from the parenchyma go to lymph nodes before reaching blood but the proportions delivered directly to lymph or indirectly via CSF which then enters lymph are as yet unclear. In addition, still not understood is why and how the absence of AQP4 which is normally highly expressed on glial endfeet lining periarterial and perivenous routes reduces rates of solute elimination from the parenchyma and of solute delivery to it from remote sites of injection. Neither the glymphatic hypothesis nor the earlier classical hypothesis adequately explain how solutes and fluid move into, through and out of the brain parenchyma. Features of a more complete description are discussed. All aspects of extravascular transport require further study.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
235
|
Chong PLH, Garic D, Shen MD, Lundgaard I, Schwichtenberg AJ. Sleep, cerebrospinal fluid, and the glymphatic system: A systematic review. Sleep Med Rev 2022; 61:101572. [PMID: 34902819 PMCID: PMC8821419 DOI: 10.1016/j.smrv.2021.101572] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 10/14/2021] [Accepted: 11/10/2021] [Indexed: 02/03/2023]
Abstract
Current theories of the glymphatic system (GS) hypothesize that it relies on cerebrospinal fluid (CSF) circulation to disseminate growth factors and remove metabolic waste from the brain with increased CSF production and circulation during sleep; thereby, linking sleep disturbance with elements of CSF circulation and GS exchange. However, our growing knowledge of the relations between sleep, CSF, and the GS are plagued by variability in sleep and CSF measures across a wide array of pathologies. Hence, this review aims to summarize the dynamic relationships between sleep, CSF-, and GS-related features in samples of typically developing individuals and those with autoimmune/inflammatory, neurodegenerative, neurodevelopmental, sleep-related, neurotraumatic, neuropsychiatric, and skull atypicalities. One hundred and ninety articles (total n = 19,129 participants) were identified and reviewed for pathology, CSF circulation and related metrics, GS function, and sleep. Numerous associations were documented between sleep problems and CSF metabolite concentrations (e.g., amyloid-beta, orexin, tau proteins) and increased CSF volumes or pressure. However, these relations were not universal, with marked differences across pathologies. It is clear that elements of CSF circulation/composition and GS exchange represent pathways influenced by sleep; however, carefully designed studies and advances in GS measurement are needed to delineate the nuanced relationships.
Collapse
Affiliation(s)
| | - D. Garic
- University of North Carolina, Chapel Hill, NC
| | - M. D. Shen
- University of North Carolina, Chapel Hill, NC
| | - I. Lundgaard
- Department of Experimental Medicine Science, Lund University, Lund, Sweden,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | | |
Collapse
|
236
|
Wang K, Zhang Y, Zhu Y, Luo Y. Associations between cortical activation and network interaction during sleep. Behav Brain Res 2022; 422:113751. [PMID: 35038462 DOI: 10.1016/j.bbr.2022.113751] [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: 10/23/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 11/02/2022]
Abstract
Cortical activation and network interaction, two characterizations of the cortical states, are separately studied in most previous studies. To further clarify the underlying mechanism, the association between these two indicators during sleep was investigated in this study. Twenty healthy individuals were enrolled and all of them underwent overnight polysomnography (PSG) recording. The relative spectral powers and the phase transfer entropy (PTE) of various frequency components were extracted from 6 electroencephalographic (EEG) channels, to assess the cortical activation and network interaction, respectively. Pearson correlation coefficient was employed to estimate their associations. The results suggested that there was a negative correlation between spectral power and phase transfer entropy in δ and α frequency bands during sleep. As the sleep deepened, an increased negative correlation in the δ frequency band was noted, but the negative correlation became less extreme in the α frequency band. The extremum of the correlation coefficient was noted in δ of N3, and α of Wake. Overall, this study provides a connection between these two cortical activity assessments, especially reveals the variable characteristics of different frequency components, which is conducive to better understand sleep state.
Collapse
Affiliation(s)
- Kejie Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Yangting Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Yongpeng Zhu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Yuxi Luo
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
237
|
Xu X, Mu L, Li L, Liang J, Zhang S, Jia L, Yang X, Dai Y, Zhang J, Wang Y, Niu S, Xia G, Yang Y, Zhang Y, Cao Y, Zhang H. Imaging and tracing the pattern of adult ovarian angiogenesis implies a strategy against female reproductive aging. SCIENCE ADVANCES 2022; 8:eabi8683. [PMID: 35020427 PMCID: PMC8754302 DOI: 10.1126/sciadv.abi8683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Robust angiogenesis is continuously active in ovaries to remodel the ovary-body connections in mammals, but understanding of this unique process remains elusive. Here, we performed high-resolution, three-dimensional ovarian vascular imaging and traced the pattern of ovarian angiogenesis and vascular development in the long term. We found that angiogenesis was mainly active on ovarian follicles and corpus luteum and that robust angiogenesis constructs independent but temporary vascular networks for each follicle. Based on the pattern of ovarian angiogenesis, we designed an angiogenesis-blocking strategy by axitinib administration to young females, and we found that the temporary suppression of angiogenesis paused ovarian development and kept the ovarian reserve in the long term, leading to postponed ovarian senescence and an extension of the female reproductive life span. Together, by uncovering the detailed model of physiological ovarian angiogenesis, our experiments suggest a potential approach to delay female reproductive aging through the manipulation of angiogenesis.
Collapse
Affiliation(s)
- Xueqiang Xu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lu Mu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lingyu Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jing Liang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Longzhong Jia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xuebing Yang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yanli Dai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiawei Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yibo Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shudong Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yan Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Corresponding author.
| |
Collapse
|
238
|
Asghar A, Naaz S. Does modulation of glymphatic system reduce delirium via waste clearance? J Anaesthesiol Clin Pharmacol 2022; 38:164-165. [PMID: 35706625 PMCID: PMC9191798 DOI: 10.4103/joacp.joacp_337_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/24/2020] [Indexed: 11/23/2022] Open
|
239
|
Szczygielski J, Kopańska M, Wysocka A, Oertel J. Cerebral Microcirculation, Perivascular Unit, and Glymphatic System: Role of Aquaporin-4 as the Gatekeeper for Water Homeostasis. Front Neurol 2021; 12:767470. [PMID: 34966347 PMCID: PMC8710539 DOI: 10.3389/fneur.2021.767470] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
In the past, water homeostasis of the brain was understood as a certain quantitative equilibrium of water content between intravascular, interstitial, and intracellular spaces governed mostly by hydrostatic effects i.e., strictly by physical laws. The recent achievements in molecular bioscience have led to substantial changes in this regard. Some new concepts elaborate the idea that all compartments involved in cerebral fluid homeostasis create a functional continuum with an active and precise regulation of fluid exchange between them rather than only serving as separate fluid receptacles with mere passive diffusion mechanisms, based on hydrostatic pressure. According to these concepts, aquaporin-4 (AQP4) plays the central role in cerebral fluid homeostasis, acting as a water channel protein. The AQP4 not only enables water permeability through the blood-brain barrier but also regulates water exchange between perivascular spaces and the rest of the glymphatic system, described as pan-cerebral fluid pathway interlacing macroscopic cerebrospinal fluid (CSF) spaces with the interstitial fluid of brain tissue. With regards to this, AQP4 makes water shift strongly dependent on active processes including changes in cerebral microcirculation and autoregulation of brain vessels capacity. In this paper, the role of the AQP4 as the gatekeeper, regulating the water exchange between intracellular space, glymphatic system (including the so-called neurovascular units), and intravascular compartment is reviewed. In addition, the new concepts of brain edema as a misbalance in water homeostasis are critically appraised based on the newly described role of AQP4 for fluid permeation. Finally, the relevance of these hypotheses for clinical conditions (including brain trauma and stroke) and for both new and old therapy concepts are analyzed.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland.,Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland
| | - Anna Wysocka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, Lublin, Poland
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
240
|
Glymphatic system dysfunction in obstructive sleep apnea evidenced by DTI-ALPS. Sleep Med 2021; 89:176-181. [PMID: 35030357 DOI: 10.1016/j.sleep.2021.12.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/13/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study aimed to evaluate the glymphatic system function in patients with obstructive sleep apnea (OSA) compared to healthy controls using diffusion tensor imaging (DTI) with the perivascular space (DTI-ALPS) method. Our hypothesis is that patients with OSA may have glymphatic system dysfunction, which is correlated with OSA severity. METHODS We enrolled 24 patients with OSA and 24 healthy controls. All participants underwent DTI magnetic resonance imaging (MRI) using the same 3T MRI scanner, and we calculated the DTI-ALPS index from the DTI. We evaluated the differences in the DTI-ALPS index between patients with OSA and healthy controls. In addition, we conducted a correlation analysis between the DTI-ALPS index and clinical characteristics. RESULTS The DTI-ALPS index was significantly different between the groups. The DTI-ALPS in patients with OSA was significantly lower than in healthy controls (1.30450 vs. 1.61600, p = 0.0006). Furthermore, the DTI-ALPS index was significantly negatively correlated with the apnea-hypopnea index in sleep stage N (r = -0.427, p = 0.042) and oxygen desaturation index during sleep N (r = -0.497, p = 0.036). CONCLUSION We successfully demonstrated glymphatic system dysfunction in patients with OSA. In addition, glymphatic system dysfunction is well correlated with OSA severity, especially during sleep stage N. Thus, these findings can explain the effects of OSA on increased risk of developing dementia and highlight the importance of OSA treatment.
Collapse
|
241
|
Decker Y, Krämer J, Xin L, Müller A, Scheller A, Fassbender K, Proulx ST. Magnetic resonance imaging of cerebrospinal fluid outflow after low-rate lateral ventricle infusion in mice. JCI Insight 2021; 7:150881. [PMID: 34905509 PMCID: PMC8855808 DOI: 10.1172/jci.insight.150881] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The anatomical routes for the clearance of cerebrospinal fluid (CSF) remain incompletely understood. However, recent evidence has given strong support for routes leading to lymphatic vessels. A current debate centers upon the routes through which CSF can access lymphatics, with evidence emerging for either direct routes to meningeal lymphatics or along cranial nerves to reach lymphatics outside the skull. Here, a method was established to infuse contrast agent into the ventricles using indwelling cannulae during imaging of mice at 2 and 12 months of age by magnetic resonance imaging. As expected, a significant decline in overall CSF turnover was found with aging. Quantifications demonstrated that the bulk of the contrast agent flowed from the ventricles to the subarachnoid space in the basal cisterns. Comparatively little contrast agent signal was found at the dorsal aspect of the skull. The imaging dynamics from the two cohorts revealed that the contrast agent cleared from the cranium through the cribriform plate to the nasopharyngeal lymphatics. On decalcified sections, we confirmed that fluorescentlylabeled ovalbumin drains through the cribriform plate and can be found within lymphatics surrounding the nasopharynx. In conclusion, routes leading to nasopharyngeal lymphatics appear to be a major efflux pathway for cranial CSF.
Collapse
Affiliation(s)
- Yann Decker
- Department of Neurology, Saarland University, Homburg, Germany
| | - Jonas Krämer
- Department of Neurology, Saarland University, Homburg, Germany
| | - Li Xin
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Andreas Müller
- Clinic for Diagnostic and Interventional Radiology, Saarland University, Homburg, Germany
| | - Anja Scheller
- Department of Physiology, Saarland University, Homburg, Germany
| | | | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| |
Collapse
|
242
|
Carlstrom LP, Eltanahy A, Perry A, Rabinstein AA, Elder BD, Morris JM, Meyer FB, Graffeo CS, Lundgaard I, Burns TC. A clinical primer for the glymphatic system. Brain 2021; 145:843-857. [PMID: 34888633 DOI: 10.1093/brain/awab428] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/02/2021] [Accepted: 11/07/2021] [Indexed: 11/14/2022] Open
Abstract
The complex and dynamic system of fluid flow through the perivascular and interstitial spaces of the central nervous system has new-found implications for neurological diseases. Cerebrospinal fluid movement throughout the CNS parenchyma is more dynamic than could be explained via passive diffusion mechanisms alone. Indeed, a semi-structured glial-lymphatic (glymphatic) system of astrocyte-supported extracellular perivascular channels serves to directionally channel extracellular fluid, clearing metabolites and peptides to optimize neurologic function. Clinical studies of the glymphatic network has to date proven challenging, with most data gleaned from rodent models and post-mortem investigations. However, increasing evidence suggests that disordered glymphatic function contributes to the pathophysiology of CNS aging, neurodegenerative disease, and CNS injuries, as well as normal pressure hydrocephalus. Unlocking such pathophysiology could provide important avenues toward novel therapeutics. We here provide a multidisciplinary overview of glymphatics and critically review accumulating evidence regarding its structure, function, and hypothesized relevance to neurological disease. We highlight emerging technologies of relevance to the longitudinal evaluation of glymphatic function in health and disease. Finally, we discuss the translational opportunities and challenges of studying glymphatic science.
Collapse
Affiliation(s)
- Lucas P Carlstrom
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Ahmed Eltanahy
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Avital Perry
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Benjamin D Elder
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Fredric B Meyer
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Iben Lundgaard
- Departments of Experimental Medical Science, Lund University, Lund 228 11 Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund 228 11 Sweden
| | - Terry C Burns
- Departments of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
243
|
Morawska MM, Moreira CG, Ginde VR, Valko PO, Weiss T, Büchele F, Imbach LL, Masneuf S, Kollarik S, Prymaczok N, Gerez JA, Riek R, Baumann CR, Noain D. Slow-wave sleep affects synucleinopathy and regulates proteostatic processes in mouse models of Parkinson's disease. Sci Transl Med 2021; 13:eabe7099. [PMID: 34878820 DOI: 10.1126/scitranslmed.abe7099] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marta M Morawska
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Carlos G Moreira
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,ETH Zurich, Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Varun R Ginde
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Philipp O Valko
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Fabian Büchele
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Lukas L Imbach
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Sophie Masneuf
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Sedef Kollarik
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Natalia Prymaczok
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Juan A Gerez
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Roland Riek
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland.,Center of Competence Sleep and Health Zurich, University of Zurich, Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland.,Center of Competence Sleep and Health Zurich, University of Zurich, Frauenklinikstrasse 26, Zurich 8091, Switzerland
| |
Collapse
|
244
|
Ray LA, Pike M, Simon M, Iliff JJ, Heys JJ. Quantitative analysis of macroscopic solute transport in the murine brain. Fluids Barriers CNS 2021; 18:55. [PMID: 34876169 PMCID: PMC8650464 DOI: 10.1186/s12987-021-00290-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Understanding molecular transport in the brain is critical to care and prevention of neurological disease and injury. A key question is whether transport occurs primarily by diffusion, or also by convection or dispersion. Dynamic contrast-enhanced (DCE-MRI) experiments have long reported solute transport in the brain that appears to be faster than diffusion alone, but this transport rate has not been quantified to a physically relevant value that can be compared to known diffusive rates of tracers. METHODS In this work, DCE-MRI experimental data is analyzed using subject-specific finite-element models to quantify transport in different anatomical regions across the whole mouse brain. The set of regional effective diffusivities ([Formula: see text]), a transport parameter combining all mechanisms of transport, that best represent the experimental data are determined and compared to apparent diffusivity ([Formula: see text]), the known rate of diffusion through brain tissue, to draw conclusions about dominant transport mechanisms in each region. RESULTS In the perivascular regions of major arteries, [Formula: see text] for gadoteridol (550 Da) was over 10,000 times greater than [Formula: see text]. In the brain tissue, constituting interstitial space and the perivascular space of smaller blood vessels, [Formula: see text] was 10-25 times greater than [Formula: see text]. CONCLUSIONS The analysis concludes that convection is present throughout the brain. Convection is dominant in the perivascular space of major surface and branching arteries (Pe > 1000) and significant to large molecules (> 1 kDa) in the combined interstitial space and perivascular space of smaller vessels (not resolved by DCE-MRI). Importantly, this work supports perivascular convection along penetrating blood vessels.
Collapse
Affiliation(s)
- Lori A Ray
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA
| | - Martin Pike
- Advanced Imaging Research Center, Oregon Health and Sciences University, Portland, USA
| | - Matthew Simon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, USA
- Neuroscience Graduate Program, Oregon Health and Science University, Portland, USA
- Denali Therapeutics, San Francisco, USA
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, USA
| | - Jeffrey J Heys
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA.
| |
Collapse
|
245
|
Semyachkina-Glushkovskaya O, Penzel T, Blokhina I, Khorovodov A, Fedosov I, Yu T, Karandin G, Evsukova A, Elovenko D, Adushkina V, Shirokov A, Dubrovskii A, Terskov A, Navolokin N, Tzoy M, Ageev V, Agranovich I, Telnova V, Tsven A, Kurths J. Night Photostimulation of Clearance of Beta-Amyloid from Mouse Brain: New Strategies in Preventing Alzheimer's Disease. Cells 2021; 10:3289. [PMID: 34943796 PMCID: PMC8699220 DOI: 10.3390/cells10123289] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022] Open
Abstract
The deposition of amyloid-β (Aβ) in the brain is a risk factor for Alzheimer's disease (AD). Therefore, new strategies for the stimulation of Aβ clearance from the brain can be useful in preventing AD. Transcranial photostimulation (PS) is considered a promising method for AD therapy. In our previous studies, we clearly demonstrated the PS-mediated stimulation of lymphatic clearing functions, including Aβ removal from the brain. There is increasing evidence that sleep plays an important role in Aβ clearance. Here, we tested our hypothesis that PS at night can stimulate Aβ clearance from the brain more effectively than PS during the day. Our results on healthy mice show that Aβ clearance from the brain occurs faster at night than during wakefulness. The PS course at night improves memory and reduces Aβ accumulation in the brain of AD mice more effectively than the PS course during the day. Our results suggest that night PS is a more promising candidate as an effective method in preventing AD than daytime PS. These data are an important informative platform for the development of new noninvasive and nonpharmacological technologies for AD therapy as well as for preventing Aβ accumulation in the brain of people with disorder of Aβ metabolism, sleep deficit, elderly age, and jet lag.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Inna Blokhina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Alexander Khorovodov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Ivan Fedosov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China;
- Collaborative Innovation Center for Biomedical Engineering, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Georgy Karandin
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Arina Evsukova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Dariya Elovenko
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Viktoria Adushkina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Alexander Shirokov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Institute of Biochemistry and Physiology of Plants and Microorganisms, Prospekt Entuziastov 13, 410049 Saratov, Russia
| | - Alexander Dubrovskii
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Andrey Terskov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Nikita Navolokin
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Department of Pathological Anatomy, Saratov Medical State University, Kazachaya 112, 410012 Saratov, Russia
| | - Maria Tzoy
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Ilana Agranovich
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Valeria Telnova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Anna Tsven
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Department of Complexity Science, Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
246
|
Jagirdar R, Fu CH, Park J, Corbett BF, Seibt FM, Beierlein M, Chin J. Restoring activity in the thalamic reticular nucleus improves sleep architecture and reduces Aβ accumulation in mice. Sci Transl Med 2021; 13:eabh4284. [PMID: 34731016 PMCID: PMC8985235 DOI: 10.1126/scitranslmed.abh4284] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sleep disruptions promote increases of amyloid β (Aβ) and tau in the brain and increase Alzheimer’s disease (AD) risk, but the precise mechanisms that give rise to sleep disturbances have yet to be defined. The thalamic reticular nucleus (TRN) is essential for sleep maintenance and for the regulation of slow-wave sleep (SWS). We examined the TRN in transgenic mice that express mutant human amyloid precursor protein (APP) and found reduced neuronal activity, increased sleep fragmentation, and decreased SWS time as compared to nontransgenic littermates. Selective activation of the TRN using excitatory DREADDs restored sleep maintenance, increased time in SWS, and reduced amyloid plaque load in both hippocampus and cortex. Our findings suggest that the TRN may play a major role in symptoms associated with AD. Enhancing TRN activity might be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Rohan Jagirdar
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Chia-Hsuan Fu
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Jin Park
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Brian F. Corbett
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Frederik M. Seibt
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Michael Beierlein
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Jeannie Chin
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
247
|
Cai X, Harding IC, Sadaka AH, Colarusso B, Kulkarni P, Ebong E, Qiao J, O'Hare NR, Ferris CF. Mild repetitive head impacts alter perivascular flow in the midbrain dopaminergic system in awake rats. Brain Commun 2021; 3:fcab265. [PMID: 34806002 PMCID: PMC8600963 DOI: 10.1093/braincomms/fcab265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
Head injury is a known risk factor for Parkinson's disease. Disruption in the perivascular clearance of metabolic waste and unwanted proteins is thought to be a contributing factor to disease progression. We hypothesized that repetitive mild head impacts, without evidence of structural brain damage, would increase microgliosis and AQP4 expression and depolarization and alter perivascular flow in the midbrain dopaminergic system. Adult male rats were subjected to sham, or two mild head impacts separated by 48 h. Three weeks later, fully awake rats were imaged using dynamic, contrast-enhanced MRI to follow the distribution of intraventricular gadobenate dimeglumine contrast agent. Images were registered to and analysed using a 3D MRI rat atlas providing site-specific data on 171 different brain areas. Following imaging, rats were tested for cognitive function using the Barnes maze assay. Histological analyses of tyrosine hydroxylase, microglia activation and AQP4 expression and polarization were performed on a parallel cohort of head impacted rats at 20 days post insult to coordinate with the time of imaging. There was no change in the global flux of contrast agent between sham and head impacted rats. The midbrain dopaminergic system showed a significant decrease in the influx of contrast agent as compared to sham controls together with a significant increase in microgliosis, AQP4 expression and depolarization. There were no deficits in cognitive function. The histology showed a significant level of neuroinflammation in the midbrain dopaminergic system 3 weeks post mild repetitive head impact but no loss in tyrosine hydroxylase. MRI revealed no structural brain damage emphasizing the potential serious consequences of mild head impacts on sustained brain neuroinflammation in this area critical to the pathophysiology of Parkinson's.
Collapse
Affiliation(s)
- Xuezhu Cai
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Ian C Harding
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Aymen H Sadaka
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Bradley Colarusso
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Praveen Kulkarni
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Eno Ebong
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ju Qiao
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Nick R O'Hare
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Craig F Ferris
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
- Department of Psychology, Northeastern University, Boston, MA 02115, USA
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
248
|
Cross NE, Pomares FB, Nguyen A, Perrault AA, Jegou A, Uji M, Lee K, Razavipour F, Ali OBK, Aydin U, Benali H, Grova C, Dang-Vu TT. An altered balance of integrated and segregated brain activity is a marker of cognitive deficits following sleep deprivation. PLoS Biol 2021; 19:e3001232. [PMID: 34735431 PMCID: PMC8568176 DOI: 10.1371/journal.pbio.3001232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/28/2021] [Indexed: 11/19/2022] Open
Abstract
Sleep deprivation (SD) leads to impairments in cognitive function. Here, we tested the hypothesis that cognitive changes in the sleep-deprived brain can be explained by information processing within and between large-scale cortical networks. We acquired functional magnetic resonance imaging (fMRI) scans of 20 healthy volunteers during attention and executive tasks following a regular night of sleep, a night of SD, and a recovery nap containing nonrapid eye movement (NREM) sleep. Overall, SD was associated with increased cortex-wide functional integration, driven by a rise of integration within cortical networks. The ratio of within versus between network integration in the cortex increased further in the recovery nap, suggesting that prolonged wakefulness drives the cortex towards a state resembling sleep. This balance of integration and segregation in the sleep-deprived state was tightly associated with deficits in cognitive performance. This was a distinct and better marker of cognitive impairment than conventional indicators of homeostatic sleep pressure, as well as the pronounced thalamocortical connectivity changes that occurs towards falling asleep. Importantly, restoration of the balance between segregation and integration of cortical activity was also related to performance recovery after the nap, demonstrating a bidirectional effect. These results demonstrate that intra- and interindividual differences in cortical network integration and segregation during task performance may play a critical role in vulnerability to cognitive impairment in the sleep-deprived state.
Collapse
Affiliation(s)
- Nathan E. Cross
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Institut Universitaire de Gériatrie de Montréal and CRIUGM, CIUSSS du Centre-Sud-de-l’Île-de-Montréal, Montreal, Canada
| | - Florence B. Pomares
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Institut Universitaire de Gériatrie de Montréal and CRIUGM, CIUSSS du Centre-Sud-de-l’Île-de-Montréal, Montreal, Canada
| | - Alex Nguyen
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
| | - Aurore A. Perrault
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Institut Universitaire de Gériatrie de Montréal and CRIUGM, CIUSSS du Centre-Sud-de-l’Île-de-Montréal, Montreal, Canada
| | - Aude Jegou
- PERFORM Centre, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
| | - Makoto Uji
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
| | - Kangjoo Lee
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Biomedical Engineering Department, Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Fatemeh Razavipour
- PERFORM Centre, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Biomedical Engineering Department, Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada
| | - Obaï Bin Ka’b Ali
- PERFORM Centre, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Biomedical Engineering Department, Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada
| | - Umit Aydin
- PERFORM Centre, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Biomedical Engineering Department, Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Habib Benali
- PERFORM Centre, Concordia University, Montreal, Canada
| | - Christophe Grova
- PERFORM Centre, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Department of Physics, Concordia University, Montreal, Canada
- Multimodal Functional Imaging Lab, Biomedical Engineering Department, Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada
| | - Thien Thanh Dang-Vu
- PERFORM Centre, Concordia University, Montreal, Canada
- Center for Studies in Behavioral Neurobiology, Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Institut Universitaire de Gériatrie de Montréal and CRIUGM, CIUSSS du Centre-Sud-de-l’Île-de-Montréal, Montreal, Canada
| |
Collapse
|
249
|
Abstract
Sleep is essential for brain function in a surprisingly diverse set of ways. In the short term, lack of sleep leads to impaired memory and attention; in the longer term, it produces neurological dysfunction or even death. I discuss recent advances in understanding how sleep maintains the physiological health of the brain through interconnected systems of neuronal activity and fluid flow. The neural dynamics that appear during sleep are intrinsically coupled to its consequences for blood flow, cerebrospinal fluid dynamics, and waste clearance. Recognizing these linked causes and consequences of sleep has shed new light on why sleep is important for such disparate aspects of brain function.
Collapse
Affiliation(s)
- Laura D Lewis
- Department of Biomedical Engineering, Boston University, Boston, MA, USA, and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
250
|
Cao J, Yao D, Li R, Guo X, Hao J, Xie M, Li J, Pan D, Luo X, Yu Z, Wang M, Wang W. Digoxin Ameliorates Glymphatic Transport and Cognitive Impairment in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurosci Bull 2021; 38:181-199. [PMID: 34704235 PMCID: PMC8821764 DOI: 10.1007/s12264-021-00772-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/04/2021] [Indexed: 02/03/2023] Open
Abstract
The glymphatic system plays a pivotal role in maintaining cerebral homeostasis. Chronic cerebral hypoperfusion, arising from small vessel disease or carotid stenosis, results in cerebrometabolic disturbances ultimately manifesting in white matter injury and cognitive dysfunction. However, whether the glymphatic system serves as a potential therapeutic target for white matter injury and cognitive decline during hypoperfusion remains unknown. Here, we established a mouse model of chronic cerebral hypoperfusion via bilateral common carotid artery stenosis. We found that the hypoperfusion model was associated with significant white matter injury and initial cognitive impairment in conjunction with impaired glymphatic system function. The glymphatic dysfunction was associated with altered cerebral perfusion and loss of aquaporin 4 polarization. Treatment of digoxin rescued changes in glymphatic transport, white matter structure, and cognitive function. Suppression of glymphatic functions by treatment with the AQP4 inhibitor TGN-020 abolished this protective effect of digoxin from hypoperfusion injury. Our research yields new insight into the relationship between hemodynamics, glymphatic transport, white matter injury, and cognitive changes after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Jie Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Rong Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xuequn Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Respiratory Medicine, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000 China
| | - Jiahuan Hao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jia Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Dengji Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|