1
|
Tong R, Su S, Liang Y, Li C, Sun L, Zhang X. Functional Connectivity Encodes Sound Locations by Lateralization Angles. Neurosci Bull 2024:10.1007/s12264-024-01312-0. [PMID: 39470972 DOI: 10.1007/s12264-024-01312-0] [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: 01/12/2024] [Accepted: 06/16/2024] [Indexed: 11/01/2024] Open
Abstract
The ability to localize sound sources rapidly allows human beings to efficiently understand the surrounding environment. Previous studies have suggested that there is an auditory "where" pathway in the cortex for processing sound locations. The neural activation in regions along this pathway encodes sound locations by opponent hemifield coding, in which each unilateral region is activated by sounds coming from the contralateral hemifield. However, it is still unclear how these regions interact with each other to form a unified representation of the auditory space. In the present study, we investigated whether functional connectivity in the auditory "where" pathway encoded sound locations during passive listening. Participants underwent functional magnetic resonance imaging while passively listening to sounds from five distinct horizontal locations (-90°, -45°, 0°, 45°, 90°). We were able to decode sound locations from the functional connectivity patterns of the "where" pathway. Furthermore, we found that such neural representation of sound locations was primarily based on the coding of sound lateralization angles to the frontal midline. In addition, whole-brain analysis indicated that functional connectivity between occipital regions and the primary auditory cortex also encoded sound locations by lateralization angles. Overall, our results reveal a lateralization-angle-based representation of sound locations encoded by functional connectivity patterns, which could add on the activation-based opponent hemifield coding to provide a more precise representation of the auditory space.
Collapse
Affiliation(s)
- Renjie Tong
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Beijing, 100069, China
| | - Shaoyi Su
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Ying Liang
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Beijing, 100069, China
| | - Chunlin Li
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Beijing, 100069, China
| | - Liwei Sun
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Beijing, 100069, China.
| | - Xu Zhang
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Beijing, 100069, China.
| |
Collapse
|
2
|
Antila H, Lilius TO, Palada V, Lohela T, Bell RF, Porkka-Heiskanen T, Kalso E. Effects of commonly used analgesics on sleep architecture-A topical review. Pain 2024; 165:00006396-990000000-00539. [PMID: 38442410 PMCID: PMC11247456 DOI: 10.1097/j.pain.0000000000003201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 03/07/2024]
Affiliation(s)
- Hanna Antila
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Finland
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
| | - Tuomas O. Lilius
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland
- Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Finland
| | - Vinko Palada
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Finland
| | - Terhi Lohela
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland
| | - Rae F. Bell
- Regional Centre of Excellence in Palliative Care, Haukeland University Hospital, Bergen, Norway
| | | | - Eija Kalso
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland
| |
Collapse
|
3
|
Hummel L, Frenzel T, Boyken J, Pietsch H, Seeliger E. Comprehensive Analysis of the Spatial Distribution of Gadolinium, Iron, Manganese, and Phosphorus in the Brain of Healthy Rats After High-Dose Administrations of Gadodiamide and Gadobutrol. Invest Radiol 2024; 59:150-164. [PMID: 38157437 PMCID: PMC11441738 DOI: 10.1097/rli.0000000000001054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
OBJECTIVES After the administration of gadolinium-based contrast agents (GBCAs), residual gadolinium (Gd) has been detected in a few distinct morphological structures of the central nervous system (CNS). However, a systematic, comprehensive, and quantitative analysis of the spatial Gd distribution in the entire brain is not yet available. The first aim of this study is to provide this analysis in healthy rats after administration of high GBCA doses. The second aim is to assess the spatial distributions and possible Gd colocalizations of endogenous iron (Fe), manganese (Mn), and phosphorus (P). In addition, the presence of Gd in proximity to blood vessels was assessed by immunohistochemistry. MATERIALS AND METHODS Male rats were randomly assigned to 3 groups (n = 3/group): saline (control), gadodiamide (linear GBCA), and gadobutrol (macrocyclic GBCA) with cumulative Gd doses of 14.4 mmol/kg of body mass. Five weeks after the last administration, the brains were collected and cryosectioned. The spatial distributions of Gd, Fe, Mn, and P were analyzed in a total of 130 sections, each covering the brain in 1 of the 3 perpendicular anatomical orientations, using laser ablation coupled with inductively coupled plasma mass spectrometry. Quantitative spatial element maps were generated, and the concentrations of Gd, Fe, and Mn were measured in 31 regions of interest covering various distinct CNS structures. Correlation analyses were performed to test for possible colocalization of Gd, Fe, and Mn. The spatial proximity of Gd and blood vessels was studied using metal-tagged antibodies against von Willebrand factor with laser ablation coupled with inductively coupled plasma mass spectrometry. RESULTS After administration of linear gadodiamide, high Gd concentrations were measured in many distinct structures of the gray matter. This involved structures previously reported to retain Gd after linear GBCA, such as the deep cerebellar nuclei or the globus pallidus, but also structures that had not been reported so far including the dorsal subiculum, the retrosplenial cortex, the superior olivary complex, and the inferior colliculus. The analysis in all 3 orientations allowed the localization of Gd in specific subregions and layers of certain structures, such as the hippocampus and the primary somatosensory cortex. After macrocyclic gadobutrol, the Gd tissue concentration was significantly lower than after gadodiamide. Correlation analyses of region of interest concentrations of Gd, Fe, and Mn revealed no significant colocalization of Gd with endogenous Fe or Mn in rats exposed to either GBCA. Immunohistochemistry revealed a colocalization of Gd traces with vascular endothelium in the deep cerebellar nuclei after gadobutrol, whereas the majority of Gd was found outside the vasculature after gadodiamide. CONCLUSIONS In rats exposed to gadodiamide but not in rats exposed to gadobutrol, high Gd concentrations were measured in various distinct CNS structures, and structures not previously reported were identified to contain Gd, including specific subregions and layers with different cytoarchitecture and function. Knowledge of these distinct spatial patterns may pave the way for tailored functional neurological testing. Signs for the localization of the remaining Gd in the vascular endothelium were prominent for gadobutrol but not gadodiamide. The results also indicate that local transmetalation with endogenous Fe or Mn is unlikely to explain the spatial patterns of Gd deposition in the brain, which argues against a general role of these metals in local transmetalation and release of Gd ions in the CNS.
Collapse
|
4
|
Murase S, Sakitani N, Maekawa T, Yoshino D, Takano K, Konno A, Hirai H, Saito T, Tanaka S, Shinohara K, Kishi T, Yoshikawa Y, Sakai T, Ayaori M, Inanami H, Tomiyasu K, Takashima A, Ogata T, Tsuchimochi H, Sato S, Saito S, Yoshino K, Matsuura Y, Funamoto K, Ochi H, Shinohara M, Nagao M, Sawada Y. Interstitial-fluid shear stresses induced by vertically oscillating head motion lower blood pressure in hypertensive rats and humans. Nat Biomed Eng 2023; 7:1350-1373. [PMID: 37414976 PMCID: PMC10651490 DOI: 10.1038/s41551-023-01061-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 05/27/2023] [Indexed: 07/08/2023]
Abstract
The mechanisms by which physical exercise benefits brain functions are not fully understood. Here, we show that vertically oscillating head motions mimicking mechanical accelerations experienced during fast walking, light jogging or treadmill running at a moderate velocity reduce the blood pressure of rats and human adults with hypertension. In hypertensive rats, shear stresses of less than 1 Pa resulting from interstitial-fluid flow induced by such passive head motions reduced the expression of the angiotensin II type-1 receptor in astrocytes in the rostral ventrolateral medulla, and the resulting antihypertensive effects were abrogated by hydrogel introduction that inhibited interstitial-fluid movement in the medulla. Our findings suggest that oscillatory mechanical interventions could be used to elicit antihypertensive effects.
Collapse
Affiliation(s)
- Shuhei Murase
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyoshi Sakitani
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takahiro Maekawa
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Daisuke Yoshino
- Division of Advanced Applied Physics, Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Kouji Takano
- Department of Rehabilitation for Brain Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Ayumu Konno
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Taku Saito
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Kishi
- Department of Cardiology, Graduate School of Medicine, International University of Health and Welfare, Okawa, Japan
| | - Yuki Yoshikawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takamasa Sakai
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | - Hirohiko Inanami
- Inanami Spine & Joint Hospital/Iwai Orthopaedic Medical Hospital, Iwai Medical Foundation, Tokyo, Japan
| | - Koji Tomiyasu
- Center of Sports Science and Health Promotion, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Atsushi Takashima
- Department of Assistive Technology, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Toru Ogata
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Rehabilitation Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Shinya Sato
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Shigeyoshi Saito
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kohzoh Yoshino
- School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| | - Yuiko Matsuura
- Department of Health and Sports, Niigata University of Health and Welfare, Niigata, Japan
| | | | - Hiroki Ochi
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Masahiro Shinohara
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Motoshi Nagao
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Yasuhiro Sawada
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan.
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan.
- Division of Advanced Applied Physics, Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan.
- Department of Clinical Research, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan.
| |
Collapse
|
5
|
Jin P, Munson JM. Fluids and flows in brain cancer and neurological disorders. WIREs Mech Dis 2023; 15:e1582. [PMID: 36000149 PMCID: PMC9869390 DOI: 10.1002/wsbm.1582] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 01/31/2023]
Abstract
Interstitial fluid (IF) and cerebrospinal fluid (CSF) are an integral part of the brain, serving to cushion and protect the brain parenchymal cells against damage and aid in their function. The brain IF contains various ions, nutrients, waste products, peptides, hormones, and neurotransmitters. IF moves primarily by pressure-dependent bulk flow through brain parenchyma, draining into the ventricular CSF. The brain ventricles and subarachnoid spaces are filled with CSF which circulates through the perivascular spaces. It also flows into the IF space regulated, in part, by aquaporin channels, removing waste solutes through a process of IF-CSF mixing. During disease development, the composition, flow, and volume of these fluids changes and can lead to brain cell dysfunction. With the improvement of imaging technology and the help of genomic profiling, more information has been and can be obtained from brain fluids; however, the role of CSF and IF in brain cancer and neurobiological disease is still limited. Here we outline recent advances of our knowledge of brain fluid flow in cancer and neurodegenerative disease based on our understanding of its dynamics and composition. This article is categorized under: Cancer > Biomedical Engineering Neurological Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Peng Jin
- Fralin Biomedical Research Institute, Department of Biomedical Engineering and Mechanics Virginia Polytechnic Institute and State University Roanoke Virginia USA
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute, Department of Biomedical Engineering and Mechanics Virginia Polytechnic Institute and State University Roanoke Virginia USA
| |
Collapse
|
6
|
Gu Z, Chen H, Zhao H, Yang W, Song Y, Li X, Wang Y, Du D, Liao H, Pan W, Li X, Gao Y, Han H, Tong Z. New insight into brain disease therapy: nanomedicines-crossing blood-brain barrier and extracellular space for drug delivery. Expert Opin Drug Deliv 2022; 19:1618-1635. [PMID: 36285632 DOI: 10.1080/17425247.2022.2139369] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Brain diseases including brain tumor, Alzheimer's disease, Parkinson's disease, etc. are difficult to treat. The blood-brain barrier (BBB) is a major obstacle for drug delivery into the brain. Although nano-package and receptor-mediated delivery of nanomedicine markedly increases BBB penetration, it yet did not extensively improve clinical cure rate. Recently, brain extracellular space (ECS) and interstitial fluid (ISF) drainage in ECS have been found to determine whether a drug dissolved in ISF can reach its target cells. Notably, an increase in tortuosity of ECS associated with slower ISF drainage induced by the accumulated harmful substances, such as: amyloid-beta (Aβ), α-synuclein, and metabolic wastes, causes drug delivery failure. AREAS COVERED The methods of nano-package and receptor-mediated drug delivery and the penetration efficacy of nanomedicines across BBB and ECS are assessed. EXPERT OPINION Invasive delivering drug via ECS and noninvasive near-infrared photo-sensitive nanomedicines may provide a promising benefit to patients with brain disease.
Collapse
Affiliation(s)
- Ziqi Gu
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Haishu Chen
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Han Zhao
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Wanting Yang
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Yilan Song
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xiang Li
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Yang Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.,Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Dan Du
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Haikang Liao
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Wenhao Pan
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, China.,NMPA key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China
| | - Hongbin Han
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.,Department of Radiology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China.,Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zhiqian Tong
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Du D, Gao Y, Zheng T, Yang L, Wang Z, Shi Q, Wu S, Liang X, Yao X, Lu J, Liu L. The Value of First-Order Features Based on the Apparent Diffusion Coefficient Map in Evaluating the Therapeutic Effect of Low-Intensity Pulsed Ultrasound for Acute Traumatic Brain Injury With a Rat Model. Front Comput Neurosci 2022; 16:923247. [PMID: 35814344 PMCID: PMC9259978 DOI: 10.3389/fncom.2022.923247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose In order to evaluate the neuroprotective effect of low-intensity pulsed ultrasound (LIPUS) for acute traumatic brain injury (TBI), we studied the potential of apparent diffusion coefficient (ADC) values and ADC-derived first-order features regarding this problem. Methods Forty-five male Sprague Dawley rats (sham group: 15, TBI group: 15, LIPUS treated: 15) were enrolled and underwent magnetic resonance imaging. Scanning layers were acquired using a multi-shot readout segmentation of long variable echo trains (RESOLVE) to decrease distortion. The ultrasound transducer was applied to the designated region in the injured cortical areas using a conical collimator and was filled with an ultrasound coupling gel. Regions of interest were manually delineated in the center of the damaged cortex on the diffusion weighted images (b = 800 s/mm2) layer by layer for the TBI and LIPUS treated groups using the open-source software ITK-SNAP. Before analysis and modeling, the features were normalized using a z-score method, and a logistic regression model with a backward filtering method was employed to perform the modeling. The entire process was completed using the R language. Results During the observation time, the ADC values ipsilateral to the trauma in the TBI and LIPUS groups increased rapidly up to 24 h. After statistical analysis, the 10th percentile, 90th percentile, mean, skewness, and uniformity demonstrated a significant difference among three groups. The receiver operating characteristic curve (ROC) analysis shows that the combined LR model exhibited the highest area under the curve value (AUC: 0.96). Conclusion The combined LR model of first-order features based on the ADC map can acquire a higher diagnostic performance than each feature only in evaluating the neuroprotective effect of LIPUS for TBI. Models based on first-order features may have potential value in predicting the therapeutic effect of LIPUS in clinical practice in the future.
Collapse
Affiliation(s)
- Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, China
- NMPA Key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China
- Peking University Shenzhen Graduate School, Shenzhen, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Linsha Yang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Zhanqiu Wang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Qinglei Shi
- MR Scientific Marketing, Siemens Healthineers Ltd., Beijing, China
| | - Shuo Wu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Xin Liang
- Graduate School of Chengde Medical University, Chengde, China
| | - Xinyu Yao
- Graduate School of Chengde Medical University, Chengde, China
| | - Jiabin Lu
- Beijing Key Laboratory of Magnetic Resonance Imaging Device and Technique, Beijing, China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
- *Correspondence: Lanxiang Liu,
| |
Collapse
|
8
|
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
|
9
|
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: 21] [Impact Index Per Article: 7.0] [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
|
10
|
Gao Y, Han H, Du J, He Q, Jia Y, Yan J, Dai H, Cui B, Yang J, Wei X, Yang L, Wang R, Long R, Ren Q, Yang X, Lu J. Early changes to the extracellular space in the hippocampus under simulated microgravity conditions. SCIENCE CHINA-LIFE SCIENCES 2021; 65:604-617. [PMID: 34185240 DOI: 10.1007/s11427-021-1932-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023]
Abstract
The smooth transportation of substances through the brain extracellular space (ECS) is crucial to maintaining brain function; however, the way this occurs under simulated microgravity remains unclear. In this study, tracer-based magnetic resonance imaging (MRI) and DECS-mapping techniques were used to image the drainage of brain interstitial fluid (ISF) from the ECS of the hippocampus in a tail-suspended hindlimb-unloading rat model at day 3 (HU-3) and 7 (HU-7). The results indicated that drainage of the ISF was accelerated in the HU-3 group but slowed markedly in the HU-7 group. The tortuosity of the ECS decreased in the HU-3 group but increased in the HU-7 group, while the volume fraction of the ECS increased in both groups. The diffusion rate within the ECS increased in the HU-3 group and decreased in the HU-7 group. The alterations to ISF drainage and diffusion in the ECS were recoverable in the HU-3 group, but neither parameter was restored in the HU-7 group. Our findings suggest that early changes to the hippocampal ECS and ISF drainage under simulated microgravity can be detected by tracer-based MRI, providing a new perspective for studying microgravity-induced nano-scale structure abnormities and developing neuroprotective approaches involving the brain ECS.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China. .,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China. .,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China.
| | - Jichen Du
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China.,Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Yanxing Jia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Junhao Yan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hui Dai
- NHC Key Laboratory of Medical Immunology, Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Bin Cui
- Department of Radiology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Jing Yang
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Xunbin Wei
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Liu Yang
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Ren Long
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Qiushi Ren
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Xing Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Jiabin Lu
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| |
Collapse
|
11
|
Naseri Kouzehgarani G, Feldsien T, Engelhard HH, Mirakhur KK, Phipps C, Nimmrich V, Clausznitzer D, Lefebvre DR. Harnessing cerebrospinal fluid circulation for drug delivery to brain tissues. Adv Drug Deliv Rev 2021; 173:20-59. [PMID: 33705875 DOI: 10.1016/j.addr.2021.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 12/31/2022]
Abstract
Initially thought to be useful only to reach tissues in the immediate vicinity of the CSF circulatory system, CSF circulation is now increasingly viewed as a viable pathway to deliver certain therapeutics deeper into brain tissues. There is emerging evidence that this goal is achievable in the case of large therapeutic proteins, provided conditions are met that are described herein. We show how fluid dynamic modeling helps predict infusion rate and duration to overcome high CSF turnover. We posit that despite model limitations and controversies, fluid dynamic models, pharmacokinetic models, preclinical testing, and a qualitative understanding of the glymphatic system circulation can be used to estimate drug penetration in brain tissues. Lastly, in addition to highlighting landmark scientific and medical literature, we provide practical advice on formulation development, device selection, and pharmacokinetic modeling. Our review of clinical studies suggests a growing interest for intra-CSF delivery, particularly for targeted proteins.
Collapse
|
12
|
Comprehensive phenotyping revealed transient startle response reduction and histopathological gadolinium localization to perineuronal nets after gadodiamide administration in rats. Sci Rep 2020; 10:22385. [PMID: 33372182 PMCID: PMC7769977 DOI: 10.1038/s41598-020-79374-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/01/2020] [Indexed: 01/28/2023] Open
Abstract
Gadolinium based contrast agents (GBCAs) are widely used in clinical MRI since the mid-1980s. Recently, concerns have been raised that trace amounts of Gadolinium (Gd), detected in brains even long time after GBCA application, may cause yet unrecognized clinical consequences. We therefore assessed the behavioral phenotype, neuro-histopathology, and Gd localization after repeated administration of linear (gadodiamide) or macrocyclic (gadobutrol) GBCA in rats. While most behavioral tests revealed no difference between treatment groups, we observed a transient and reversible decrease of the startle reflex after gadodiamide application. Residual Gd in the lateral cerebellar nucleus was neither associated with a general gene expression pathway deregulation nor with neuronal cell loss, but in gadodiamide-treated rats Gd was associated with the perineuronal net protein aggrecan and segregated to high molecular weight fractions. Our behavioral finding together with Gd distribution and speciation support a substance class difference for Gd presence in the brain after GBCA application.
Collapse
|
13
|
Li Y, Han H, Shi K, Cui D, Yang J, Alberts IL, Yuan L, Zhao G, Wang R, Cai X, Teng Z. The Mechanism of Downregulated Interstitial Fluid Drainage Following Neuronal Excitation. Aging Dis 2020; 11:1407-1422. [PMID: 33269097 PMCID: PMC7673848 DOI: 10.14336/ad.2020.0224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
The drainage of brain interstitial fluid (ISF) has been observed to slow down following neuronal excitation, although the mechanism underlying this phenomenon is yet to be elucidated. In searching for the changes in the brain extracellular space (ECS) induced by electrical pain stimuli in the rat thalamus, significantly decreased effective diffusion coefficient (DECS) and volume fraction (α) of the brain ECS were shown, accompanied by the slowdown of ISF drainage. The morphological basis for structural changes in the brain ECS was local spatial deformation of astrocyte foot processes following neuronal excitation. We further studied aquaporin-4 gene (APQ4) knockout rats in which the changes of the brain ECS structure were reversed and found that the slowed DECS and ISF drainage persisted, confirming that the down-regulation of ISF drainage following neuronal excitation was mainly attributable to the release of neurotransmitters rather than to structural changes of the brain ECS. Meanwhile, the dynamic changes in the DECS were synchronized with the release and elimination processes of neurotransmitters following neuronal excitation. In conclusion, the downregulation of ISF drainage following neuronal excitation was found to be caused by the restricted diffusion in the brain ECS, and DECS mapping may be used to track the neuronal activity in the deep brain.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, 3010 Bern, Switzerland.
- Department of Informatics, Technical University of Munich, Garching 85748, Germany.
| | - Dehua Cui
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Jun Yang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
| | - Ian Leigh Alberts
- Department of Nuclear Medicine, University of Bern, 3010 Bern, Switzerland.
| | - Lan Yuan
- Peking University Medical and Health Analysis Center, Peking University Health Science Center, Beijing, China.
| | - Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xianjie Cai
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Ze Teng
- Department of Radiology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
14
|
Dong Y, Yuan Y, Fang Y, Zheng T, Du D, Gao D, Du J, Liu L, He Q. Effect of aquaporin 4 protein overexpression in nigrostriatal system on development of Parkinson's disease. Int J Neurosci 2020; 131:666-673. [PMID: 32259464 DOI: 10.1080/00207454.2020.1753727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTS Recent studies indicated that aquaporin 4 (AQP4), as the main water channel in the central nervous system (CNS), participated in the onset and progression of Parkinson's disease (PD). But how the AQP4 influenced the exacerbation of PD has not been described in detail. In this study, the effect of the AQP4 protein overexpression in nigrostriatal system that include substantia nigra (SN) and striatum (CPu) on the development of PD was investigated. METHODS Forty male Sprague Dawley rats were equally divided into two groups at random: PD group and control group, PD group undergoing surgery and receiving 6-hydroxydopamine (6-OHDA). Using MRI tracer-based method, extracellular space (ECS) diffusion parameters of nigrostriatal system for all rats were measured, including the clearance coefficient (k') and the half-life (t1/2). Immunohistochemistry of AQP4 was performed for 20 rats. RESULTS The area of dark-stained AQP4 immunoreactivity increased markedly in SN of PD rats, there were significant differences between two groups (SN: t = 5.809, p < 0.0001; CPu: t = 5.943, p < 0.0001). And the diffusion parameters were significantly greater in PD group than that of control group, including k' (SN: t = 5.519, p < 0.0001; CPu: t = 2.149, p = 0.045) and t1/2 (SN: t = 6.131, p < 0.0001; CPu: t = 6.708, p < 0.0001). There was a significant positive correlation between the AQP4 expression level and the k' values (SN: r = 0.827, p = 0.0031; CPu: r = 0.641, p = 0.0046), and a significant negative correlation between AQP4 and the t1/2 values (SN: r=-0.654, p = 0.0403; CPu: r=-0.664, p = 0.0362). CONCLUSIONS The results indicated that AQP4 expression was increased in nigrostriatal system of PD rats, therefore, the overexpression of AQP4 led to acceleration of the diffusion and drainage process of drugs in ECS, reduced the effect of drugs for the treatment of PD, inhibited the development of PD.
Collapse
Affiliation(s)
- Yanchao Dong
- Department of Interventional Therapy, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Yi Yuan
- College of Electrical Engineering, Yanshan University, Qinhuangdao, China
| | - Yuan Fang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Dawei Gao
- College of Chemical Engineering, Yanshan University, Qinhuangdao, China
| | - Juan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
15
|
Zhao G, Han H, Yang J, Sun M, Cui D, Li Y, Gao Y, Zou J. Brain interstitial fluid drainage and extracellular space affected by inhalational isoflurane: in comparison with intravenous sedative dexmedetomidine and pentobarbital sodium. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1363-1379. [PMID: 32133594 DOI: 10.1007/s11427-019-1633-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/06/2020] [Indexed: 12/09/2022]
Abstract
Brain interstitial fluid drainage and extracellular space are closely related to waste clearance from the brain. Different anesthetics may cause different changes of brain interstitial fluid drainage and extracellular space but these still remain unknown. Herein, effects of the inhalational isoflurane, intravenous sedative dexmedetomidine and pentobarbital sodium on deep brain matters' interstitial fluid drainage and extracellular space and underlying mechanisms were investigated. When compared to intravenous anesthetic dexmedetomidine or pentobarbital sodium, inhalational isoflurane induced a restricted diffusion of extracellular space, a decreased extracellular space volume fraction, and an increased norepinephrine level in the caudate nucleus or thalamus with the slowdown of brain interstitial fluid drainage. A local administration of norepinephrine receptor antagonists, propranolol, atipamezole and prazosin into extracellular space increased diffusion of extracellular space and interstitial fluid drainage whilst norepinephrine decreased diffusion of extracellular space and interstitial fluid drainage. These findings suggested that restricted diffusion in brain extracellular space can cause slowdown of interstitial fluid drainage, which may contribute to the neurotoxicity following the waste accumulation in extracellular space under inhaled anesthesia per se.
Collapse
Affiliation(s)
- Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China. .,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China. .,Institute of Medical Technology, Peking University, Beijing, 100191, China.
| | - Jun Yang
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China. .,Department of Neurosurgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Min Sun
- Department of Cardiology, Peking University Shougang Hospital, Beijing, 100144, China
| | - Dehua Cui
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Jing Zou
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| |
Collapse
|
16
|
Novel imaging and related techniques for studies of diseases of the central nervous system: a review. Cell Tissue Res 2020; 380:415-424. [PMID: 32072308 DOI: 10.1007/s00441-020-03183-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 01/29/2020] [Indexed: 10/25/2022]
Abstract
Imaging technologies for the analysis of the central nervous system are rapidly developing. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry imaging, tracer-based magnetic resonance imaging, CLARITY technology and optogenetics can be used to visualize small molecules in brain tissues, the interstitial system of the brain and neuronal circuits in whole-brain samples. These tools serve as powerful technical means to explore the mechanisms underlying disease models and to evaluate the effects of drugs. Here, we review the constituting principles of these imaging techniques and describe their applications in the field of neuroscience.
Collapse
|
17
|
Shetty AK, Zanirati G. The Interstitial System of the Brain in Health and Disease. Aging Dis 2020; 11:200-211. [PMID: 32010493 PMCID: PMC6961771 DOI: 10.14336/ad.2020.0103] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022] Open
Abstract
The brain interstitial fluid (ISF) and the cerebrospinal fluid (CSF) cushion and support the brain cells. The ISF occupies the brain interstitial system (ISS), whereas the CSF fills the brain ventricles and the subarachnoid space. The brain ISS is an asymmetrical, tortuous, and exceptionally confined space between neural cells and the brain microvasculature. Recently, with a newly developed in vivo measuring technique, a series of discoveries have been made in the brain ISS and the drainage of ISF. The goal of this review is to confer recent advances in our understanding of the brain ISS, including its structure, function, and the various processes mediating or disrupting ISF drainage in physiological and pathological conditions. The brain ISF in the deep brain regions has recently been demonstrated to drain in a compartmentalized ISS instead of a highly connected system, together with the drainage of ISF into the cerebrospinal fluid (CSF) at the surface of the cerebral cortex and the transportation from CSF into cervical lymph nodes. Besides, accumulation of tau in the brain ISS in conditions such as Alzheimer’s disease and its link to the sleep-wake cycle and sleep deprivation, clearance of ISF in a deep sleep via increased CSF flow, novel approaches to remove beta-amyloid from the brain ISS, and obstruction to the ISF drainage in neurological conditions are deliberated. Moreover, the role of ISS in the passage of extracellular vesicles (EVs) released from neural cells and the rapid targeting of therapeutic EVs into neural cells in the entire brain following an intranasal administration, and the promise and limitations of ISS based drug delivery approaches are discussed
Collapse
Affiliation(s)
- Ashok K Shetty
- 1Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX 77843, USA
| | - Gabriele Zanirati
- 2Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
18
|
Wang A, Wang R, Cui D, Huang X, Yuan L, Liu H, Fu Y, Liang L, Wang W, He Q, Shi C, Guan X, Teng Z, Zhao G, Li Y, Gao Y, Han H. The Drainage of Interstitial Fluid in the Deep Brain is Controlled by the Integrity of Myelination. Aging Dis 2019; 10:937-948. [PMID: 31595193 PMCID: PMC6764732 DOI: 10.14336/ad.2018.1206] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
In searching for the drainage route of the interstitial fluid (ISF) in the deep brain, we discovered a regionalized ISF drainage system as well as a new function of myelin in regulating the drainage. The traced ISF from the caudate nucleus drained to the ipsilateral cortex along myelin fiber tracts, while in the opposite direction, its movement to the adjacent thalamus was completely impeded by a barrier structure, which was identified as the converged, compact myelin fascicle. The regulating and the barrier effects of myelin were unchanged in AQP4-knockout rats but were impaired as the integrity of boundary structure of drainage system was destroyed in a demyelinated rat model. We thus proposed that the brain homeostasis was maintained within each ISF drainage division locally, rather than across the brain as a whole. A new brain division system and a new pathogenic mechanism of demyelination are therefore proposed.
Collapse
Affiliation(s)
- Aibo Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Dehua Cui
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xinrui Huang
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Lan Yuan
- Peking University Medical and Health Analysis Center, Peking University Health Science Center, Beijing, China.
| | - Huipo Liu
- Institute of Applied Physics and Computational Mathematics, Beijing, China.
| | - Yu Fu
- Department of Neurology, Peking University Third Hospital, Beijing, China.
| | - Lei Liang
- Department of Medical Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Wei Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Chunyan Shi
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xiangping Guan
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Ze Teng
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yajuan Gao
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| |
Collapse
|
19
|
Han HB. [Discovery of a new division system in brain and the regionalized drainage route of brain interstitial fluid]. JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2019; 51:397-401. [PMID: 31209408 DOI: 10.19723/j.issn.1671-167x.2019.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Brain extracellular space (ECS) is a narrow, irregular space, which provides immediate living environment for neural cells and accounts for approximately 15%-20% of the total volume of living brain. Twenty-five years ago, as an interventional radiologist, the author was engaged in investigating early diagnosis and treatment of cerebral ischemic stroke, and the parameters of brain ECS was firstly derived and demonstrated during the study of the permeability of blood-brain barrier (BBB) and its diffusion changes in the cerebral ischemic tissue. Since then, the author and his team had been working on developing a novel measuring method of ECS: tracer-based magnetic resonance imaging (MRI), which could measure brain ECS parameters in the whole brain scale and make the dynamic drainage process of the labelled brain interstitial fluid (ISF) visualized. By using the new method, the team made a series of new findings about the brain ECS and ISF, including the discovery of a new division system in the brain, named regionalized ISF drainage system. We found that the ISF drainage in the deep brain was regionalized and the structural and functional parameters in different interstitial system (ISS) divisions were disparate. The ISF in the caudate nucleus could be drained to ipsilateral cortex and finally into the subarachnoid space, which maintained the pathway of ISF-cerebrospinal fluid (CSF) exchange. However, the ISF in the thalamus was eliminated locally in its anatomical division. After verifying the nature of the barrier structure between different drainage divisions, the author proposed the hypothesis of "regionalized brain homeostasis". Thus, we demonstrated that the brain was protected not only by the BBB, which avoided potential exogenous damage through the vascular system, but was also protected by an internal ISF drainage barrier to avoid potentially harmful interference from other ECS divisions in the deep brain. With the new findings and the proposed hypothesis, an innovative therapeutic method for the treatment of encephalopathy with local drug delivery via the brain ECS pathway was established. By using this new administration method, the drug was achieved directly to the space around neurons or target regions, overwhelming the impendence from the blood-brain barrier, thus solved the obstacles of low efficiency in traditional drug investigation. At present, new methods and discoveries developed by the author and his team have been widely applied in several frontier fields including neuroscience, new drug research and development, neurodevelopment aerospace medicine, clinical encephalopathy treatment,new neural network modeling and so on.
Collapse
Affiliation(s)
- H B Han
- Department of Radiology, Peking University Third Hospital; Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing 100191, China
| |
Collapse
|
20
|
王 伟, 侯 进, 黄 文. [Temporary acceleration of interstitial fluid drainage in excited brain region induced by movement]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2019; 51:206-209. [PMID: 30996355 PMCID: PMC7441198 DOI: 10.19723/j.issn.1671-167x.2019.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To investigate the changes of brain interstitial fluid (ISF) induced by movement. METHODS Twenty mature male Sprague-Dawley rats were randomly divided into two groups: control group and movement group. Electrophysiological neurons in caudate nuclear of additional five rats were recorded and the differences analyzed between under anesthesia and by movement. In the control group, the rats were anesthetized using isoflurane continuously during the experiment process. In the meantime the magnetic tracer was injected into the center of the caudate nucleus and multi-period magnetic resonance scanning was performed at several time points until high signal intensity invisible in the images. In the movement group, the rats were anesthetized for the injection of the tracer, and the first post-injection magnetic resonance scanning was performed. Then the rats were waken and allowed moving voluntarily for 20 minutes. The rats were anesthetized again and multi-period magnetic resonance scanning was performed until the experiment ended. NanoDetect system (Version 1.2, MRI lab, Beijing, China) was used to measure the parameters on ISF, which included the weighed signal intensity (weighed ΔSI) , the term predicting the amount of the tracer, and half-time of the tracer. In movement group, the weighed ΔSI at the time points of pre-movement and 10, 40, 70, 130, and 190 minutes after movement were calculated respectively. In control group, the weighed ΔSI at the same time points also were measured. The weighed ΔSI and half-time were compared between the two groups. RESULTS The electrophysiological recording and data analysis showed significant difference in the local field potential of Caudate Nucleus between under anesthesia and by movement. The weighed ΔSI (unit: ΔSI×mm3) values of the two groups, presented by movement group vs. control group, were as followings, 60 257.1±23 069.2 vs. 61 072.0±19 547.3 at pre-move, 83 624.3±21 475.7 vs. 71 218.1±12 586.5 at 10 min after movement, 57 336.0±36 243.4 vs. 69 756.1±13 306.0 at 40 min after movement, 43 705.9±10 246.3 vs. 55 443.2±20 733.3 at 70 min after movement, 7 734.9±2 645.2 vs. 8 967.6±2 007.3 at 130 min after movement and 2 497.3±987.5 vs. 3 013.2±1 760.8 at 190 min after movement. Moreover, at 40 min after movement, the weighed ΔSI of movement group was significantly reduced compared with control group (P<0.05). The half-time was not significantly different [(104.3±54.1) min vs. (113.4±47.3) min, P>0.05]. CONCLUSION ISF drainage of caudate nuclear can be acclerated temporarily by movement.
Collapse
Affiliation(s)
- 伟 王
- 佛山市第一人民医院影像科, 广东佛山 528000Department of Radiology, The First People’s Hospital of Foshan, Foshan 528000, Guangdong, China;
| | - 进 侯
- 广州医科大学附属第二医院放射科, 广州 510260Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 文强 黄
- 首都师范大学心理学院, 北京 100048School of Psychology, Capital Normal University, Beijing 100048, China
| |
Collapse
|
21
|
王 伟, 侯 进, 黄 文. [Temporary acceleration of interstitial fluid drainage in excited brain region induced by movement]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2019; 51:206-209. [PMID: 30996355 PMCID: PMC7441198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To investigate the changes of brain interstitial fluid (ISF) induced by movement. METHODS Twenty mature male Sprague-Dawley rats were randomly divided into two groups: control group and movement group. Electrophysiological neurons in caudate nuclear of additional five rats were recorded and the differences analyzed between under anesthesia and by movement. In the control group, the rats were anesthetized using isoflurane continuously during the experiment process. In the meantime the magnetic tracer was injected into the center of the caudate nucleus and multi-period magnetic resonance scanning was performed at several time points until high signal intensity invisible in the images. In the movement group, the rats were anesthetized for the injection of the tracer, and the first post-injection magnetic resonance scanning was performed. Then the rats were waken and allowed moving voluntarily for 20 minutes. The rats were anesthetized again and multi-period magnetic resonance scanning was performed until the experiment ended. NanoDetect system (Version 1.2, MRI lab, Beijing, China) was used to measure the parameters on ISF, which included the weighed signal intensity (weighed ΔSI) , the term predicting the amount of the tracer, and half-time of the tracer. In movement group, the weighed ΔSI at the time points of pre-movement and 10, 40, 70, 130, and 190 minutes after movement were calculated respectively. In control group, the weighed ΔSI at the same time points also were measured. The weighed ΔSI and half-time were compared between the two groups. RESULTS The electrophysiological recording and data analysis showed significant difference in the local field potential of Caudate Nucleus between under anesthesia and by movement. The weighed ΔSI (unit: ΔSI×mm3) values of the two groups, presented by movement group vs. control group, were as followings, 60 257.1±23 069.2 vs. 61 072.0±19 547.3 at pre-move, 83 624.3±21 475.7 vs. 71 218.1±12 586.5 at 10 min after movement, 57 336.0±36 243.4 vs. 69 756.1±13 306.0 at 40 min after movement, 43 705.9±10 246.3 vs. 55 443.2±20 733.3 at 70 min after movement, 7 734.9±2 645.2 vs. 8 967.6±2 007.3 at 130 min after movement and 2 497.3±987.5 vs. 3 013.2±1 760.8 at 190 min after movement. Moreover, at 40 min after movement, the weighed ΔSI of movement group was significantly reduced compared with control group (P<0.05). The half-time was not significantly different [(104.3±54.1) min vs. (113.4±47.3) min, P>0.05]. CONCLUSION ISF drainage of caudate nuclear can be acclerated temporarily by movement.
Collapse
Affiliation(s)
- 伟 王
- 佛山市第一人民医院影像科, 广东佛山 528000Department of Radiology, The First People’s Hospital of Foshan, Foshan 528000, Guangdong, China;
| | - 进 侯
- 广州医科大学附属第二医院放射科, 广州 510260Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 文强 黄
- 首都师范大学心理学院, 北京 100048School of Psychology, Capital Normal University, Beijing 100048, China
| |
Collapse
|
22
|
Li K, Zhang Y, He D, Han H, Yuan L. Homogeneous orthogonal diffusion of drugs in the rat brain. Clin Exp Pharmacol Physiol 2019; 46:398-403. [PMID: 30570785 DOI: 10.1111/1440-1681.13057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/23/2018] [Accepted: 12/12/2018] [Indexed: 11/28/2022]
Abstract
The study introduced a homogeneous orthogonal diffusion model to describe the diffusion pattern of agents in the rat brain. Magnetic resonance imaging was performed post-injection of paramagnetic drugs into the caudatum of the rat brain at predetermined time intervals. The signal intensity on magnetic resonance images was converted to agents' concentration, and the standard least square method was employed to estimate the diffusion coefficients. The diffusion coefficients calculated along the three orthogonal axes were D1 = (0.0097 ± 0.0036) mm2 /h, D2 = (0.0153 ± 0.0033) mm2 /h, and D3 = (0.0293 ± 0.0155) mm2 /h; the clearance rate constant was k = (0.2177 ± 0.0112)/h. The theoretical homogeneous orthogonal model can enhance our knowledge on both the biophysical characteristics of brain extracellular space and the interstitial drug delivery in the brain.
Collapse
Affiliation(s)
- Kai Li
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Yunjun Zhang
- Department of Applied Science, Peking University Health Science Center, Beijing, China
| | - Dongqi He
- Department of Applied Science, Peking University Health Science Center, Beijing, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Beijing Key Lab of MRI Device and Technique, Beijing, China
| | - Lan Yuan
- Center of Medical and Health Analysis, Peking University, Beijing, China
| |
Collapse
|
23
|
Wang W, He Q, Hou J, Chui D, Gao M, Wang A, Han H, Liu H. Stimulation Modeling on Three-Dimensional Anisotropic Diffusion of MRI Tracer in the Brain Interstitial Space. Front Neuroinform 2019; 13:6. [PMID: 30837860 PMCID: PMC6390635 DOI: 10.3389/fninf.2019.00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/29/2019] [Indexed: 01/04/2023] Open
Abstract
Purpose: To build a mathematical model based magnetic resonance (MR) method to simulate drug anisotropic distribution in vivo in the interstitial space (ISS) of the brain. Materials and Methods: An injection of signal intensity-related gadolinium-diethylenetriaminepentaacetic acid (Gd-DTPA), which is an exogenous drug, was administered, and its diffusion was traced in the ISS of the brain using MRI. Dynamic MRI scans were performed to monitor and record the changes in signal intensity in each pixel of the region of interest. The transport parameters were calculated using the modified equation to simulate three-dimensional anisotropic diffusion, which was resolved using a Laplace transform and a linear regressive model. Results: After Gd-DTPA was introduced into the caudate nucleus, its distribution was demonstrated in real time. As the Gd-DTPA gradually cleared, the associated hyperintensity attenuated over time. The average diffusion coefficient (D) and the clearance rate constant (k) were (1.305 ± 0.364) × 10−4 mm2/s and (1.40 ± 0.206) × 10−5 s−1, respectively. Discussion: The combination of trace-based MRI and modified diffusion mathematical models can visualize and measure the three-dimensional anisotropic distribution of drugs in the ISS of the brain.
Collapse
Affiliation(s)
- Wei Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Department of Radiology, The First People's Hospital of FoShan, Affiliated FoShan Hospital of Sun Yat-sen University, Foshan, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Jin Hou
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dehua Chui
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Mingyong Gao
- Department of Radiology, The First People's Hospital of FoShan, Affiliated FoShan Hospital of Sun Yat-sen University, Foshan, China
| | - Aibo Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, China
| | - Huipo Liu
- Institute of Applied Physics and Computational Mathematics, Beijing, China
| |
Collapse
|
24
|
Teng Z, Wang A, Wang P, Wang R, Wang W, Han H. The Effect of Aquaporin-4 Knockout on Interstitial Fluid Flow and the Structure of the Extracellular Space in the Deep Brain. Aging Dis 2018; 9:808-816. [PMID: 30271658 PMCID: PMC6147590 DOI: 10.14336/ad.2017.1115] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022] Open
Abstract
It has been reported that aquaporin-4 (AQP4) deficiency impairs transportation between the cerebrospinal fluid and interstitial fluid (ISF) as well as the clearance of interstitial solutes in the superficial brain. However, the effect of AQP4 on ISF flow in the deep brain remains unclear. This study compared the brain ISF flow in the caudate nucleus and thalamus of normal rats (NO) and AQP4 knockout rats (KO) using tracer-based magnetic resonance imaging. The rate of brain ISF flow slowed to different degrees in the two regions of KO rats’ brains. Compared with NO rats, the half-life of ISF in the thalamus of KO rats was significantly prolonged, with a corresponding decrease in the clearance coefficient. The tortuosity of the brain extracellular space (ECS) was unchanged in the thalamus of KO rats. In the caudate nucleus of KO rats, the volume fraction of the ECS and the diffusion coefficient were increased, with significantly decreased tortuosity; no significant changes in brain ISF flow were demonstrated. Combined with a change in the expression of glial fibrillary acidic protein and AQP4 in two brain regions, we found that the effect of AQP4 knockout on ISF flow and ECS structure in these two regions differed. This difference may be related to the distribution of astrocytes and the extent of AQP4 decline. This study provides evidence for the involvement of AQP4 in ISF transportation in the deep brain and provides a basis for the establishment of a pharmacokinetic model of the brain’s interstitial pathway.
Collapse
Affiliation(s)
- Ze Teng
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Aibo Wang
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Peng Wang
- 3Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Rui Wang
- 2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Wei Wang
- 2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Hongbin Han
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| |
Collapse
|
25
|
Guan X, Wang W, Wang A, Teng Z, Han H. Brain Interstitial Fluid Drainage Alterations in Glioma-Bearing Rats. Aging Dis 2018. [DOI: 10.14336/ad.2017.0415] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
26
|
Fang Y, Zheng T, Liu L, Gao D, Shi Q, Dong Y, Du D. Role of the combination of FA and T2* parameters as a new diagnostic method in therapeutic evaluation of parkinson's disease. J Magn Reson Imaging 2017; 48:84-93. [PMID: 29148118 DOI: 10.1002/jmri.25900] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/06/2017] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Simple diffusion delivery (SDD) has attained good effects with only tiny amounts of drugs. Fractional anisotropy (FA) and relaxation time T2* that indicate the integrity of fiber tracts and iron concentration within brain tissue were used to evaluate the therapeutic effect of SDD. PURPOSE To evaluate therapeutic effect of SDD in the Parkinson's disease (PD) rat model with FA and T2* parameters. STUDY TYPE Prospective case-control animal study. POPULATION Thirty-two male Sprague Dawley rats (eight normal, eight PD, eight SDD, and eight subcutaneous injection rats). FIELD STRENGTH/SEQUENCE Single-shot spin echo echo-planar imaging and fast low-angle shot T2 WI sequences at 3.0T. ASSESSMENT Parameters of FA and T2* on the treated side of the substantia nigra were measured to evaluate the therapeutic effect of SDD in a PD rat model. STATISTICAL TESTS The effects of time on FA and T2* values were analyzed by repeated measurement tests. A one-way analysis of variance was conducted, followed by individual comparisons of the mean FA and T2* values at different timepoints. RESULTS The FA values on the treated side of the substantia nigra in the SDD treatment group and subcutaneous injection treatment group were significantly higher at week 1 and lower at week 6 than that of the PD control group (SDD vs. PD, week 1, adjusted P = 0.012; subcutaneous vs. PD, week 1, adjusted P < 0.001; SDD vs. PD, week 6, adjusted P = 0.004; subcutaneous vs. PD, week 6, adjusted P = 0.024). The T2* parameter in the SDD treatment group and subcutaneous injection treatment group was significantly higher than that in the PD control group at week 6 (SDD vs. PD, adjusted P = 0.032; subcutaneous vs. PD, adjusted P < 0.001). DATA CONCLUSION The combination of FA and T2* parameters can potentially serve as a new effective evaluation method of the therapeutic effect of SDD. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 4 J. Magn. Reson. Imaging 2017.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P.R. China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P.R. China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P.R. China
| | - Dawei Gao
- Applied Chemical Key Lab of Hebei Province, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao, P.R. China
| | - Qinglei Shi
- Scientific Clinical Specialist, Siemens Ltd., Beijing, P.R. China
| | - Yanchao Dong
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P.R. China
| | - Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P.R. China
| |
Collapse
|
27
|
Yuan Y, Dong Y, Hu S, Zheng T, Du D, Du J, Liu L. Reduced Apparent Diffusion Coefficient in Various Brain Areas following Low-Intensity Transcranial Ultrasound Stimulation. Front Neurosci 2017; 11:562. [PMID: 29062269 PMCID: PMC5640877 DOI: 10.3389/fnins.2017.00562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/25/2017] [Indexed: 01/30/2023] Open
Abstract
Diffusion of water molecules closely related to physiological and pathological information of brain tissue. Low-intensity transcranial ultrasound stimulation (TUS) has advantages of noninvasive, high spatial resolution and penetration depth. Previous studies have demonstrate that TUS can modulate neuronal activity and alter cortical hemodynamic. However, how TUS affect diffusion of water molecules remain unclear. In this paper, in order to evaluate the effect of low-intensity TUS on the diffusion of water molecules in brain tissue, diffusion-weighted magnetic resonance (MR) imaging was performed in 19 healthy Sprague-Dawley rats in sham surgery group (six rats) and TUS group (thirteen rats) Subsequently, rats were stimulated by low-intensity transcranial ultrasound for 5 min in TUS group. Finally, rats of sham surgery group and TUS group were imaged again by diffusion-weighted MR imaging. The apparent diffusion coefficient (ADC) was measured in caudate putamen region and middle brain motor-related region of each rat in sham surgery group and TUS group. Surgery-related and TUS-related changes were calculated using a statistical analysis. The mean ADC values of marked regions of six rats in sham surgery group were 0.743 ± 0.031 (pre-surgery) and 0.745 ± 0.029 (post-surgery). The mean ADC values of marked regions of 13 rats in TUS group were 0.749 ± 0.032 (pre-TUS) and 0.712 ± 0.033 (post-TUS) Compared to the pre-TUS values, the mean ADC values of the rats decreased 4.9% (*P < 0.05) post-TUS. These results of this study demonstrate that low-intensity TUS can restrict the diffusion of water molecules in brain tissue.
Collapse
Affiliation(s)
- Yi Yuan
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, China
| | - Yanchao Dong
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Shuo Hu
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Juan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| |
Collapse
|
28
|
Hou J, Wang W, Quan X, Liang W, Li Z, Chen D, Han H. Quantitative Visualization of Dynamic Tracer Transportation in the Extracellular Space of Deep Brain Regions Using Tracer-Based Magnetic Resonance Imaging. Med Sci Monit 2017; 23:4260-4268. [PMID: 28866708 PMCID: PMC5595121 DOI: 10.12659/msm.903010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background This study assessed an innovative tracer-based magnetic resonance imaging (MRI) system to visualize the dynamic transportation of tracers in regions of deep brain extracellular space (ECS) and to measure transportation ability and ECS structure. Material/Methods Gadolinium-diethylene triamine pentaacetic acid (Gd-DTPA) was the chosen tracer and was injected into the caudate nucleus and thalamus. Real-time dynamic transportation of Gd-DTPA in ECS was observed and the results were verified by laser scanning confocal microscopy. Using Transwell assay across the blood-brain barrier, a modified diffusion equation was further simplified. Effective diffusion coefficient D* and tortuosity λ were calculated. Immunohistochemical staining and Western blot analysis were used to investigate the extracellular matrix contributing to ECS structure. Results Tracers injected into the caudate nucleus were transported to the ipsilateral frontal and temporal cortices away from the injection points, while both of them injected into the thalamus were only distributed on site. Although the caudate nucleus was closely adjacent to the thalamus, tracer transportation between partitions was not observed. In addition, D* and the λ showed statistically significant differences between partitions. ECS was shown to be a physiologically partitioned system, and its division is characterized by the unique distribution territory and transportation ability of substances located in it. Versican and Tenascin R are possible contributors to the tortuosity of ECS. Conclusions Tracer-based MRI will improve our understanding of the brain microenvironment, improve the techniques for local delivery of drugs, and highlight brain tissue engineering fields in the future.
Collapse
Affiliation(s)
- Jin Hou
- Department of Radiology, The 2nd Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Wei Wang
- Department of Radiology and Peking Magnetic Resonance Imaging Technology Research Laboratory, 3rd Hospital of Peking University, Beijing, China (mainland)
| | - Xianyue Quan
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Wen Liang
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Zhiming Li
- Department of Radiology, The 2nd Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Deji Chen
- Department of Radiology, The 2nd Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Hongbin Han
- Department of Radiology and Peking Magnetic Resonance Imaging Technology Research Laboratory, 3rd Hospital of Peking University, Beijing, China (mainland)
| |
Collapse
|
29
|
Fang Y, Dong Y, Zheng T, Du D, Wen J, Gao D, Liu L. Altered Tracer Distribution and Clearance in the Extracellular Space of the Substantia Nigra in a Rodent Model of Parkinson's Disease. Front Neurosci 2017; 11:409. [PMID: 28790882 PMCID: PMC5524830 DOI: 10.3389/fnins.2017.00409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 06/30/2017] [Indexed: 01/12/2023] Open
Abstract
The relationship between extracellular space (ECS) diffusion parameters and brain drug clearance is not well-studied, especially in the context of Parkinson's disease (PD). Therefore, we used a rodent model of PD to explore the distribution and clearance of a magnetic resonance tracer. Forty male Sprague Dawley rats were randomized into four different groups: a PD group, a Madopar group (PD + Madopar treatment), a sham group, and a control group. All rats received an injection of the extracellular tracer gadolinium-diethylene triaminepentacetic acid (Gd-DTPA) directly into the substantia nigra (SN). ECS diffusion parameters including the effective diffusion coefficient (D*), clearance coefficient (k'), ratio of the maximum distribution volume of the tracer (Vd-max%), and half-life (t1/2) were measured. We found that all parameters were significantly increased in the PD group compared to the other three groups (D*: F = 5.774, p = 0.0025; k': F = 20.00, P < 0.0001; Vd-max%: F = 12.81, P < 0.0001; and t1/2: F = 23.35, P < 0.0001). In conclusion, the PD group exhibited a wider distribution and lower clearance of the tracer compared to the other groups. Moreover, k' was more sensitive than D* for monitoring morphological and functional changes in the ECS in a rodent model of PD.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| | - Yanchao Dong
- Department of Interventional Therapy, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| | - Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| | - Jiexia Wen
- Department of Central Laboratory, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| | - Dawei Gao
- Institute of Chemical and Environmental Engineering, Yanshan UniversityQinhuangdao, China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 HospitalQinhuangdao, China
| |
Collapse
|
30
|
Lv D, Li J, Li H, Fu Y, Wang W. Imaging and Quantitative Analysis of the Interstitial Space in the Caudate Nucleus in a Rotenone-Induced Rat Model of Parkinson's Disease Using Tracer-based MRI. Aging Dis 2017; 8:1-6. [PMID: 28203477 PMCID: PMC5287382 DOI: 10.14336/ad.2016.0625] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/26/2016] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by pathological changes within several deep structures of the brain, including the substantia nigra and caudate nucleus. However, changes in interstitial fluid (ISF) flow and the microstructure of the interstitial space (ISS) in the caudate nucleus in PD have not been reported. In this study, we used tracer-based magnetic resonance imaging (MRI) to quantitatively investigate the alterations in ISS and visualize ISF flow in the caudate nucleus in a rotenone-induced rat model of PD treated with and without madopar. In the rotenone-induced rat model, the ISF flow was slowed and the tortuosity of the ISS was significantly decreased. Administration of madopar partially prevented these changes of ISS and ISF. Therefore, our data suggest that tracer-based MRI can be used to monitor the parameters related to ISF flow and ISS microstructure. It is a promising technique to investigate the microstructure and functional changes in the deep brain regions of PD.
Collapse
Affiliation(s)
- Deyong Lv
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China; 2Beijing Key Laboratory of Magnetic Resonance Imaging Device and Technique, Beijing 100191, China; 3Department of Radiology, Dongying People's Hospital of Shandong, Shandong, 257091, China
| | - Jingbo Li
- 4Department of Ultrasound, Dongying People's Hospital of Shandong, Shandong, 257091, China
| | - Hongfu Li
- 3Department of Radiology, Dongying People's Hospital of Shandong, Shandong, 257091, China
| | - Yu Fu
- 5Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wang
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China; 2Beijing Key Laboratory of Magnetic Resonance Imaging Device and Technique, Beijing 100191, China
| |
Collapse
|
31
|
Fu Y, Li Y, Guo J, Liu B, Liu H, Zhang W, Sun W, Gao Y, Ren Q, Han H. Bloodletting at Jing-well points decreases interstitial fluid flow in the thalamus of rats. J TRADIT CHIN MED 2016; 36:107-12. [PMID: 26946627 DOI: 10.1016/s0254-6272(16)30016-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To investigate the changes in the neuronal microenvironment of the middle cerebral artery (MCA) territory induced by Jing-well points bloodletting acupuncture (WPBA) and to explore the neuroprotective mechanism of WPBA in stroke. METHODS Adult male Sprague Dawley (n = 32) rats were randomly divided into four groups of eight animals each: WPBA-thalamus group (WT), WPBA-caudate nucleus group (WC), sham-control thalamus group (ST) and sham-control caudate nucleus group (SC). Animals in the WT and WC groups received 2 µL of the extracellular tracer gadolinium-diethylene triamine pentaacetic acid (Gd-DTPA) injected into the thalamus or caudate nucleus, respectively, and 12 Jing-well points in the distal ends of the rats' digits were used for WPBA. Although 2 µL of Gd-DTPA was injected into the thalamus or caudate nucleus, respectively, for animals in the two sham groups (ST and SC), no acupuncture or bloodletting was performed. Brain extracellular space and interstitial fluid flow parameters were measured using Gd-DTPA-enhanced magnetic resonance imaging. RESULTS The brain interstitial fluid flow speed was decreased in the thalamus after WPBA, with a significantly lower Gd-DTPA clearance rate and longer half-life of Gd-DTPA in the thalamus of treated rats than those in sham-control rats [WPBA-treated rats' clearance rate, (7.47 ± 3.15) x 10(-5)/s (P.= 0.009); half-life, (1.52 ± 0.13) h, P = 0.000]. By contrast, no significant changes in brain extracellular space and interstitial fluid flow parameters were detected in the caudate nucleus after WPBA (P = 0.649). In addition, no differences in the morphology of the brain extracellular space or the final distribution of the traced brain interstitial fluid were demonstrated between the WT and WC groups (P = 0.631, P = 0.970, respectively). CONCLUSION The WPBA decreased the speed of the local thalamic ISF flow in rats, which is assumed to be a beneficial protection by down-modulated the metabolic rate of the attacked neurons under stroke.
Collapse
|