1
|
Fontana F, Donato AC, Malik A, Gelain F. Unveiling Interactions between Self-Assembling Peptides and Neuronal Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26811-26823. [PMID: 39653368 DOI: 10.1021/acs.langmuir.4c02050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
The use of self-assembling peptide hydrogels in the treatment of spinal cord and brain injuries, especially when combined with adult neural stem cells, has shown great potential. To advance tissue engineering, it is essential to understand the effect of mechanochemical signaling on cellular differentiation. The elucidation of the molecular interactions at the level of the neuronal membrane still represents a promising area of investigation for many drug delivery and tissue engineering applications. An innovative molecular dynamics framework has been introduced to investigate the effect of SAP fibrils with different charges on neural membrane lipid domain dynamics. Such advance enables the in silico exploration of the biomimetic properties of SAP hydrogels and other polymeric biomaterials for tissue engineering applications.
Collapse
Affiliation(s)
- Federico Fontana
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Alice Cristina Donato
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Histology Unit, Department of Molecular Medicine, University of Padova, Padova 35121, Italy
| | - Ashish Malik
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Fabrizio Gelain
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| |
Collapse
|
2
|
Kim D, Tithof J. Lumped parameter simulations of cervical lymphatic vessels: dynamics of murine cerebrospinal fluid efflux from the skull. Fluids Barriers CNS 2024; 21:104. [PMID: 39702363 DOI: 10.1186/s12987-024-00605-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Growing evidence suggests that for rodents, a substantial fraction of cerebrospinal fluid (CSF) drains by crossing the cribriform plate into the nasopharyngeal lymphatics, eventually reaching the cervical lymphatic vessels (CLVs). Disruption of this drainage pathway is associated with various neurological disorders. METHODS We employ a lumped parameter method to numerically model CSF drainage across the cribriform plate to CLVs. Our model uses intracranial pressure as an inlet pressure and central venous blood pressure as an outlet pressure. The model incorporates initial lymphatic vessels (modeling those in the nasal region) that absorb the CSF and collecting lymphatic vessels (modeling CLVs) to transport the CSF against an adverse pressure gradient. To determine unknown parameters such as wall stiffness and valve properties, we utilize a Monte Carlo approach and validate our simulation against recent in vivo experimental measurements. RESULTS Our parameter analysis reveals the physical characteristics of CLVs. Our results suggest that the stiffness of the vessel wall and the closing state of the valve are crucial for maintaining the vessel size and volume flow rate observed in vivo. We find that a decreased contraction amplitude and frequency leads to a reduction in volume flow rate, and we test the effects of varying the different pressures acting on the CLVs. Finally, we provide evidence that branching of initial lymphatic vessels may deviate from Murray's law to reduce sensitivity to elevated intracranial pressure. CONCLUSIONS This is the first numerical study of CSF drainage through CLVs. Our comprehensive parameter analysis offers guidance for future numerical modeling of CLVs. This study also provides a foundation for understanding physiology of CSF drainage, helping guide future experimental studies aimed at identifying causal mechanisms of reduction in CLV transport and potential therapeutic approaches to enhance flow.
Collapse
Affiliation(s)
- Daehyun Kim
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis, MN, 55455, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Xu H, He J, Du H, Jing X, Liu X. Evaluation of the Choroid Plexus Epithelium Inflammation TLR4/NF-κB/NKCC1 Signal Pathway Activation in the Development of Hydrocephalus. CNS Neurosci Ther 2024; 30:e70085. [PMID: 39450988 PMCID: PMC11503839 DOI: 10.1111/cns.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Hydrocephalus is characterized by secretion, circulation, and absorption disorder of cerebrospinal fluid (CSF) with high morbidity and complication rate. The relationship between inflammation and abnormal secretion of CSF by choroid plexus epithelium (CPE) had received more attention. In this study, we aim to detect the role of Toll-like receptor 4/nuclear factor-kappa B/Na+/K+/2Cl-cotransporter 1(TLR4/NF-κB/NKCC1) signal pathway in the development of hydrocephalus. METHOD Hydrocephalus was induced in adult rats (8 weeks) by intracisternal kaolin injection, then pyrrolidinedithiocarbamate (PDTC) and bumetanide were administrated to the rats mode. Then the rat model was evaluated, and ventricular volume was calculated at different time points. Then CPE, cortex, preventricular tissue, and CSF were obtained. Protein expressions of TLR-4, NKCC/serine-threonine STE20/SPS1-related, proline-alanine-rich kinase (SPAK), pNKCC1, pSPAK, GFAP, AQP1, and AQP4 were measured by RT-PCR, western blot, and immunofluorescence (IF) stains in CPE, respectively. RESULT Our data showed that inflammation factors tumor necrosis factor-(TNF-α), interleukin 18(IL-18), and glial fibrillary acidic protein (GFAP) concentrations were significantly higher in the model group than in controls. The TLR4/NF-κB/NKCC1 signal pathway were actived by NF-κB-p65, NKCC1, pNKCC1- pSPAK complex, and Aquaporin1 (AQP1) high expression. PDTC and bumetanide use can help regular TLR4/NF-κB/NKCC1 expression and reduced AQP1 expression by down-regulate NF-B-p65 and inhibiting NKCC1, respectively. As a result, the treatment groups alleviated CPE abnormal secretion and ventricle enlargement. CONCLUSION These results confirmed that the inflammatory reaction contributes TLR4/NF-κB/NKCC1 mediated CPE abnormal secretion and consequent hydrocephalus. Regulation of TLR4/NF-κB/NKCC1 and AQP1 can prevent this process. Our study provides a strong rationale for further exploring alleviating CPE abnormal secretion as a therapeutic perspective of hydrocephalus.
Collapse
Affiliation(s)
- Hao Xu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Jiawei He
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianChina
| | - Hua Du
- Key Laboratory of High Magnetic Field and Ion Beam Physical BiologyHefei Institutes of Physical Science, CASHefeiAnhuiP. R. China
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control TechnologyHefei Institutes of Physical Science, CASHefeiAnhuiP. R. China
| | - Xiaolei Jing
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Xinfeng Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| |
Collapse
|
4
|
Lee AH, Tai SH, Huang SY, Chang LD, Chen LY, Chen YN, Hsu HH, Lee EJ. Melatonin Improves Vasogenic Edema via Inhibition to Water Channel Aquaporin-4 (AQP4) and Metalloproteinase-9 (MMP-9) Following Permanent Focal Cerebral Ischemia. Biomedicines 2024; 12:2184. [PMID: 39457496 PMCID: PMC11504272 DOI: 10.3390/biomedicines12102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Background: The efficacy of melatonin in reducing vasogenic and cytotoxic edema was investigated using a model of permanent middle cerebral artery occlusion (pMCAO). Methods: Rats underwent pMCAO, followed by intravenous administration of either melatonin (5 mg/kg) or a vehicle 10 min post-insult. Brain infarction and edema were assessed, and Western blot analyses were conducted to examine the expression levels of aquaporin-4 (AQP4), metalloproteinase-9 (MMP-9), and the neurovascular tight-junction protein ZO-1 upon sacrifice. The permeability of the blood-brain barrier (BBB) was measured using spectrophotometric quantification of Evans blue dye leakage. Results: Compared to controls, melatonin-treated rats exhibited a significant reduction in infarct volume by 26.9% and showed improved neurobehavioral outcomes (p < 0.05 for both). Melatonin treatment also led to decreased Evans blue dye extravasation and brain edema (p < 0.05 for both), along with lower expression levels of AQP4 and MMP-9 proteins and better preservation of ZO-1 protein (p < 0.05 for all). Conclusions: Therefore, melatonin offers neuroprotection against brain swelling induced by ischemia, possibly through its modulation of AQP4 and MMP-9 activities in glial cells and the extracellular matrix (ECM) during the early phase of ischemic injury.
Collapse
Affiliation(s)
- Ai-Hua Lee
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
- Department of Occupational Safety and Health, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan
| | - Shih-Huang Tai
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Sheng-Yang Huang
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Li-Der Chang
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Liang-Yi Chen
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Yu-Ning Chen
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Hao-Hsiang Hsu
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - E-Jian Lee
- Neurophysiology Laboratory, Neurosurgical Service, Departments of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
5
|
Tang G, Zhou H, Zeng C, Jiang Y, Li Y, Hou L, Liao K, Tan Z, Wu H, Tang Y, Cheng Y, Ling X, Guo Q, Xu H. Alterations of apparent diffusion coefficient from ultra high b-values in the bilateral thalamus and striatum in MRI-negative drug-resistant epilepsy. Epilepsia Open 2024; 9:1515-1525. [PMID: 38943548 PMCID: PMC11296122 DOI: 10.1002/epi4.12990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 04/01/2024] [Accepted: 05/26/2024] [Indexed: 07/01/2024] Open
Abstract
OBJECTIVE Subcortical nuclei such as the thalamus and striatum have been shown to be related to seizure modulation and termination, especially in drug-resistant epilepsy. Enhance diffusion-weighted imaging (eDWI) technique and tri-component model have been used in previous studies to calculate apparent diffusion coefficient from ultra high b-values (ADCuh). This study aimed to explore the alterations of ADCuh in the bilateral thalamus and striatum in MRI-negative drug-resistant epilepsy. METHODS Twenty-nine patients with MRI-negative drug-resistant epilepsy and 18 healthy controls underwent eDWI scan with 15 b-values (0-5000 s/mm2). The eDWI parameters including standard ADC (ADCst), pure water diffusion (D), and ADCuh were calculated from the 15 b-values. Regions-of-interest (ROIs) analyses were conducted in the bilateral thalamus, caudate nucleus, putamen, and globus pallidus. ADCst, D, and ADCuh values were compared between the MRI-negative drug-resistant epilepsy patients and controls using multivariate generalized linear models. Inter-rater reliability was assessed using the intra-class correlation coefficient (ICC) and Bland-Altman (BA) analysis. False discovery rate (FDR) method was applied for multiple comparisons correction. RESULTS ADCuh values in the bilateral thalamus, caudate nucleus, putamen, and globus pallidus in MRI-negative drug-resistant epilepsy were significantly higher than those in the healthy control subjects (all p < 0.05, FDR corrected). SIGNIFICANCE The alterations of the ADCuh values in the bilateral thalamus and striatum in MRI-negative drug-resistant epilepsy might reflect abnormal membrane water permeability in MRI-negative drug-resistant epilepsy. ADCuh might be a sensitive measurement for evaluating subcortical nuclei-related brain damage in epilepsy patients. PLAIN LANGUAGE SUMMARY This study aimed to explore the alterations of apparent diffusion coefficient calculated from ultra high b-values (ADCuh) in the subcortical nuclei such as the bilateral thalamus and striatum in MRI-negative drug-resistant epilepsy. The bilateral thalamus and striatum showed higher ADCuh in epilepsy patients than healthy controls. These findings may add new evidences of subcortical nuclei abnormalities related to water and ion hemostasis in epilepsy patients, which might help to elucidate the underlying epileptic neuropathophysiological mechanisms and facilitate the exploration of therapeutic targets.
Collapse
Affiliation(s)
- Guixian Tang
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Hailing Zhou
- Department of RadiologyCentral People's Hospital of ZhanjiangZhanjiangChina
| | - Chunyuan Zeng
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yuanfang Jiang
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Ying Li
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Lu Hou
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Kai Liao
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Zhiqiang Tan
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Huanhua Wu
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yongjin Tang
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yong Cheng
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Xueying Ling
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Qiang Guo
- Epilepsy Center, Guangdong 999 Brain HospitalAffiliated Brain Hospital of Jinan UniversityGuangzhouChina
| | - Hao Xu
- Department of Nuclear Medicine, PET/CT‐MRI Center, Center of Cyclotron and PET RadiopharmaceuticalsThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| |
Collapse
|
6
|
Sriram S, Carstens K, Dewing W, Fiacco TA. Astrocyte regulation of extracellular space parameters across the sleep-wake cycle. Front Cell Neurosci 2024; 18:1401698. [PMID: 38988660 PMCID: PMC11233815 DOI: 10.3389/fncel.2024.1401698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple subfields of neuroscience research are beginning to incorporate astrocytes into current frameworks of understanding overall brain physiology, neuronal circuitry, and disease etiology that underlie sleep and sleep-related disorders. Astrocytes have emerged as a dynamic regulator of neuronal activity through control of extracellular space (ECS) volume and composition, both of which can vary dramatically during different levels of sleep and arousal. Astrocytes are also an attractive target of sleep research due to their prominent role in the glymphatic system, a method by which toxic metabolites generated during wakefulness are cleared away. In this review we assess the literature surrounding glial influences on fluctuations in ECS volume and composition across the sleep-wake cycle. We also examine mechanisms of astrocyte volume regulation in glymphatic solute clearance and their role in sleep and wake states. Overall, findings highlight the importance of astrocytes in sleep and sleep research.
Collapse
Affiliation(s)
- Sandhya Sriram
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Kaira Carstens
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Wayne Dewing
- Undergraduate Major in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Todd A Fiacco
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
7
|
Yaghoobi Z, Seyed Bagher Nazeri SS, Asadi A, Derafsh E, Talebi Taheri A, Tamtaji Z, Dadgostar E, Rahmati-Dehkordi F, Aschner M, Mirzaei H, Tamtaji OR, Nabavizadeh F. Non-coding RNAs and Aquaporin 4: Their Role in the Pathogenesis of Neurological Disorders. Neurochem Res 2024; 49:583-596. [PMID: 38114727 DOI: 10.1007/s11064-023-04067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Abstract
Neurological disorders are a major group of non-communicable diseases affecting quality of life. Non-Coding RNAs (ncRNAs) have an important role in the etiology of neurological disorders. In studies on the genesis of neurological diseases, aquaporin 4 (AQP4) expression and activity have both been linked to ncRNAs. The upregulation or downregulation of several ncRNAs leads to neurological disorder progression by targeting AQP4. The role of ncRNAs and AQP4 in neurological disorders is discussed in this review.
Collapse
Affiliation(s)
- Zahra Yaghoobi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, I.R. of Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, I.R. of Iran
| | | | - Amir Asadi
- Psychiatry and Behavioral Sciences Research Center, School of Medicine, Addiction Institute, and Department of Psychiatry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Derafsh
- Windsor University School of Medicine, Cayon, St Kitts and Nevis
| | - Abdolkarim Talebi Taheri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Tamtaji
- Student Research Committee, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, I.R. of Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, I.R. of Iran
| | - Fatemeh Rahmati-Dehkordi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, I.R. of Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, I.R. of Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. of Iran.
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, I.R. of Iran.
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, I.R. of Iran.
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, I.R. of Iran.
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, I.R. of Iran.
| |
Collapse
|
8
|
Čivrný J, Tomáš D, Černá M. MRI of cerebral oedema in ischaemic stroke and its current use in routine clinical practice. Neuroradiology 2024; 66:305-315. [PMID: 38102491 PMCID: PMC10859334 DOI: 10.1007/s00234-023-03262-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Currently, with the knowledge of the role of collateral circulation in the development of cerebral ischaemia, traditional therapeutic windows are being prolonged, with time not being the only criterion. Instead, a more personalised approach is applied to select additional patients who might benefit from active treatment. This review briefly describes the current knowledge of the pathophysiology of the development of early ischaemic changes, the capabilities of MRI to depict such changes, and the basics of the routinely used imaging techniques broadly available for the assessment of individual phases of cerebral ischaemia, and summarises the possible clinical use of routine MR imaging, including patient selection for active treatment and assessment of the outcome on the basis of imaging.
Collapse
Affiliation(s)
- Jakub Čivrný
- Department of Radiology, Palacky University and University Hospital, Olomouc, Czech Republic.
- Fakultní nemocnice Olomouc, Radiologická klinika, Zdravotníků 248/7, 779 00, Olomouc, Czech Republic.
| | - Dorňák Tomáš
- Fakultní nemocnice Olomouc, Radiologická klinika, Zdravotníků 248/7, 779 00, Olomouc, Czech Republic
- Department of Neurology, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Marie Černá
- Department of Radiology, Palacky University and University Hospital, Olomouc, Czech Republic
- Fakultní nemocnice Olomouc, Radiologická klinika, Zdravotníků 248/7, 779 00, Olomouc, Czech Republic
| |
Collapse
|
9
|
Jazaeri SZ, Taghizadeh G, Babaei JF, Goudarzi S, Saadatmand P, Joghataei MT, Khanahmadi Z. Aquaporin 4 beyond a water channel; participation in motor, sensory, cognitive and psychological performances, a comprehensive review. Physiol Behav 2023; 271:114353. [PMID: 37714320 DOI: 10.1016/j.physbeh.2023.114353] [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: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
Aquaporin 4 (AQP4) is a protein highly expressed in the central nervous system (CNS) and peripheral nervous system (PNS) as well as various other organs, whose different sites of action indicate its importance in various functions. AQP4 has a variety of essential roles beyond water homeostasis. In this article, we have for the first time summarized different roles of AQP4 in motor and sensory functions, besides cognitive and psychological performances, and most importantly, possible physiological mechanisms by which AQP4 can exert its effects. Furthermore, we demonstrated that AQP4 participates in pathology of different neurological disorders, various effects depending on the disease type. Since neurological diseases involve a spectrum of dysfunctions and due to the difficulty of obtaining a treatment that can simultaneously affect these deficits, it is therefore suggested that future studies consider the role of this protein in different functional impairments related to neurological disorders simultaneously or separately by targeting AQP4 expression and/or polarity modulation.
Collapse
Affiliation(s)
- Seyede Zohreh Jazaeri
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of medical Sciences, Tehran, Iran
| | - Pegah Saadatmand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Innovation in Medical Education, Faculty of Medicine, Ottawa University, Ottawa, Canada.
| | - Zohreh Khanahmadi
- Department of Occupational Therapy, School of Rehabilitation Services, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Bordoni L, Thoren AE, Gutiérrez‐Jiménez E, Åbjørsbråten KS, Bjørnstad DM, Tang W, Stern M, Østergaard L, Nagelhus EA, Frische S, Ottersen OP, Enger R. Deletion of aquaporin-4 improves capillary blood flow distribution in brain edema. Glia 2023; 71:2559-2572. [PMID: 37439315 PMCID: PMC10952478 DOI: 10.1002/glia.24439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023]
Abstract
Brain edema is a feared complication to disorders and insults affecting the brain. It can be fatal if the increase in intracranial pressure is sufficiently large to cause brain herniation. Moreover, accruing evidence suggests that even slight elevations of intracranial pressure have adverse effects, for instance on brain perfusion. The water channel aquaporin-4 (AQP4), densely expressed in perivascular astrocytic endfeet, plays a key role in brain edema formation. Using two-photon microscopy, we have studied AQP4-mediated swelling of astrocytes affects capillary blood flow and intracranial pressure (ICP) in unanesthetized mice using a mild brain edema model. We found improved regulation of capillary blood flow in mice devoid of AQP4, independently of the severity of ICP increase. Furthermore, we found brisk AQP4-dependent astrocytic Ca2+ signals in perivascular endfeet during edema that may play a role in the perturbed capillary blood flow dynamics. The study suggests that astrocytic endfoot swelling and pathological signaling disrupts microvascular flow regulation during brain edema formation.
Collapse
Affiliation(s)
- Luca Bordoni
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Anna E. Thoren
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Eugenio Gutiérrez‐Jiménez
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Knut S. Åbjørsbråten
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Daniel M. Bjørnstad
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Wannan Tang
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
- Department of Neurology, NeuroclinicSt. Olavs HospitalTrondheimNorway
| | - Mette Stern
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of NeuroradiologyAarhus University HospitalAarhusDenmark
| | - Erlend A. Nagelhus
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | | | - Ole P. Ottersen
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Rune Enger
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| |
Collapse
|
11
|
Contreras-Zárate MJ, Alvarez-Eraso KLF, Jaramillo-Gómez JA, Littrell Z, Tsuji N, Ormond DR, Karam SD, Kabos P, Cittelly DM. Short-term topiramate treatment prevents radiation-induced cytotoxic edema in preclinical models of breast-cancer brain metastasis. Neuro Oncol 2023; 25:1802-1814. [PMID: 37053041 PMCID: PMC10547511 DOI: 10.1093/neuonc/noad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Brain edema is a common complication of brain metastases (BM) and associated treatment. The extent to which cytotoxic edema, the first step in the sequence that leads to ionic edema, vasogenic edema, and brain swelling, contributes to radiation-induced brain edema during BM remains unknown. This study aimed to determine whether radiation-associated treatment of BM induces cytotoxic edema and the consequences of blocking the edema in preclinical models of breast-cancer brain metastases (BCBM). METHODS Using in vitro and in vivo models, we measured astrocytic swelling, trans-electric resistance (TEER), and aquaporin 4 (AQP4) expression following radiation. Genetic and pharmacological inhibition of AQP4 in astrocytes and cancer cells was used to assess the role of AQP4 in astrocytic swelling and brain water intake. An anti-epileptic drug that blocks AQP4 function (topiramate) was used to prevent cytotoxic edema in models of BM. RESULTS Radiation-induced astrocytic swelling and transient upregulation of AQP4 occurred within the first 24 hours following radiation. Topiramate decreased radiation-induced astrocytic swelling and loss of TEER in astrocytes in vitro, and acute short-term treatment (but not continuous administration), prevented radiation-induced increase in brain water content without pro-tumorigenic effects in multiple preclinical models of BCBM. AQP4 was expressed in clinical BM and breast-cancer cell lines, but AQP4 targeting had limited direct pro-tumorigenic or radioprotective effects in cancer cells that could impact its clinical translation. CONCLUSIONS Patients with BM could find additional benefits from acute and temporary preventive treatment of radiation-induced cytotoxic edema using anti-epileptic drugs able to block AQP4 function.
Collapse
Affiliation(s)
| | - Karen L F Alvarez-Eraso
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jenny A Jaramillo-Gómez
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zachary Littrell
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nikki Tsuji
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - D Ryan Ormond
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Peter Kabos
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diana M Cittelly
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
12
|
Yang LY, Chen YR, Lee JE, Chen KW, Luh HT, Chen YT, Wang KC, Hsieh ST. Dental Pulp Stem Cell-Derived Conditioned Medium Alleviates Subarachnoid Hemorrhage-Induced Microcirculation Impairment by Promoting M2 Microglia Polarization and Reducing Astrocyte Swelling. Transl Stroke Res 2023; 14:688-703. [PMID: 36181630 PMCID: PMC10444696 DOI: 10.1007/s12975-022-01083-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/23/2022] [Accepted: 09/12/2022] [Indexed: 10/07/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) can cause severe neurological deficits and high mortality. Early brain edema following SAH contributes to the initiation of microcirculation impairment and may further lead to delayed ischemic neurologic deficit (DIND). This study aimed to investigate whether dental pulp stem cell conditioned medium (DPSC-CM) ameliorates SAH-induced microcirculation impairment and the underlying mechanisms. SAH was induced via intrathecal injection of fresh autologous blood in Wistar male adult rat. DPSC-CM or DPSC-CM + insulin growth factor-1 (IGF-1) antibody was randomly administered by intrathecal route 5 min after SAH induction. To evaluate the underlying mechanisms of DPSC-CM in the treatment of SAH, primary rat astrocyte and microglia co-cultures were challenged with hemolysate or SAH-patient CSF in the presence or absence of DPSC-CM. The results showed that in vivo, DPSC-CM treatment decreased the brain water content, improved microcirculation impairment and enhanced functional recovery at 24 h post-SAH. DPSC-CM treatment also alleviated the expressions of water channel protein aquaporin-4 (AQP4) and pro-inflammatory cytokines, and enhanced the expressions of anti-inflammatory factors in the cortical region. However, all the beneficial effects of DPSC-CM were abrogated after treatment with IGF-1 neutralizing antibody. The in vitro results further showed that DPSC-CM treatment reduced hemolysate/SAH-patient CSF-induced astrocyte swelling and promoted M2 microglia polarization, partially through IGF-1/AKT signaling. The data suggested that DPSC-CM significantly reduced brain edema and rescued microcirculation impairment with concomitant anti-inflammatory benefits after SAH, and may potentially be developed into a novel therapeutic strategy for SAH.
Collapse
Affiliation(s)
- Ling-Yu Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yong-Ren Chen
- Non-Invasive Cancer Therapy Research Institute, Taipei, Taiwan
| | - Jing-Er Lee
- Department of Neurology, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Kuo-Wei Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hui-Tzung Luh
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Tzu Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuo-Chuan Wang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
13
|
Exton J, Higgins JMG, Chen J. Acute brain slice elastic modulus decreases over time. Sci Rep 2023; 13:12826. [PMID: 37550376 PMCID: PMC10406937 DOI: 10.1038/s41598-023-40074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023] Open
Abstract
A common benchmark in the brain tissue mechanics literature is that the properties of acute brain slices should be measured within 8 h of the experimental animal being sacrificed. The core assumption is that-since there is no substantial protein degradation during this time-there will be no change to elastic modulus. This assumption overlooks the possibility of other effects (such as osmotic swelling) that may influence the mechanical properties of the tissue. To achieve consistent and accurate analysis of brain mechanics, it is important to account for or mitigate these effects. Using atomic force microscopy (AFM), tissue hydration and volume measurements, we find that acute brain slices in oxygenated artificial cerebrospinal fluid (aCSF) with a standard osmolarity of 300 mOsm/l experience rapid swelling, softening, and increases in hydration within the first 2 hours after slicing. Reductions in elastic modulus can be partly mitigated by addition of chondroitinase ABC enzyme (CHABC). Increasing aCSF osmolarity to 400 mOsm/l does not prevent softening but may hasten equilibration of samples to a point where measurements of relative elastic modulus are consistent across experiments.
Collapse
Affiliation(s)
- John Exton
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Jinju Chen
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| |
Collapse
|
14
|
Gao Y, Liu K, Zhu J. Glymphatic system: an emerging therapeutic approach for neurological disorders. Front Mol Neurosci 2023; 16:1138769. [PMID: 37485040 PMCID: PMC10359151 DOI: 10.3389/fnmol.2023.1138769] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
The functions of the glymphatic system include clearance of the metabolic waste and modulation of the water transport in the brain, and it forms a brain-wide fluid network along with cerebrospinal fluid (CSF) and interstitial fluid (ISF). The glymphatic pathway consists of periarterial influx of CSF, astrocyte-mediated interchange between ISF and CSF supported by aquaporin-4 (AQP4) on the endfeet of astrocyte around the periarterioles, and perivenous efflux of CSF. Finally, CSF is absorbed by the arachnoid granules or flows into the cervical lymphatic vessels. There is growing evidence from animal experiments that the glymphatic system dysfunction is involved in many neurological disorders, such as Alzheimer's disease, stroke, epilepsy, traumatic brain injury and meningitis. In this review, we summarize the latest progress on the glymphatic system and its driving factors, as well as changes in the glymphatic pathway in different neurological diseases. We significantly highlight the likely therapeutic approaches for glymphatic pathway in neurological diseases, and the importance of AQP4 and normal sleep architecture in this process.
Collapse
Affiliation(s)
- Ying Gao
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Kangding Liu
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
15
|
Stokum JA, Shim B, Negoita S, Tsymbalyuk N, Tsymbalyuk O, Ivanova S, Keledjian K, Bryan J, Blaustein MP, Jha RM, Kahle KT, Gerzanich V, Simard JM. Cation flux through SUR1-TRPM4 and NCX1 in astrocyte endfeet induces water influx through AQP4 and brain swelling after ischemic stroke. Sci Signal 2023; 16:eadd6364. [PMID: 37279286 PMCID: PMC10369355 DOI: 10.1126/scisignal.add6364] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Brain swelling causes morbidity and mortality in various brain injuries and diseases but lacks effective treatments. Brain swelling is linked to the influx of water into perivascular astrocytes through channels called aquaporins. Water accumulation in astrocytes increases their volume, which contributes to brain swelling. Using a mouse model of severe ischemic stroke, we identified a potentially targetable mechanism that promoted the cell surface localization of aquaporin 4 (AQP4) in perivascular astrocytic endfeet, which completely ensheathe the brain's capillaries. Cerebral ischemia increased the abundance of the heteromeric cation channel SUR1-TRPM4 and of the Na+/Ca2+ exchanger NCX1 in the endfeet of perivascular astrocytes. The influx of Na+ through SUR1-TRPM4 induced Ca2+ transport into cells through NCX1 operating in reverse mode, thus raising the intra-endfoot concentration of Ca2+. This increase in Ca2+ stimulated calmodulin-dependent translocation of AQP4 to the plasma membrane and water influx, which led to cellular edema and brain swelling. Pharmacological inhibition or astrocyte-specific deletion of SUR1-TRPM4 or NCX1 reduced brain swelling and improved neurological function in mice to a similar extent as an AQP4 inhibitor and was independent of infarct size. Thus, channels in astrocyte endfeet could be targeted to reduce postischemic brain swelling in stroke patients.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Bosung Shim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Serban Negoita
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Natalya Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph Bryan
- Pacific Northwest Diabetes Research Institute, Seattle, WA 98122, USA
| | - Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ruchira M Jha
- Department of Neurology, Barrow Neurological Institute and St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Datta A, Suthar P, Sarmah D, Jadhav P, Shah J, Katamneni M, Bhosale N, Gupta V, Bohra M, Baidya F, Rana N, Ghosh B, Kaur H, Borah A, Rathod R, Sengupta P, Bhattacharya P. Inosine attenuates post-stroke neuroinflammation by modulating inflammasome mediated microglial activation and polarization. Biochim Biophys Acta Mol Basis Dis 2023:166771. [PMID: 37286144 DOI: 10.1016/j.bbadis.2023.166771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
To date, various agents and molecules have been developed to treat post-stroke neuroinflammation; however, none of them are clinically successful. Post-stroke neuroinflammation is primarily attributed to microglial polarization as the generation of inflammasome complexes shifts microglia to their M1 phenotype and regulate the downstream cascade. Inosine, an adenosine derivative reported to maintain cellular energy homeostasis in stressed condition. Although, the exact mechanism is still unexplored, various studies have reported that it can stimulate axonal sprouting in different neurodegenerative diseases. Hence, our present study aims to decipher the molecular mechanism of inosine mediated neuroprotection by modulating inflammasome signaling towards altered microglial polarization in ischemic stroke. Inosine was administered intraperitoneally to male Sprague Dawley rats at 1 h post ischemic stroke and were further evaluated for neurodeficit score, motor coordination and long-term neuroprotection. Brains were harvested for infarct size estimation, biochemical assays and molecular studies. Inosine administration at 1 h post ischemic stroke decreased infarct size, neurodeficit score, and improved motor co-ordination. Normalization of biochemical parameters were achieved in the treatment groups. Microglial polarization towards its anti-inflammatory phenotype and modulation of inflammation were evident by relevant gene and protein expression studies. The outcome provides preliminary evidence of inosine mediated alleviation of post-stroke neuroinflammation via modulation of microglial polarization towards its anti-inflammatory form through regulating the inflammasome activation.
Collapse
Affiliation(s)
- Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Pramod Suthar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Poonam Jadhav
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Jinagna Shah
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Mounika Katamneni
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Nikhil Bhosale
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Vishal Gupta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Mariya Bohra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Falguni Baidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Nikita Rana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Bijoyani Ghosh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar 788011, Assam, India
| | - Rajeshwari Rathod
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
17
|
Oue H, Yamazaki Y, Qiao W, Yuanxin C, Ren Y, Kurti A, Shue F, Parsons TM, Perkerson RB, Kawatani K, Wang N, Starling SC, Roy B, Mosneag IE, Aikawa T, Holm ML, Liu CC, Inoue Y, Sullivan PM, Asmann YW, Kim BY, Bu G, Kanekiyo T. LRP1 in vascular mural cells modulates cerebrovascular integrity and function in the presence of APOE4. JCI Insight 2023; 8:e163822. [PMID: 37036005 PMCID: PMC10132158 DOI: 10.1172/jci.insight.163822] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/17/2023] [Indexed: 04/11/2023] Open
Abstract
Cerebrovasculature is critical in maintaining brain homeostasis; its dysregulation often leads to vascular cognitive impairment and dementia (VCID) during aging. VCID is the second most prevalent cause of dementia in the elderly, after Alzheimer's disease (AD), with frequent cooccurrence of VCID and AD. While multiple factors are involved in the pathogenesis of AD and VCID, APOE4 increases the risk for both diseases. A major apolipoprotein E (apoE) receptor, the low-density lipoprotein receptor-related protein 1 (LRP1), is abundantly expressed in vascular mural cells (pericytes and smooth muscle cells). Here, we investigated how deficiency of vascular mural cell LRP1 affects the cerebrovascular system and cognitive performance using vascular mural cell-specific Lrp1-KO mice (smLrp1-/-) in a human APOE3 or APOE4 background. We found that spatial memory was impaired in the 13- to 16-month-old APOE4 smLrp1-/- mice but not in the APOE3 smLrp1-/- mice, compared with their respective littermate control mice. These disruptions in the APOE4 smLrp1-/- mice were accompanied with excess paravascular glial activation and reduced cerebrovascular collagen IV. In addition, blood-brain barrier (BBB) integrity was disrupted in the APOE4 smLrp1-/- mice. Together, our results suggest that vascular mural cell LRP1 modulates cerebrovasculature integrity and function in an APOE genotype-dependent manner.
Collapse
Affiliation(s)
| | | | | | | | - Yingxue Ren
- Department of Quantitative Health Sciences, and
| | | | - Francis Shue
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Tammee M. Parsons
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Ralph B. Perkerson
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | | | | | | | | | | | | | | | - Patrick M. Sullivan
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Betty Y.S. Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Takahisa Kanekiyo
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
18
|
Marsan E, Velmeshev D, Ramsey A, Patel RK, Zhang J, Koontz M, Andrews MG, de Majo M, Mora C, Blumenfeld J, Li AN, Spina S, Grinberg LT, Seeley WW, Miller BL, Ullian EM, Krummel MF, Kriegstein AR, Huang EJ. Astroglial toxicity promotes synaptic degeneration in the thalamocortical circuit in frontotemporal dementia with GRN mutations. J Clin Invest 2023; 133:e164919. [PMID: 36602862 PMCID: PMC10014110 DOI: 10.1172/jci164919] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Mutations in the human progranulin (GRN) gene are a leading cause of frontotemporal lobar degeneration (FTLD). While previous studies implicate aberrant microglial activation as a disease-driving factor in neurodegeneration in the thalamocortical circuit in Grn-/- mice, the exact mechanism for neurodegeneration in FTLD-GRN remains unclear. By performing comparative single-cell transcriptomics in the thalamus and frontal cortex of Grn-/- mice and patients with FTLD-GRN, we have uncovered a highly conserved astroglial pathology characterized by upregulation of gap junction protein GJA1, water channel AQP4, and lipid-binding protein APOE, and downregulation of glutamate transporter SLC1A2 that promoted profound synaptic degeneration across the two species. This astroglial toxicity could be recapitulated in mouse astrocyte-neuron cocultures and by transplanting induced pluripotent stem cell-derived astrocytes to cortical organoids, where progranulin-deficient astrocytes promoted synaptic degeneration, neuronal stress, and TDP-43 proteinopathy. Together, these results reveal a previously unappreciated astroglial pathology as a potential key mechanism in neurodegeneration in FTLD-GRN.
Collapse
Affiliation(s)
| | - Dmitry Velmeshev
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | | | - Mark Koontz
- Department of Ophthalmology, UCSF, San Francisco, California, USA
| | - Madeline G. Andrews
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, USA
| | - Martina de Majo
- Department of Ophthalmology, UCSF, San Francisco, California, USA
| | | | | | - Alissa N. Li
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, California, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, California, USA
| | - Lea T. Grinberg
- Department of Pathology
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, California, USA
| | - William W. Seeley
- Department of Pathology
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, California, USA
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, UCSF, San Francisco, California, USA
| | - Erik M. Ullian
- Department of Ophthalmology, UCSF, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | | | - Arnold R. Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | - Eric J. Huang
- Department of Pathology
- ImmunoX Initiative, and
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
- Pathology Service, San Francisco Veterans Health Care System, San Francisco, California, USA
| |
Collapse
|
19
|
Ishida Y, Nosaka M, Ishigami A, Kondo T. Forensic application of aquaporins. Leg Med (Tokyo) 2023; 63:102249. [PMID: 37060638 DOI: 10.1016/j.legalmed.2023.102249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/04/2023] [Indexed: 04/03/2023]
Abstract
Aquaporins (AQPs) are a family of water channel proteins that primarily elicit the basic functions of water transport and osmotic homeostasis. To date, at least 17 mammalian AQPs have been identified, AQP-0 to -12 have been found in higher orders including human, and AQP-13 to -16 have been described in older lineages. Moreover, these proteins have recently been shown to regulate many biological processes through unique activities, such as cell proliferation, migration, apoptosis, and mitochondrial metabolism. Several studies have focused on the involvement of AQPs in cell biology aspect, showing that they are involved in a variety of physiological processes and pathophysiological conditions. Furthermore, in the field of forensic medicine, studies on whether AQPs can be a useful marker for diagnosing various causes of death have been conducted using autopsy samples and animal experiments, which have produced interesting results. Herein, we review certain observations regarding AQPs and discuss their potential to contribute to the future practice of forensic research.
Collapse
|
20
|
Harris WJ, Asselin MC, Hinz R, Parkes LM, Allan S, Schiessl I, Boutin H, Dickie BR. In vivo methods for imaging blood-brain barrier function and dysfunction. Eur J Nucl Med Mol Imaging 2023; 50:1051-1083. [PMID: 36437425 PMCID: PMC9931809 DOI: 10.1007/s00259-022-05997-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/09/2022] [Indexed: 11/29/2022]
Abstract
The blood-brain barrier (BBB) is the interface between the central nervous system and systemic circulation. It tightly regulates what enters and is removed from the brain parenchyma and is fundamental in maintaining brain homeostasis. Increasingly, the BBB is recognised as having a significant role in numerous neurological disorders, ranging from acute disorders (traumatic brain injury, stroke, seizures) to chronic neurodegeneration (Alzheimer's disease, vascular dementia, small vessel disease). Numerous approaches have been developed to study the BBB in vitro, in vivo, and ex vivo. The complex multicellular structure and effects of disease are difficult to recreate accurately in vitro, and functional aspects of the BBB cannot be easily studied ex vivo. As such, the value of in vivo methods to study the intact BBB cannot be overstated. This review discusses the structure and function of the BBB and how these are affected in diseases. It then discusses in depth several established and novel methods for imaging the BBB in vivo, with a focus on MRI, nuclear imaging, and high-resolution intravital fluorescence microscopy.
Collapse
Affiliation(s)
- William James Harris
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Laura Michelle Parkes
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Stuart Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Herve Boutin
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
| | - Ben Robert Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Contreras-Zárate MJ, Alvarez-Eraso KL, Jaramillo-Gómez JA, Littrell Z, Tsuji N, Ormond DR, Karam SD, Kabos P, Cittelly DM. Short-term Topiramate treatment prevents radiation-induced cytotoxic edema in preclinical models of breast-cancer brain metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528559. [PMID: 36824740 PMCID: PMC9948992 DOI: 10.1101/2023.02.14.528559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Background Brain edema is a common complication of brain metastases (BM) and associated treatment. The extent to which cytotoxic edema, the first step in the sequence that leads to ionic edema, vasogenic edema and brain swelling, contributes to radiation-induced brain edema during BM remains unknown. This study aimed to determine whether radiation-associated treatment of BM induces cytotoxic edema and the consequences of blocking the edema in pre-clinical models of breast cancer brain metastases (BCBM). Methods Using in vitro and in vivo models, we measured astrocytic swelling, trans-electric resistance (TEER) and aquaporin 4 (AQP4) expression following radiation. Genetic and pharmacological inhibition of AQP4 in astrocytes and cancer cells was used to assess the role of AQP4 in astrocytic swelling and brain water intake. An anti-epileptic drug that blocks AQP4 function (topiramate) was used to prevent cytotoxic edema in models of BM. Results Radiation-induced astrocytic swelling and transient upregulation of AQP4 within the first 24 hours following radiation. Topiramate decreased radiation-induced astrocytic swelling, loss of TEER in astrocytes in vitro , and acute short term treatment (but not continuous administration), prevented radiation-induced increase in brain water content without pro-tumorigenic effects in multiple pre-clinical models of BCBM. AQP4 was expressed in clinical BM and breast cancer cell lines, but AQP4 targeting had limited direct pro-tumorigenic or radioprotective effects in cancer cells that could impact its clinical translation. Conclusions Patients with BM could find additional benefits from acute and temporary preventive treatment of radiation-induced cytotoxic edema using anti-epileptic drugs able to block AQP4 function. Key points Radiation induces cytotoxic edema via acute dysregulation of AQP4 in astrocytes in preclinical models of BM. Pharmacologic blockage of AQP4 function prevents water intake, astrocytic swelling and restores TEER in vitro. Pre-treatment with single-dose Topiramate prevents brain radiation-induced brain edema without direct tumor effects in pre-clinical models of BCBM. IMPORTANCE OF THE STUDY In this study we describe a novel role for astrocytic swelling and cytotoxic edema in the progression of radiation-induced brain edema during BM treatment. While radiation-induced edema has been fully attributed to the disruption of the blood-brain barrier (BBB) and ensuing vasogenic effects, our results suggest that cytotoxic edema affecting astrocytes in the acute setting plays an important role in the progression of brain edema during BM standard of care. Current standard of care for brain edema involves pre-treatment with steroids and the use of bevacizumab only after clinically significant edema develops. Both interventions are presumed to target vasogenic edema. This study suggests that patients with BM could find additional benefits from acute and temporary preventive treatment of radiation-induced cytotoxic edema using an already FDA-approved anti-epileptic drug. Such early prevention strategy can be easily clinically implemented with the goal of minimizing treatment-related toxicities.
Collapse
|
22
|
Sarmah D, Sarkar A, Datta A, Ghosh B, Rana N, Sahu S, Gupta V, Thongire V, Chaudhary A, Vadak N, Kaur H, Raut S, Singh U, Borah A, Bhattacharya P. Cardiolipin-Mediated Alleviation of Mitochondrial Dysfunction Is a Neuroprotective Effect of Statin in Animal Model of Ischemic Stroke. ACS Chem Neurosci 2023; 14:709-724. [PMID: 36706354 DOI: 10.1021/acschemneuro.2c00645] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In clinical settings, the benefit of statin for stroke is debatable as regular statin users may suffer from myalgia, statin-associated myopathy (SAM), and rarely rhabdomyolysis. Studies suggest that patients on statin therapy show lesser vulnerability toward ischemic stroke and post-stroke frailty. Both pre- and post-treatment benefits of statin have been reported as evident by its neuroprotective effects in both cases. As mitochondrial dysfunction following stroke is the fulcrum for neuronal death, we hereby explore the role of statin in alleviating mitochondrial dysfunction by regulating the mitochondrial dynamics. In the present study, we intend to evaluate the role of statin in modulating cardiolipin-mediated mitochondrial functionality and further providing a therapeutic rationale for repurposing statins either as preventive or an adjunctive therapy for stroke.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Sarkar
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Aishika Datta
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Bijoyani Ghosh
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Nikita Rana
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Shubhrakanta Sahu
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Vishal Gupta
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Vrushali Thongire
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Antra Chaudhary
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Namrata Vadak
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Harpreet Kaur
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Swapnil Raut
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Upasna Singh
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam 788011, India
| | - Pallab Bhattacharya
- Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
23
|
Endothelial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032909. [PMID: 36769234 PMCID: PMC9918222 DOI: 10.3390/ijms24032909] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The cerebral vascular system stringently regulates cerebral blood flow (CBF). The components of the blood-brain barrier (BBB) protect the brain from pathogenic infections and harmful substances, efflux waste, and exchange substances; however, diseases develop in cases of blood vessel injuries and BBB dysregulation. Vascular pathology is concurrent with the mechanisms underlying aging, Alzheimer's disease (AD), and vascular dementia (VaD), which suggests its involvement in these mechanisms. Therefore, in the present study, we reviewed the role of vascular dysfunction in aging and neurodegenerative diseases, particularly AD and VaD. During the development of the aforementioned diseases, changes occur in the cerebral blood vessel morphology and local cells, which, in turn, alter CBF, fluid dynamics, and vascular integrity. Chronic vascular inflammation and blood vessel dysregulation further exacerbate vascular dysfunction. Multitudinous pathogenic processes affect the cerebrovascular system, whose dysfunction causes cognitive impairment. Knowledge regarding the pathophysiology of vascular dysfunction in neurodegenerative diseases and the underlying molecular mechanisms may lead to the discovery of clinically relevant vascular biomarkers, which may facilitate vascular imaging for disease prevention and treatment.
Collapse
|
24
|
Aquaporins Display a Diversity in their Substrates. J Membr Biol 2023; 256:1-23. [PMID: 35986775 DOI: 10.1007/s00232-022-00257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/13/2022] [Indexed: 02/07/2023]
Abstract
Aquaporins constitute a family of transmembrane proteins that function to transport water and other small solutes across the cell membrane. Aquaporins family members are found in diverse life forms. Aquaporins share the common structural fold consisting of six transmembrane alpha helices with a central water-transporting channel. Four such monomers assemble together to form tetramers as their biological unit. Initially, aquaporins were discovered as water-transporting channels, but several studies supported their involvement in mediating the facilitated diffusion of different solutes. The so-called water channel is able to transport a variety of substrates ranging from a neutral molecule to a charged molecule or a small molecule to a bulky molecule or even a gas molecule. This article gives an overview of a diverse range of substrates conducted by aquaporin family members. Prime focus is on human aquaporins where aquaporins show a wide tissue distribution and substrate specificity leading to various physiological functions. This review also highlights the structural mechanisms leading to the transport of water and glycerol. More research is needed to understand how one common fold enables the aquaporins to transport an array of solutes.
Collapse
|
25
|
Li X, Yang B. Non-Transport Functions of Aquaporins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:65-80. [PMID: 36717487 DOI: 10.1007/978-981-19-7415-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Although it has been more than 20 years since the first aquaporin was discovered, the specific functions of many aquaporins are still under investigation, because various mice lacking aquaporins have no significant phenotypes. And in many studies, the function of aquaporin is not directly related to its transport function. Therefore, this chapter will focus on some unexpected functions of aquaporins, such the decreased tumor angiogenesis in AQP1 knockout mice, and AQP1 promotes cell migration, possibly by accelerating the water transport in lamellipodia of migrating cells. AQP transports glycerol, and water regulates glycerol content in epidermis and fat, thereby regulating skin hydration/biosynthesis and fat metabolism. AQPs may also be involved in neural signal transduction, cell volume regulation, and organelle physiology. AQP1, AQP3, and AQP5 are also involved in cell proliferation. In addition, AQPs have also been reported to play roles in inflammation in various tissues and organs. The functions of these AQPs may not depend on the permeability of small molecules such as water and glycerol, suggesting AQPs may play more roles in different biological processes in the body.
Collapse
Affiliation(s)
- Xiaowei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
26
|
Xiao M, Hou J, Xu M, Li S, Yang B. Aquaporins in Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:99-124. [PMID: 36717489 DOI: 10.1007/978-981-19-7415-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aquaporins (AQPs) mediate water flux between the four distinct water compartments in the central nervous system (CNS). In the present chapter, we mainly focus on the expression and function of the nine AQPs expressed in the CNS, which include five members of aquaporin subfamily: AQP1, AQP4, AQP5, AQP6, and AQP8; three members of aquaglyceroporin subfamily: AQP3, AQP7, and AQP9; and one member of superaquaporin subfamily: AQP11. In addition, AQP1, AQP2, and AQP4 expressed in the peripheral nervous system are also reviewed. AQP4, the predominant water channel in the CNS, is involved both in the astrocyte swelling of cytotoxic edema and the resolution of vasogenic edema and is of pivotal importance in the pathology of brain disorders such as neuromyelitis optica, brain tumors, and neurodegenerative disorders. Moreover, AQP4 has been demonstrated as a functional regulator of recently discovered glymphatic system that is a main contributor to clearance of toxic macromolecule from the brain. Other AQPs are also involved in a variety of important physiological and pathological process in the brain. It has been suggested that AQPs could represent an important target in treatment of brain disorders like cerebral edema. Future investigations are necessary to elucidate the pathological significance of AQPs in the CNS.
Collapse
Affiliation(s)
- Ming Xiao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jiaoyu Hou
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengmeng Xu
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shao Li
- Department of Physiology, Dalian Medical University, Dalian, China
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
27
|
Cryptococcal Immune Reconstitution Inflammatory Syndrome: From Clinical Studies to Animal Experiments. Microorganisms 2022; 10:microorganisms10122419. [PMID: 36557672 PMCID: PMC9780901 DOI: 10.3390/microorganisms10122419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Cryptococcus neoformans is an encapsulated pathogenic fungus that initially infects the lung but can migrate to the central nervous system (CNS), resulting in meningoencephalitis. The organism causes the CNS infection primarily in immunocompromised individuals including HIV/AIDS patients, but also, rarely, in immunocompetent individuals. In HIV/AIDS patients, limited inflammation in the CNS, due to impaired cellular immunity, cannot efficiently clear a C. neoformans infection. Antiretroviral therapy (ART) can rapidly restore cellular immunity in HIV/AIDS patients. Paradoxically, ART induces an exaggerated inflammatory response, termed immune reconstitution inflammatory syndrome (IRIS), in some HIV/AIDS patients co-infected with C. neoformans. A similar excessive inflammation, referred to as post-infectious inflammatory response syndrome (PIIRS), is also frequently seen in previously healthy individuals suffering from cryptococcal meningoencephalitis. Cryptococcal IRIS and PIIRS are life-threatening complications that kill up to one-third of affected people. In this review, we summarize the inflammatory responses in the CNS during HIV-associated cryptococcal meningoencephalitis. We overview the current understanding of cryptococcal IRIS developed in HIV/AIDS patients and cryptococcal PIIRS occurring in HIV-uninfected individuals. We also describe currently available animal models that closely mimic aspects of cryptococcal IRIS observed in HIV/AIDS patients.
Collapse
|
28
|
Walch E, Fiacco TA. Honey, I shrunk the extracellular space: Measurements and mechanisms of astrocyte swelling. Glia 2022; 70:2013-2031. [PMID: 35635369 PMCID: PMC9474570 DOI: 10.1002/glia.24224] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022]
Abstract
Astrocyte volume fluctuation is a physiological phenomenon tied closely to the activation of neural circuits. Identification of underlying mechanisms has been challenging due in part to use of a wide range of experimental approaches that vary between research groups. Here, we first review the many methods that have been used to measure astrocyte volume changes directly or indirectly. While the field has recently shifted towards volume analysis using fluorescence microscopy to record cell volume changes directly, established metrics corresponding to extracellular space dynamics have also yielded valuable insights. We then turn to analysis of mechanisms of astrocyte swelling derived from many studies, with a focus on volume changes tied to increases in extracellular potassium concentration ([K+ ]o ). The diverse methods that have been utilized to generate the external [K+ ]o environment highlight multiple scenarios of astrocyte swelling mediated by different mechanisms. Classical potassium buffering theories are tempered by many recent studies that point to different swelling pathways optimized at particular [K+ ]o and that depend on local/transient versus more sustained increases in [K+ ]o .
Collapse
Affiliation(s)
- Erin Walch
- Division of Biomedical Sciences, School of MedicineUniversity of California, RiversideRiversideCaliforniaUSA
| | - Todd A. Fiacco
- Department of Molecular, Cell and Systems BiologyUniversity of California, RiversideRiversideCaliforniaUSA
- Center for Glial‐Neuronal InteractionsUniversity of California, RiversideRiversideCaliforniaUSA
| |
Collapse
|
29
|
Lu H, Ashiqueali R, Lin CI, Walchale A, Clendaniel V, Matheson R, Fisher M, Lo EH, Selim M, Shehadah A. Histone Deacetylase 3 Inhibition Decreases Cerebral Edema and Protects the Blood–Brain Barrier After Stroke. Mol Neurobiol 2022; 60:235-246. [DOI: 10.1007/s12035-022-03083-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
Abstract
AbstractWe have previously shown that selective inhibition of histone deacetylase 3 (HDAC3) decreases infarct volume and improves long-term functional outcomes after stroke. In this study, we examined the effects of HDAC3 inhibition on cerebral edema and blood–brain barrier (BBB) leakage and explored its underlying mechanisms. Adult male Wistar rats were subjected to 2-h middle cerebral artery occlusion (MCAO) and randomly treated i.p. with either vehicle or a selective HDAC3 inhibitor (RGFP966) at 2 and 24 h after stroke. Modified neurological severity scores (mNSS) were calculated at 2 h, 1 day, and 3 days. H&E, Evans blue dye (EBD) assay, and fluorescein isothiocyanate (FITC)-dextran were employed to assess cerebral edema and BBB leakage. Western blot for matrix metalloproteinase-9 (MMP9), MMP-9 zymography, and immunostaining for HDAC3, GFAP, Iba-1, albumin, aquaporin-4, claudin-5, ZO-1, and NF-kB were performed. Early RGFP966 administration decreased cerebral edema (p = 0.002) and BBB leakage, as measured by EBD assay, FITC-dextran, and albumin extravasation (p < 0.01). RGFP966 significantly increased tight junction proteins (claudin-5 and ZO-1) in the peri-infarct area. RGFP966 also significantly decreased HDAC3 in GFAP + astrocytes, which correlated with better mNSS (r = 0.67, p = 0.03) and decreased cerebral edema (r = 0.64, p = 0.04). RGFP966 decreased aquaporin-4 in GFAP + astrocytes (p = 0.002), as well as, the inflammatory markers Iba-1, NF-kB, and MMP9 in the ischemic brain (p < 0.05). Early HDAC3 inhibition decreases cerebral edema and BBB leakage. BBB protection by RGFP966 is mediated in part by the upregulation of tight junction proteins, downregulation of aquaporin-4 and HDAC3 in astrocytes, and decreased neuroinflammation.
Collapse
|
30
|
Zhu A, Lin Y, Hu X, Lin Z, Lin Y, Xie Q, Ni S, Cheng H, Lu Q, Lai S, Pan G, Chen X, Pang W, Liu C. Treadmill exercise decreases cerebral edema in rats with local cerebral infarction by modulating AQP4 polar expression through the caveolin-1/TRPV4 signaling pathway. Brain Res Bull 2022; 188:155-168. [PMID: 35961528 DOI: 10.1016/j.brainresbull.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/16/2022] [Accepted: 08/06/2022] [Indexed: 11/02/2022]
Abstract
Rehabilitation therapy is beneficial for patients with ischemic stroke. Our previous study showed that treadmill training is conducive to neurological function in rats that underwent middle cerebral artery occlusion (MCAO). However, whether exercise benefits cerebral edema and the underlying mechanism remain unclear. This study investigated the influence of treadmill exercise on brain edema and the mechanism of its formation and elimination. The MCAO model was established with Sprague-Dawley (SD) rats, and lentivirus-mediated caveolin-1 shRNA was used to investigate the role of caveolin-1 in brain edema. As expected, we found that treadmill exercise has a beneficial effect on brain edema after stroke. Training led to a significant increase in the expression of caveolin-1 and TRPV4; and reduced brain water content and blood-brain barrier (BBB) damage. This treatment also changed the localization of aquaporin-4 (AQP4). Moreover, the effect of treadmill training on the polar expression of AQP4 differed over time. The results showed that early treadmill training inhibited the polar expression of AQP4, and later promoted its expression. However, the rats that were injected with the caveolin-1 shRNA lentivirus exhibited enhanced edema. Caveolin-1 shRNA eliminated the protective effect induced by exercise, which is consistent with the downregulation of TRPV4 expression. The findings indicate that treadmill training improves brain edema through the caveolin-1/TRPV4/AQP4 pathway.
Collapse
Affiliation(s)
- Anqi Zhu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Yao Lin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Xuanbo Hu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Zaizai Lin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Yongqiang Lin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Qingfeng Xie
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Shaobo Ni
- The Third Affiliated Hospital of Wenzhou Medical University, No. 108, Wansong Road, Ruian, Zhejiang, China
| | - Hui Cheng
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Qiaoya Lu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Shanshan Lai
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China
| | - Guoyuan Pan
- Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, Zhejiang, China
| | - Xiang Chen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China.
| | - Wei Pang
- The Third Affiliated Hospital of Jiamusi University, No. 419, Dexiang Street, Jiamusi, Heilongjiang, China.
| | - Chan Liu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuan West Road, Wenzhou, Zhejiang, China.
| |
Collapse
|
31
|
The glymphatic system: implications for drugs for central nervous system diseases. Nat Rev Drug Discov 2022; 21:763-779. [PMID: 35948785 DOI: 10.1038/s41573-022-00500-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2022] [Indexed: 12/14/2022]
Abstract
In the past decade, evidence for a fluid clearance pathway in the central nervous system known as the glymphatic system has grown. According to the glymphatic system concept, cerebrospinal fluid flows directionally through the brain and non-selectively clears the interstitium of metabolic waste. Importantly, the glymphatic system may be modulated by particular drugs such as anaesthetics, as well as by non-pharmacological factors such as sleep, and its dysfunction has been implicated in central nervous system disorders such as Alzheimer disease. Although the glymphatic system is best described in rodents, reports using multiple neuroimaging modalities indicate that a similar transport system exists in the human brain. Here, we overview the evidence for the glymphatic system and its role in disease and discuss opportunities to harness the glymphatic system therapeutically; for example, by improving the effectiveness of intrathecally delivered drugs.
Collapse
|
32
|
Alanazi AH, Adil MS, Lin X, Chastain DB, Henao-Martínez AF, Franco-Paredes C, Somanath PR. Elevated Intracranial Pressure in Cryptococcal Meningoencephalitis: Examining Old, New, and Promising Drug Therapies. Pathogens 2022; 11:783. [PMID: 35890028 PMCID: PMC9321092 DOI: 10.3390/pathogens11070783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/02/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
Despite the availability of effective antifungal therapy, cryptococcal meningoencephalitis (CM) remains associated with elevated mortality. The spectrum of symptoms associated with the central nervous system (CNS) cryptococcosis is directly caused by the high fungal burden in the subarachnoid space and the peri-endothelial space of the CNS vasculature, which results in intracranial hypertension (ICH). Management of intracranial pressure (ICP) through aggressive drainage of cerebrospinal fluid by lumbar puncture is associated with increased survival. Unfortunately, these procedures are invasive and require specialized skills and supplies that are not readily available in resource-limited settings that carry the highest burden of CM. The institution of pharmacologic therapies to reduce the production or increase the resorption of cerebrospinal fluid would likely improve clinical outcomes associated with ICH in patients with CM. Here, we discuss the potential role of multiple pharmacologic drug classes such as diuretics, corticosteroids, and antiepileptic agents used to decrease ICP in various neurological conditions as potential future therapies for CM.
Collapse
Affiliation(s)
- Abdulaziz H. Alanazi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30902, USA; (A.H.A.); (M.S.A.)
- Research Department, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Mir S. Adil
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30902, USA; (A.H.A.); (M.S.A.)
- Research Department, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA;
| | - Daniel B. Chastain
- Department of Clinical and Administrative Pharmacy, UGA College of Pharmacy, SWGA Clinical Campus, Phoebe Putney Memorial Hospital, Albany, GA 31701, USA;
| | - Andrés F. Henao-Martínez
- Division of Infectious Diseases, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (A.F.H.-M.); (C.F.-P.)
| | - Carlos Franco-Paredes
- Division of Infectious Diseases, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (A.F.H.-M.); (C.F.-P.)
- Hospital Infantil de México, Federico Gómez, Ciudad de México 06720, Mexico
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30902, USA; (A.H.A.); (M.S.A.)
- Research Department, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
33
|
TNF-α induces AQP4 overexpression in astrocytes through the NF-κB pathway causing cellular edema and apoptosis. Biosci Rep 2022; 42:230993. [PMID: 35260880 PMCID: PMC8935387 DOI: 10.1042/bsr20212224] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
Aquaporin 4 (AQP4) is highly expressed on astrocytes and is critical for controlling brain water transport in neurological diseases. Tumor necrosis factor (TNF)-α is a common cytokine found in disease microenvironment. The aim of this study was to determine whether TNF-α can regulate the expression of AQP4 in astrocytes. Primary astrocyte cultures were treated with different concentrations of TNF-α and the cell viability was assessed through cell counting kit-8 assay and AQP4 expression was detected by qPCR, western blots, and immunofluorescence assays. The activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway was detected by western blot. Further, dual-luciferase reporting system and chromatin immunoprecipitation were used to detect the transcriptional regulation of AQP4 by p65. These experiments demonstrated that treatment with TNF-α can lead to astrocyte edema and an increase in AQP4 expression. Following TNF-α treatment, the expression levels of P-IKKα/β-IκBα and P-p65 increased significantly over time. The results of the dual-luciferase reporter system and chromatin immunoprecipitation assays revealed that p65 protein and AQP4 promoter had a robust binding effect after TNF-α treatment, and the NF-κB pathway inhibitor, BAY 11-7082 could inhibit these effects of TNF-α. The expression level of AQP4 was significantly decreased upon p65 interference, while the astrocyte viability was significantly increased compared to that in the TNF-α only group. In conclusion, TNF-α activated NF-κB pathway, which promoted the binding of p65 to the AQP4 gene promoter region, and enhanced AQP4 expression, ultimately reducing astrocyte viability and causing cell edema.
Collapse
|
34
|
Post-stroke Impairment of the Blood–Brain Barrier and Perifocal Vasogenic Edema Is Alleviated by Endovascular Mesenchymal Stem Cell Administration: Modulation of the PKCδ/MMP9/AQP4-Mediated Pathway. Mol Neurobiol 2022; 59:2758-2775. [DOI: 10.1007/s12035-022-02761-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022]
|
35
|
Wagner K, Unger L, Salman MM, Kitchen P, Bill RM, Yool AJ. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int J Mol Sci 2022; 23:1388. [PMID: 35163313 PMCID: PMC8836214 DOI: 10.3390/ijms23031388] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The aquaporins (AQPs) are a family of small integral membrane proteins that facilitate the bidirectional transport of water across biological membranes in response to osmotic pressure gradients as well as enable the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body. Here, we review their key roles in fluid homeostasis, glandular secretions, signal transduction and sensation, barrier function, immunity and inflammation, cell migration, and angiogenesis. Evidence from a wide variety of studies now supports a view of the functions of AQPs being much more complex than simply mediating the passive flow of water across biological membranes. The discovery and development of small-molecule AQP inhibitors for research use and therapeutic development will lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.
Collapse
Affiliation(s)
- Kim Wagner
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Mootaz M. Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Roslyn M. Bill
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
36
|
Kozler P, Marešová D, Pokorný J. Effect of methylprednisolone on experimental brain edema in rats - own experience reviewed. Physiol Res 2021; 70:S289-S300. [PMID: 35099248 DOI: 10.33549/physiolres.934818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Brain edema - a frequently fatal pathological state in which brain volume increases resulting in intracranial pressure elevation - can result from almost any insult to the brain, including traumatic brain injury. For many years, the objective of experimental studies was to find a method to prevent the development of brain edema at the onset. From this perspective, the use of methylprednisolone (MP) appears promising. High molecular MP (MW>50 kDa) can be incorporated into the brain - in the conditions of the experimental model - either by osmotic blood-brain barrier disruption (BBBd) or during the induction of cellular edema by water intoxication (WI) - a condition that increases the BBB permeability. The time window for administration of the MP should be at the earliest stages of edema. The neuroprotective effect of MP on the permeability of cytoplasmatic membranes of neuronal populations was proved. MP was administrated in three alternative ways: intraperitoneally during the induction of cytotoxic edema or immediately after finishing cytotoxic edema induction in a dose of 100 mg/kg b.w.; into the internal carotid artery within 2 h after finishing cytotoxic edema induction in a dose of 50 mg/kg b.w.; into internal carotid artery 10 min after edema induction by BBBd in a dose of 50 mg/kg b.w.
Collapse
Affiliation(s)
- P Kozler
- Institute of Physiology, First Faculty of Medicine, Charles University, Praha 2, Czech Republic.
| | | | | |
Collapse
|
37
|
Guo S, Song Z, He J, Yin G, Zhu J, Liu H, Yang L, Ji X, Xu X, Liu Z, Liu J. Akt/Aquaporin-4 Signaling Aggravates Neuropathic Pain by Activating Astrocytes after Spinal Nerve Ligation in Rats. Neuroscience 2021; 482:116-131. [PMID: 34942314 DOI: 10.1016/j.neuroscience.2021.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
Aquaporins (AQPs) play critical physiological roles in water balance in the central nervous system (CNS). Aquaporin-4 (AQP4), the principal aquaporin expressed in the CNS, has been implicated in the processing of sensory and pain transmission. Akt signaling is also involved in pain mediation, such as neuroinflammatory pain and bone cancer pain. Previously, we found that expression of AQP4 and p-Akt was altered in the rat spinal cord after spinal nerve ligation (SNL). Here, we further investigated the effects of the AQP4 and Akt pathways in the spinal dorsal horn (SDH) on the pathogenesis of neuropathic pain (NP). Spinal AQP4 was significantly upregulated after SNL and was primarily expressed in astrocytes in the SDH. Inhibition of AQP4 with TGN-020 attenuated the development and maintenance of NP by inhibiting glial activation and anti-neuroinflammatory mechanisms. Moreover, inhibition of AQP4 suppressed astrocyte activation both in the SDH and in primary cultures. Similar to AQP4, we found that p-Akt was also significantly elevated after SNL. Inhibition of Akt with MK2206 suppressed AQP4 upregulation and astrocyte activation both in vivo and in vitro. Furthermore, Akt blockade with MK2206 alleviated NP in the early and late phases after SNL. These results elucidate the mechanisms involved in the roles of Akt/AQP4 signaling in the development and maintenance of NP. AQP4 is likely to be a novel therapeutic target for NP management.
Collapse
Affiliation(s)
- Shiwu Guo
- Department of Spinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiwen Song
- Department of Spinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Junsheng He
- Department of Spinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Gang Yin
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Jianguo Zhu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Haifeng Liu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Lei Yang
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Xubiao Ji
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Xu Xu
- Department of Spinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiyuan Liu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Jinbo Liu
- Department of Spinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| |
Collapse
|
38
|
ZHANG J, ZHU YZ, JIA HL, ZHANG ZL, ZHANG YC. 劳宫穴主治病症和配伍规律的古今对比研究. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2021. [DOI: 10.1016/j.wjam.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
39
|
Hellas JA, Andrew RD. Neuronal Swelling: A Non-osmotic Consequence of Spreading Depolarization. Neurocrit Care 2021; 35:112-134. [PMID: 34498208 PMCID: PMC8536653 DOI: 10.1007/s12028-021-01326-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/04/2021] [Indexed: 01/22/2023]
Abstract
An acute reduction in plasma osmolality causes rapid uptake of water by astrocytes but not by neurons, whereas both cell types swell as a consequence of lost blood flow (ischemia). Either hypoosmolality or ischemia can displace the brain downwards, potentially causing death. However, these disorders are fundamentally different at the cellular level. Astrocytes osmotically swell or shrink because they express functional water channels (aquaporins), whereas neurons lack functional aquaporins and thus maintain their volume. Yet both neurons and astrocytes immediately swell when blood flow to the brain is compromised (cytotoxic edema) as following stroke onset, sudden cardiac arrest, or traumatic brain injury. In each situation, neuronal swelling is the direct result of spreading depolarization (SD) generated when the ATP-dependent sodium/potassium ATPase (the Na+/K+ pump) is compromised. The simple, and incorrect, textbook explanation for neuronal swelling is that increased Na+ influx passively draws Cl- into the cell, with water following by osmosis via some unknown conduit. We first review the strong evidence that mammalian neurons resist volume change during acute osmotic stress. We then contrast this with their dramatic swelling during ischemia. Counter-intuitively, recent research argues that ischemic swelling of neurons is non-osmotic, involving ion/water cotransporters as well as at least one known amino acid water pump. While incompletely understood, these mechanisms argue against the dogma that neuronal swelling involves water uptake driven by an osmotic gradient with aquaporins as the conduit. Promoting clinical recovery from neuronal cytotoxic edema evoked by spreading depolarizations requires a far better understanding of molecular water pumps and ion/water cotransporters that act to rebalance water shifts during brain ischemia.
Collapse
Affiliation(s)
- Julia A Hellas
- Center for Neuroscience Studies, Queen's University, Kingston, ON, K7L 3N6, Canada.
| | - R David Andrew
- Center for Neuroscience Studies, Queen's University, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
40
|
Jeon H, Kim M, Park W, Lim JS, Lee E, Cha H, Ahn JS, Kim JH, Hong SH, Park JE, Lee EJ, Woo CW, Lee S. Upregulation of AQP4 Improves Blood-Brain Barrier Integrity and Perihematomal Edema Following Intracerebral Hemorrhage. Neurotherapeutics 2021; 18:2692-2706. [PMID: 34545550 PMCID: PMC8804112 DOI: 10.1007/s13311-021-01126-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/08/2023] Open
Abstract
In intracerebral hemorrhage (ICH), delayed secondary neural damages largely occur from perihematomal edema (PHE) resulting from the disruption of the blood-brain barrier (BBB). PHE is often considered the principal cause of morbidity and mortality in patients with ICH. Nevertheless, the main cellular mechanism as well as the specific BBB component involved in the formation of PHE after ICH remains elusive. Herein, we evaluated the role of AQP4, a water channel expressed on the astrocytes of the BBB, in the formation of PHE in ICH. The static and dynamic functions of the BBB were evaluated by analyzing the microstructure and leakage assay. Protein changes in the PHE lesion were analyzed and the control mechanism of AQP4 expression by reactive oxygen species was also investigated. Delayed PHE formation due to BBB disruption after ICH was confirmed by the decreased coverage of multiple BBB components and increased dynamic leakages. Microstructure assay showed that among the BBB components, AQP4 showed a markedly decreased expression in the PHE lesions. The decrease in AQP4 was due to microenvironmental ROS derived from the hemorrhage and was restored by treatment with ROS scavenger. AQP4-deficient mice had significantly larger PHE lesions and unfavorable survival outcomes compared with wild-type mice. Our data identify AQP4 as a specific BBB-modulating target for alleviating PHE in ICH. Further comprehensive studies are needed to form the preclinical basis for the use of AQP4 enhancers as BBB modulators for preventing delayed cerebral edema after ICH.
Collapse
Affiliation(s)
- Hanwool Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Moinay Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Wonhyoung Park
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunyeup Lee
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyeuk Cha
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Sung Ahn
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seok Ho Hong
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Park
- University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Neuroradiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun-Jae Lee
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chul-Woong Woo
- Convergence Medicine Research Center, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seungjoo Lee
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Chen S, Shao L, Ma L. Cerebral Edema Formation After Stroke: Emphasis on Blood-Brain Barrier and the Lymphatic Drainage System of the Brain. Front Cell Neurosci 2021; 15:716825. [PMID: 34483842 PMCID: PMC8415457 DOI: 10.3389/fncel.2021.716825] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Brain edema is a severe stroke complication that is associated with prolonged hospitalization and poor outcomes. Swollen tissues in the brain compromise cerebral perfusion and may also result in transtentorial herniation. As a physical and biochemical barrier between the peripheral circulation and the central nervous system (CNS), the blood–brain barrier (BBB) plays a vital role in maintaining the stable microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the dysfunction of the BBB results in increased paracellular permeability, directly contributing to the extravasation of blood components into the brain and causing cerebral vasogenic edema. Recent studies have led to the discovery of the glymphatic system and meningeal lymphatic vessels, which provide a channel for cerebrospinal fluid (CSF) to enter the brain and drain to nearby lymph nodes and communicate with the peripheral immune system, modulating immune surveillance and brain responses. A deeper understanding of the function of the cerebral lymphatic system calls into question the known mechanisms of cerebral edema after stroke. In this review, we first discuss how BBB disruption after stroke can cause or contribute to cerebral edema from the perspective of molecular and cellular pathophysiology. Finally, we discuss how the cerebral lymphatic system participates in the formation of cerebral edema after stroke and summarize the pathophysiological process of cerebral edema formation after stroke from the two directions of the BBB and cerebral lymphatic system.
Collapse
Affiliation(s)
- Sichao Chen
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linqian Shao
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Ma
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Molecular Mechanisms of Neuroimmune Crosstalk in the Pathogenesis of Stroke. Int J Mol Sci 2021; 22:ijms22179486. [PMID: 34502395 PMCID: PMC8431165 DOI: 10.3390/ijms22179486] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022] Open
Abstract
Stroke disrupts the homeostatic balance within the brain and is associated with a significant accumulation of necrotic cellular debris, fluid, and peripheral immune cells in the central nervous system (CNS). Additionally, cells, antigens, and other factors exit the brain into the periphery via damaged blood–brain barrier cells, glymphatic transport mechanisms, and lymphatic vessels, which dramatically influence the systemic immune response and lead to complex neuroimmune communication. As a result, the immunological response after stroke is a highly dynamic event that involves communication between multiple organ systems and cell types, with significant consequences on not only the initial stroke tissue injury but long-term recovery in the CNS. In this review, we discuss the complex immunological and physiological interactions that occur after stroke with a focus on how the peripheral immune system and CNS communicate to regulate post-stroke brain homeostasis. First, we discuss the post-stroke immune cascade across different contexts as well as homeostatic regulation within the brain. Then, we focus on the lymphatic vessels surrounding the brain and their ability to coordinate both immune response and fluid homeostasis within the brain after stroke. Finally, we discuss how therapeutic manipulation of peripheral systems may provide new mechanisms to treat stroke injury.
Collapse
|
43
|
Wu J, Li Z, Yuan W, Zhang Q, Liang Y, Zhang M, Qin H, Li C. Shenfu injection improves cerebral microcirculation and reduces brain injury in a porcine model of hemorrhagic shock. Clin Hemorheol Microcirc 2021; 78:175-185. [PMID: 33579831 DOI: 10.3233/ch-211100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Shenfu injection (SFI) is a traditional Chinese herbal medicine which has been clinically used for treatment of septic shock and cardiac shock. The aim of this study was to clarify effects of SFI on cerebral microcirculation and brain injury after hemorrhagic shock (HS). METHODS Twenty-one domestic male Beijing Landrace pigs were randomly divided into three groups: SFI group (SFI, n = 8), saline group (SA, n = 8) or sham operation group (SO, n = 5). In the SFI group, animals were induced to HS by rapid bleeding to a mean arterial pressure of 40 mmHg within 10 minutes and maintained at 40±3 mmHg for 60 minutes. Volume resuscitation (shed blood and crystalloid) and SFI were given after 1 hour of HS. In the SA group, animals received the same dose of saline instead of SFI. In the SO group, the same surgical procedure was performed but without inducing HS and volume resuscitation. The cerebral microvascular flow index (MFI), nitric oxide synthase (NOS) expression, aquaporin-4 expression, interleukin-6, tumor necrosis factor-α (TNF-α) and ultrastructural of microvascular endothelia were measured. RESULTS Compared with the SA group, SFI significantly improved cerebral MFI after HS. SFI up regulated cerebral endothelial NOS expression, but down regulated interleukin-6, TNF-α, inducible NOS and aquaporin-4 expression compared with the SA group. The cerebral microvascular endothelial injury and interstitial edema in the SFI group were lighter than those in the SA group. CONCLUSIONS Combined application of SFI with volume resuscitation after HS can improve cerebral microcirculation and reduce brain injury.
Collapse
Affiliation(s)
- Junyuan Wu
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhiwei Li
- Department of Neurology, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing, China
| | - Wei Yuan
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Qiang Zhang
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yong Liang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mingqing Zhang
- Department of Emergency Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Hongjie Qin
- Department of Emergency Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chunsheng Li
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Huang B, Wang H, Zhong D, Meng J, Li M, Yang B, Ran J. Expression of Urea Transporter B in Normal and Injured Brain. Front Neuroanat 2021; 15:591726. [PMID: 34122018 PMCID: PMC8194276 DOI: 10.3389/fnana.2021.591726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
Urea transporter B (UT-B) is a membrane channel protein widely distributed in mammals, and plays a significant physiological role by regulating urea and water transportation in different tissues. More and more studies have found that UT-B is related to neurological diseases, including myelinopathy and depression. When urea accumulates in the brains of UT-B knockout mice, the synaptic plasticity of neurons is reduced, and the morphology and function of glial cells are also changed. However, the distribution and expression change of UT-B remain unclear. The purpose of this study is to determine the expression characteristics of UT-B in the brain. Through single-cell RNA sequencing, UT-B was found to express universally and substantially throughout the various cells in the central nervous system except for endothelial and smooth muscle cells. UT-B was detected in the third cerebral ventricular wall, granule cell layer of the dentate gyrus, and other parts of the hippocampal, cerebral cortex, substantia nigra, habenular, and lateral hypothalamic nucleus by immunohistochemistry. Compared with the membrane expression of UT-B in glial cells, the subcellular localization of UT-B is in the Golgi apparatus of neurons. Further, the expression of UT-B was regulated by osmotic pressure in vitro. In the experimental traumatic brain injury model (TBI), the number of UT-B positive neurons near the ipsilateral cerebral cortex increased first and then decreased over time, peaking at the 24 h. We inferred that change in UT-B expression after the TBI was an adaptation to changed urea levels. The experimental data suggest that the UT-B may be a potential target for the treatment of TBI and white matter edema.
Collapse
Affiliation(s)
- Boyue Huang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China.,Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Hongkai Wang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China.,Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Dandan Zhong
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Jia Meng
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Min Li
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Jianhua Ran
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Zhang X, O’Callaghan P, Li H, Tan Y, Zhang G, Barash U, Wang X, Lannfelt L, Vlodavsky I, Lindahl U, Li JP. Heparanase overexpression impedes perivascular clearance of amyloid-β from murine brain: relevance to Alzheimer's disease. Acta Neuropathol Commun 2021; 9:84. [PMID: 33971986 PMCID: PMC8111754 DOI: 10.1186/s40478-021-01182-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Defective amyloid-β (Aβ) clearance from the brain is a major contributing factor to the pathophysiology of Alzheimer's disease (AD). Aβ clearance is mediated by macrophages, enzymatic degradation, perivascular drainage along the vascular basement membrane (VBM) and transcytosis across the blood-brain barrier (BBB). AD pathology is typically associated with cerebral amyloid angiopathy due to perivascular accumulation of Aβ. Heparan sulfate (HS) is an important component of the VBM, thought to fulfill multiple roles in AD pathology. We previously showed that macrophage-mediated clearance of intracortically injected Aβ was impaired in the brains of transgenic mice overexpressing heparanase (Hpa-tg). This study revealed that perivascular drainage was impeded in the Hpa-tg brain, evidenced by perivascular accumulation of the injected Aβ in the thalamus of Hpa-tg mice. Furthermore, endogenous Aβ accumulated at the perivasculature of Hpa-tg thalamus, but not in control thalamus. This perivascular clearance defect was confirmed following intracortical injection of dextran that was largely retained in the perivasculature of Hpa-tg brains, compared to control brains. Hpa-tg brains presented with thicker VBMs and swollen perivascular astrocyte endfeet, as well as elevated expression of the BBB-associated water-pump protein aquaporin 4 (AQP4). Elevated levels of both heparanase and AQP4 were also detected in human AD brain. These findings indicate that elevated heparanase levels alter the organization and composition of the BBB, likely through increased fragmentation of BBB-associated HS, resulting in defective perivascular drainage. This defect contributes to perivascular accumulation of Aβ in the Hpa-tg brain, highlighting a potential role for heparanase in the pathogenesis of AD.
Collapse
|
46
|
Chen J, Wang L, Xu H, Wang Y, Liang Q. The lymphatic drainage system of the CNS plays a role in lymphatic drainage, immunity, and neuroinflammation in stroke. J Leukoc Biol 2021; 110:283-291. [PMID: 33884651 DOI: 10.1002/jlb.5mr0321-632r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/31/2022] Open
Abstract
The lymphatic drainage system of the central nervous system (CNS) plays an important role in maintaining interstitial fluid balance and regulating immune responses and immune surveillance. The impaired lymphatic drainage system of the CNS might be involved in the onset and progression of various neurodegenerative diseases, neuroinflammation, and cerebrovascular diseases. A significant immune response and brain edema are observed after stroke, resulting from disrupted homeostasis in the brain. Thus, understanding the lymphatic drainage system of the CNS in stroke may lead to the development of new approaches for therapeutic interventions in the future. Here, we review recent evidence implicating the lymphatic drainage system of the CNS in stroke.
Collapse
Affiliation(s)
- Jinman Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Linmei Wang
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| |
Collapse
|
47
|
Yao Y, Zhang Y, Liao X, Yang R, Lei Y, Luo J. Potential Therapies for Cerebral Edema After Ischemic Stroke: A Mini Review. Front Aging Neurosci 2021; 12:618819. [PMID: 33613264 PMCID: PMC7890111 DOI: 10.3389/fnagi.2020.618819] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/28/2020] [Indexed: 02/05/2023] Open
Abstract
Stroke is the leading cause of global mortality and disability. Cerebral edema and intracranial hypertension are common complications of cerebral infarction and the major causes of mortality. The formation of cerebral edema includes three stages (cytotoxic edema, ionic edema, and vasogenic edema), which involve multiple proteins and ion channels. A range of therapeutic agents that successfully target cerebral edema have been developed in animal studies, some of which have been assessed in clinical trials. Herein, we review the mechanisms of cerebral edema and the research progress of anti-edema therapies for use after ischemic stroke.
Collapse
Affiliation(s)
- Yi Yao
- International Medical Center, Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, Chengdu, China
- Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- International Medical Center, Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Yang
- International Medical Center, Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Lei
- International Medical Center, Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jianzhao Luo
- International Medical Center, Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Bloodletting Puncture at Hand Twelve Jing-Well Points Relieves Brain Edema after Severe Traumatic Brain Injury in Rats via Inhibiting MAPK Signaling Pathway. Chin J Integr Med 2021; 27:291-299. [PMID: 33515398 DOI: 10.1007/s11655-021-3326-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2019] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To investigate whether blood-brain barrier (BBB) served a key role in the edema-relief effect of bloodletting puncture at hand twelve Jing-well points (HTWP) in traumatic brain injury (TBI) and the potential molecular signaling pathways. METHODS Adult male Sprague-Dawley rats were assigned to the sham-operated (sham), TBI, and bloodletting puncture (bloodletting) groups (n=24 per group) using a randomized number table. The TBI model rats were induced by cortical contusion and then bloodletting puncture were performed at HTWP twice a day for 2 days. The neurological function and cerebral edema were evaluated by modified neurological severity score (mNSS), cerebral water content, magnetic resonance imaging and hematoxylin and eosin staining. Cerebral blood flow was measured by laser speckles. The protein levels of aquaporin 4 (AQP4), matrix metalloproteinases 9 (MMP9) and mitogen-activated protein kinase pathway (MAPK) signaling were detected by immunofluorescence staining and Western blot. RESULTS Compared with TBI group, bloodletting puncture improved neurological function at 24 and 48 h, alleviated cerebral edema at 48 h, and reduced the permeability of BBB induced by TBI (all P<0.05). The AQP4 and MMP9 which would disrupt the integrity of BBB were downregulated by bloodletting puncture (P<0.05 or P<0.01). In addition, the extracellular signal-regulated kinase (ERK) and p38 signaling pathways were inhibited by bloodletting puncture (P<0.05). CONCLUSIONS Bloodletting puncture at HTWP might play a significant role in protecting BBB through regulating the expressions of MMP9 and AQP4 as well as corresponding regulatory upstream ERK and p38 signaling pathways. Therefore, bloodletting puncture at HTWP may be a promising therapeutic strategy for TBI-induced cerebral edema.
Collapse
|
49
|
KOZLER P, HERYNEK V, MAREŠOVÁ D, PEREZ P, ŠEFC L, POKORNÝ J. Effect of Methylprednisolone on Experimental Brain Edema in Magnetic Resonance Imaging. Physiol Res 2020; 69:919-926. [DOI: 10.33549/physiolres.934460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Magnetic resonance imaging has been used for evaluating of a brain edema in experimental animals to assess cytotoxic and vasogenic edema by the apparent diffusion coefficient (ADC) and T2 imaging. This paper brings information about the effectiveness of methylprednisolone (MP) on experimental brain edema. A total of 24 rats were divided into three groups of 8 animals each. Rats with cytotoxic/intracellular brain edema induced by water intoxication were assigned to the group WI. These rats also served as the additional control group CG when measured before the induction of edema. A third group (WIMP) was intraperitoneally administered with methylprednisolone 100 mg/kg during water intoxication treatment. The group WI+MP was injected with methylprednisolone 50 mg/kg into the carotid artery within two hours after the water intoxication treatment. We evaluated the results in four groups. Two control groups (CG, WI) and two experimental groups (WIMP, WI+MP). Rats were subjected to MR scanning 24 h after edema induction. We observed significantly increased ADC values in group WI in both evaluated areas – cortex and hippocampus, which proved the occurrence of experimental vasogenic edema, while ADC values in groups WIMP and WI+MP were not increased, indicating that the experimental edema was not developed and thus confirming the protective effect of MP.
Collapse
Affiliation(s)
- P KOZLER
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - V HERYNEK
- Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - D MAREŠOVÁ
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - P PEREZ
- Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - L ŠEFC
- Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - J POKORNÝ
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
50
|
Khaw YM, Aggarwal N, Barclay WE, Kang E, Inoue M, Shinohara ML. Th1-Dependent Cryptococcus-Associated Immune Reconstitution Inflammatory Syndrome Model With Brain Damage. Front Immunol 2020; 11:529219. [PMID: 33133067 PMCID: PMC7550401 DOI: 10.3389/fimmu.2020.529219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/04/2020] [Indexed: 01/02/2023] Open
Abstract
Cryptococcus-associated immune reconstitution inflammatory syndrome (C-IRIS) is identified upon immune reconstitution in immunocompromised patients, who have previously contracted an infection of Cryptococcus neoformans (Cn). C-IRIS can be lethal but how the immune system triggers life-threatening outcomes in patients is still poorly understood. Here, we establish a mouse model for C-IRIS with Cn serotype A strain H99, which is highly virulent and the most intensively studied. C-IRIS in mice is induced by the adoptive transfer of CD4+ T cells in immunocompromised Rag1-deficient mice infected with a low inoculum of Cn. The mice with C-IRIS exhibit symptoms which mimic clinical presentations of C-IRIS. This C-IRIS model is Th1-dependent and shows host mortality. This model is characterized with minimal lung injury, but infiltration of Th1 cells in the brain. C-IRIS mice also exhibited brain swelling with resemblance to edema and upregulation of aquaporin-4, a critical protein that regulates water flux in the brain in a Th1-dependent fashion. Our C-IRIS model may be used to advance our understanding of the paradoxical inflammatory phenomenon of C-IRIS in the context of neuroinflammation.
Collapse
Affiliation(s)
- Yee Ming Khaw
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Nupur Aggarwal
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - William E. Barclay
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Eunjoo Kang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Makoto Inoue
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mari L. Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|