1
|
Zhao Y, Huang Y, Cao Y, Yang J. Astrocyte-Mediated Neuroinflammation in Neurological Conditions. Biomolecules 2024; 14:1204. [PMID: 39456137 PMCID: PMC11505625 DOI: 10.3390/biom14101204] [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: 08/09/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Astrocytes are one of the key glial types of the central nervous system (CNS), accounting for over 20% of total glial cells in the brain. Extensive evidence has established their indispensable functions in the maintenance of CNS homeostasis, as well as their broad involvement in neurological conditions. In particular, astrocytes can participate in various neuroinflammatory processes, e.g., releasing a repertoire of cytokines and chemokines or specific neurotrophic factors, which result in both beneficial and detrimental effects. It has become increasingly clear that such astrocyte-mediated neuroinflammation, together with its complex crosstalk with other glial cells or immune cells, designates neuronal survival and the functional integrity of neurocircuits, thus critically contributing to disease onset and progression. In this review, we focus on the current knowledge of the neuroinflammatory responses of astrocytes, summarizing their common features in neurological conditions. Moreover, we highlight several vital questions for future research that promise novel insights into diagnostic or therapeutic strategies against those debilitating CNS diseases.
Collapse
Affiliation(s)
- Yanxiang Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- The Affiliated High School, Peking University, Beijing 100080, China
| | - Yingying Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Third Hospital Cancer Center, Beijing 100191, China
| |
Collapse
|
2
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
3
|
Lia A, Di Spiezio A, Vitalini L, Tore M, Puja G, Losi G. Ion Channels and Ionotropic Receptors in Astrocytes: Physiological Functions and Alterations in Alzheimer's Disease and Glioblastoma. Life (Basel) 2023; 13:2038. [PMID: 37895420 PMCID: PMC10608464 DOI: 10.3390/life13102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The human brain is composed of nearly one hundred billion neurons and an equal number of glial cells, including macroglia, i.e., astrocytes and oligodendrocytes, and microglia, the resident immune cells of the brain. In the last few decades, compelling evidence has revealed that glial cells are far more active and complex than previously thought. In particular, astrocytes, the most abundant glial cell population, not only take part in brain development, metabolism, and defense against pathogens and insults, but they also affect sensory, motor, and cognitive functions by constantly modulating synaptic activity. Not surprisingly, astrocytes are actively involved in neurodegenerative diseases (NDs) and other neurological disorders like brain tumors, in which they rapidly become reactive and mediate neuroinflammation. Reactive astrocytes acquire or lose specific functions that differently modulate disease progression and symptoms, including cognitive impairments. Astrocytes express several types of ion channels, including K+, Na+, and Ca2+ channels, transient receptor potential channels (TRP), aquaporins, mechanoreceptors, and anion channels, whose properties and functions are only partially understood, particularly in small processes that contact synapses. In addition, astrocytes express ionotropic receptors for several neurotransmitters. Here, we provide an extensive and up-to-date review of the roles of ion channels and ionotropic receptors in astrocyte physiology and pathology. As examples of two different brain pathologies, we focus on Alzheimer's disease (AD), one of the most diffuse neurodegenerative disorders, and glioblastoma (GBM), the most common brain tumor. Understanding how ion channels and ionotropic receptors in astrocytes participate in NDs and tumors is necessary for developing new therapeutic tools for these increasingly common neurological conditions.
Collapse
Affiliation(s)
- Annamaria Lia
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
| | - Alessandro Di Spiezio
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
- Neuroscience Institute (CNR-IN), Padova Section, 35131 Padova, Italy
| | - Lorenzo Vitalini
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Manuela Tore
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Puja
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Gabriele Losi
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
4
|
Sanicola HW, Stewart CE, Luther P, Yabut K, Guthikonda B, Jordan JD, Alexander JS. Pathophysiology, Management, and Therapeutics in Subarachnoid Hemorrhage and Delayed Cerebral Ischemia: An Overview. PATHOPHYSIOLOGY 2023; 30:420-442. [PMID: 37755398 PMCID: PMC10536590 DOI: 10.3390/pathophysiology30030032] [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: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke resulting from the rupture of an arterial vessel within the brain. Unlike other stroke types, SAH affects both young adults (mid-40s) and the geriatric population. Patients with SAH often experience significant neurological deficits, leading to a substantial societal burden in terms of lost potential years of life. This review provides a comprehensive overview of SAH, examining its development across different stages (early, intermediate, and late) and highlighting the pathophysiological and pathohistological processes specific to each phase. The clinical management of SAH is also explored, focusing on tailored treatments and interventions to address the unique pathological changes that occur during each stage. Additionally, the paper reviews current treatment modalities and pharmacological interventions based on the evolving guidelines provided by the American Heart Association (AHA). Recent advances in our understanding of SAH will facilitate clinicians' improved management of SAH to reduce the incidence of delayed cerebral ischemia in patients.
Collapse
Affiliation(s)
- Henry W. Sanicola
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Caleb E. Stewart
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Patrick Luther
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Kevin Yabut
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Bharat Guthikonda
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
5
|
Tesler F, Linne ML, Destexhe A. Modeling the relationship between neuronal activity and the BOLD signal: contributions from astrocyte calcium dynamics. Sci Rep 2023; 13:6451. [PMID: 37081004 PMCID: PMC10119111 DOI: 10.1038/s41598-023-32618-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/30/2023] [Indexed: 04/22/2023] Open
Abstract
Functional magnetic resonance imaging relies on the coupling between neuronal and vascular activity, but the mechanisms behind this coupling are still under discussion. Recent experimental evidence suggests that calcium signaling may play a significant role in neurovascular coupling. However, it is still controversial where this calcium signal is located (in neurons or elsewhere), how it operates and how relevant is its role. In this paper we introduce a biologically plausible model of the neurovascular coupling and we show that calcium signaling in astrocytes can explain main aspects of the dynamics of the coupling. We find that calcium signaling can explain so-far unrelated features such as the linear and non-linear regimes, the negative vascular response (undershoot) and the emergence of a (calcium-driven) Hemodynamic Response Function. These features are reproduced here for the first time by a single model of the detailed neuronal-astrocyte-vascular pathway. Furthermore, we analyze how information is coded and transmitted from the neuronal to the vascular system and we predict that frequency modulation of astrocytic calcium dynamics plays a key role in this process. Finally, our work provides a framework to link neuronal activity to the BOLD signal, and vice-versa, where neuronal activity can be inferred from the BOLD signal. This opens new ways to link known alterations of astrocytic calcium signaling in neurodegenerative diseases (e.g. Alzheimer's and Parkinson's diseases) with detectable changes in the neurovascular coupling.
Collapse
Affiliation(s)
- Federico Tesler
- CNRS, Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, 91400, Saclay, France.
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, 33720, Tampere, Finland
| | - Alain Destexhe
- CNRS, Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, 91400, Saclay, France
| |
Collapse
|
6
|
Ladthavorlaphatt K, Surti FBS, Beishon LC, Panerai RB, Robinson TG. Challenging neurovascular coupling through complex and variable duration cognitive paradigms: A subcomponent analysis. Med Eng Phys 2022; 110:103921. [PMID: 36564144 DOI: 10.1016/j.medengphy.2022.103921] [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: 05/31/2022] [Revised: 10/04/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
A similar pattern of cerebral blood velocity (CBv) response has been observed for neurovascular coupling (NVC) assessment with cognitive tasks of varying complexity and duration. This lack of specificity could result from parallel changes in arterial blood pressure (BP) and PaCO2, which could confound the estimates of NVC integrity. Healthy participants (n = 16) underwent recordings at rest (5 min sitting) and during randomized paradigms of different complexity (naming words (NW) beginning with P-, R-, V- words and serial subtractions (SS) of 100-2, 100-7, 1000-17, with durations of 5, 30 and 60 s). Bilateral CBv (middle cerebral arteries, transcranial Doppler), end-tidal CO2 (EtCO2, capnography), blood pressure (BP, Finapres) and heart rate (HR, ECG) were recorded continuously. The bilateral CBv response to all paradigms was classified under objective criteria to select only responders, then the repeated data were averaged between visits. Bilateral CBv change to tasks was decomposed into the relative contributions (subcomponents) of arterial BP (VBP; neurogenic), critical closing pressure (VCrCP; metabolic) and resistance area product (VRAP; myogenic). A temporal effect was demonstrated in bilateral VBP and VRAP during all tasks (p<0.002), increased VBP early (between 0 and 10 s) and followed by decreases of VRAP late (25-35 s) in the response. VCrCP varied by complexity and duration (p<0.046). The main contributions to CBv responses to cognitive tasks of different complexity and duration were VBP and VRAP, whilst a smaller contribution from VCrCP would suggest sensitivity to metabolic demands. Further studies are needed to assess the influence of different paradigms, ageing and cerebrovascular conditions.
Collapse
Affiliation(s)
- Kannaphob Ladthavorlaphatt
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; Medical Diagnostics Unit, Thammasat University Hospital, Thammasat University, Pathumthani, Thailand.
| | - Farhaana B S Surti
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom
| | - Lucy C Beishon
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Ronney B Panerai
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Thompson G Robinson
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| |
Collapse
|
7
|
Chen Z, Yuan Z, Yang S, Zhu Y, Xue M, Zhang J, Leng L. Brain Energy Metabolism: Astrocytes in Neurodegenerative Diseases. CNS Neurosci Ther 2022; 29:24-36. [PMID: 36193573 PMCID: PMC9804080 DOI: 10.1111/cns.13982] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are the most abundant cells in the brain. They have many important functions in the central nervous system (CNS), including the maintenance of glutamate and ion homeostasis, the elimination of oxidative stress, energy storage in glycogen, tissue repair, regulating synaptic activity by releasing neurotransmitters, and participating in synaptic formation. Astrocytes have special highly ramified structure. Their branches contact with synapses of neurons inwardly, with fine structure and wrapping synapses; their feet contact with blood vessels of brain parenchyma outward, almost wrapping the whole brain. The adjacent astrocytes rarely overlap and communicate with each other through gap junction channels. The ideal location of astrocytes enables them to sense the weak changes of their surroundings and provide the structural basis for the energy supply of neurons. Neurons and astrocytes are closely coupled units of energy metabolism in the brain. Neurons consume a lot of ATPs in the process of neurotransmission. Astrocytes provide metabolic substrates for neurons, maintain high activity of neuron, and facilitate information transmission of neurons. This article reviews the characteristics of glucose metabolism, lipid metabolism, and amino acid metabolism of astrocytes. The metabolic interactions between astrocytes and neurons, astrocytes and microglia were also detailed discussed. Finally, we classified analyzed the role of metabolic disorder of astrocytes in the occurrence and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhenlei Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Ziqi Yuan
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Shangchen Yang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Yufei Zhu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Maoqiang Xue
- Department of Basic Medical Science, School of MedicineXiamen UniversityXiamenChina
| | - Jie Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Lige Leng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| |
Collapse
|
8
|
Bohr T, Hjorth PG, Holst SC, Hrabětová S, Kiviniemi V, Lilius T, Lundgaard I, Mardal KA, Martens EA, Mori Y, Nägerl UV, Nicholson C, Tannenbaum A, Thomas JH, Tithof J, Benveniste H, Iliff JJ, Kelley DH, Nedergaard M. The glymphatic system: Current understanding and modeling. iScience 2022; 25:104987. [PMID: 36093063 PMCID: PMC9460186 DOI: 10.1016/j.isci.2022.104987] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We review theoretical and numerical models of the glymphatic system, which circulates cerebrospinal fluid and interstitial fluid around the brain, facilitating solute transport. Models enable hypothesis development and predictions of transport, with clinical applications including drug delivery, stroke, cardiac arrest, and neurodegenerative disorders like Alzheimer's disease. We sort existing models into broad categories by anatomical function: Perivascular flow, transport in brain parenchyma, interfaces to perivascular spaces, efflux routes, and links to neuronal activity. Needs and opportunities for future work are highlighted wherever possible; new models, expanded models, and novel experiments to inform models could all have tremendous value for advancing the field.
Collapse
Affiliation(s)
- Tomas Bohr
- Department of Physics, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Poul G. Hjorth
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads, 2800 Kgs. Lyngby, Denmark
| | - Sebastian C. Holst
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Sabina Hrabětová
- Department of Cell Biology and The Robert Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Vesa Kiviniemi
- Oulu Functional NeuroImaging, Department of Diagnostic Radiology, MRC, Oulu University Hospital, Oulu, Finland
- Medical Imaging, Physics and Technology, the Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Tuomas Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Kent-Andre Mardal
- Department of Mathematics, University of Oslo, Oslo, Norway
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Oslo, Norway
| | | | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - U. Valentin Nägerl
- Instítut Interdisciplinaire de Neurosciences, Université de Bordeaux / CNRS UMR 5297, Centre Broca Nouvelle-Aquitaine, 146 rue Léo Saignat, CS 61292 Case 130, 33076 Bordeaux Cedex France
| | - Charles Nicholson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Allen Tannenbaum
- Departments of Computer Science/ Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - John H. Thomas
- Department of Mechanical Engineering, University of Rochester, Rochester, 14627 NY, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey J. Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, 14627 NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, 14642 NY, USA
| |
Collapse
|
9
|
Turner MP, Zhao Y, Abdelkarim D, Liu P, Spence JS, Hutchison JL, Sivakolundu DK, Thomas BP, Hubbard NA, Xu C, Taneja K, Lu H, Rypma B. Altered linear coupling between stimulus-evoked blood flow and oxygen metabolism in the aging human brain. Cereb Cortex 2022; 33:135-151. [PMID: 35388407 PMCID: PMC9758587 DOI: 10.1093/cercor/bhac057] [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: 07/22/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
Neural-vascular coupling (NVC) is the process by which oxygen and nutrients are delivered to metabolically active neurons by blood vessels. Murine models of NVC disruption have revealed its critical role in healthy neural function. We hypothesized that, in humans, aging exerts detrimental effects upon the integrity of the neural-glial-vascular system that underlies NVC. To test this hypothesis, calibrated functional magnetic resonance imaging (cfMRI) was used to characterize age-related changes in cerebral blood flow (CBF) and oxygen metabolism during visual cortex stimulation. Thirty-three younger and 27 older participants underwent cfMRI scanning during both an attention-controlled visual stimulation task and a hypercapnia paradigm used to calibrate the blood-oxygen-level-dependent signal. Measurement of stimulus-evoked blood flow and oxygen metabolism permitted calculation of the NVC ratio to assess the integrity of neural-vascular communication. Consistent with our hypothesis, we observed monotonic NVC ratio increases with increasing visual stimulation frequency in younger adults but not in older adults. Age-related changes in stimulus-evoked cerebrovascular and neurometabolic signal could not fully explain this disruption; increases in stimulus-evoked neurometabolic activity elicited corresponding increases in stimulus-evoked CBF in younger but not in older adults. These results implicate age-related, demand-dependent failures of the neural-glial-vascular structures that comprise the NVC system.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Yuguang Zhao
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dema Abdelkarim
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Peiying Liu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Binu P Thomas
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Nicholas A Hubbard
- Department of Psychology, Center for Brain, Biology, and Behavior, University of Nebraska, Lincoln, NE 68588, USA
| | - Cuimei Xu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Kamil Taneja
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Hanzhang Lu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Bart Rypma
- Corresponding author: School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA.
| |
Collapse
|
10
|
Paul G, Elabi OF. Microvascular Changes in Parkinson’s Disease- Focus on the Neurovascular Unit. Front Aging Neurosci 2022; 14:853372. [PMID: 35360216 PMCID: PMC8960855 DOI: 10.3389/fnagi.2022.853372] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/11/2022] [Indexed: 12/27/2022] Open
Abstract
Vascular alterations emerge as a common denominator for several neurodegenerative diseases. In Parkinson’s disease (PD), a number of observations have been made suggesting that the occurrence of vascular pathology is an important pathophysiological aspect of the disease. Specifically, pathological activation of pericytes, blood-brain barrier (BBB) disruption, pathological angiogenesis and vascular regression have been reported. This review summarizes the current evidence for the different vascular alterations in patients with PD and in animal models of PD. We suggest a possible sequence of vascular pathology in PD ranging from early pericyte activation and BBB leakage to an attempt for compensatory angiogenesis and finally vascular rarefication. We highlight different pathogenetic mechanisms that play a role in these vascular alterations including perivascular inflammation and concomitant metabolic disease. Awareness of the contribution of vascular events to the pathogenesis of PD may allow the identification of targets to modulate those mechanisms. In particular the BBB has for decades only been viewed as an obstacle for drug delivery, however, preservation of its integrity and/or modulation of the signaling at this interface between the blood and the brain may prove to be a new avenue to take in order to develop disease-modifying strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Gesine Paul
- Translational Neurology Group, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- *Correspondence: Gesine Paul,
| | - Osama F. Elabi
- Translational Neurology Group, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Pouso MR, Cairrao E. Effect of retinoic acid on the neurovascular unit: A review. Brain Res Bull 2022; 184:34-45. [DOI: 10.1016/j.brainresbull.2022.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
|
12
|
Balachandar L, Borrego D, Diaz JR. Serotype-based evaluation of an optogenetic construct in rat cortical astrocytes. Biochem Biophys Res Commun 2022; 593:35-39. [DOI: 10.1016/j.bbrc.2022.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/08/2022] [Indexed: 11/16/2022]
|
13
|
Liu L, Gao H, Zaikin A, Chen S. Unraveling Aβ-Mediated Multi-Pathway Calcium Dynamics in Astrocytes: Implications for Alzheimer's Disease Treatment From Simulations. Front Physiol 2021; 12:767892. [PMID: 34777023 PMCID: PMC8581622 DOI: 10.3389/fphys.2021.767892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/08/2021] [Indexed: 02/02/2023] Open
Abstract
The accumulation of amyloid β peptide (Aβ) in the brain is hypothesized to be the major factor driving Alzheimer's disease (AD) pathogenesis. Mounting evidence suggests that astrocytes are the primary target of Aβ neurotoxicity. Aβ is known to interfere with multiple calcium fluxes, thus disrupting the calcium homeostasis regulation of astrocytes, which are likely to produce calcium oscillations. Ca2+ dyshomeostasis has been observed to precede the appearance of clinical symptoms of AD; however, it is experimentally very difficult to investigate the interactions of many mechanisms. Given that Ca2+ disruption is ubiquitously involved in AD progression, it is likely that focusing on Ca2+ dysregulation may serve as a potential therapeutic approach to preventing or treating AD, while current hypotheses concerning AD have so far failed to yield curable therapies. For this purpose, we derive and investigate a concise mathematical model for Aβ-mediated multi-pathway astrocytic intracellular Ca2+ dynamics. This model accounts for how Aβ affects various fluxes contributions through voltage-gated calcium channels, Aβ-formed channels and ryanodine receptors. Bifurcation analysis of Aβ level, which reflected the corresponding progression of the disease, revealed that Aβ significantly induced the increasing [Ca2+] i and frequency of calcium oscillations. The influence of inositol 1,4,5-trisphosphate production (IP3) is also investigated in the presence of Aβ as well as the impact of changes in resting membrane potential. In turn, the Ca2+ flux can be considerably changed by exerting specific interventions, such as ion channel blockers or receptor antagonists. By doing so, a "combination therapy" targeting multiple pathways simultaneously has finally been demonstrated to be more effective. This study helps to better understand the effect of Aβ, and our findings provide new insight into the treatment of AD.
Collapse
Affiliation(s)
- Langzhou Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Gao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Alexey Zaikin
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Institute for Women's Health and Department of Mathematics, University College London, London, United Kingdom.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Shangbin Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Nelissen E, Argyrousi EK, Van Goethem NP, Zhao F, Hines CDG, Swaminath G, Gerisch M, Hueser J, Sandner P, Prickaerts J. Soluble Guanylate Cyclase Stimulator Vericiguat Enhances Long-Term Memory in Rats without Altering Cerebral Blood Volume. Biomedicines 2021; 9:1047. [PMID: 34440254 PMCID: PMC8393324 DOI: 10.3390/biomedicines9081047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 01/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) is characterized by impairments in cerebral blood flow (CBF), endothelial function and blood-brain barrier (BBB) integrity. These processes are all physiologically regulated by the nitric oxide (NO)-soluble guanylate cyclase (sGC)-cGMP signaling pathway. Additionally, cGMP signaling plays an important role in long-term potentiation (LTP) underlying memory formation. Therefore, targeting the NO-sGC-cGMP pathway may be a therapeutic strategy for treating VCI. Hence, in this study we investigated whether sGC stimulator vericiguat has potential as a cognitive enhancer. The effects of vericiguat on long-term memory were measured in rats using an object location task. Due to the low brain-penetrance of vericiguat found in this study, it was investigated whether in the absence of BBB limitations, vericiguat enhanced hippocampal plasticity using an ex vivo memory acquisition-like chemical LTP model. Finally, peripheral effects were measured by means of blood pressure and cerebral blood volume. Vericiguat successfully enhanced long-term memory and increased hippocampal plasticity via enhanced translocation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors to the cell membrane, while blood pressure and cerebral blood volume were unaltered. Although the memory enhancing effects in this study are likely due to peripheral effects on the cerebral microvasculature, sGC stimulation may provide a new therapeutic strategy for treating VCI, especially when BBB integrity is reduced.
Collapse
Affiliation(s)
- Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (E.N.); (E.K.A.); (N.P.V.G.)
| | - Elentina K. Argyrousi
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (E.N.); (E.K.A.); (N.P.V.G.)
| | - Nick P. Van Goethem
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (E.N.); (E.K.A.); (N.P.V.G.)
| | - Fuqiang Zhao
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (F.Z.); (C.D.G.H.)
| | | | | | - Michael Gerisch
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; (M.G.); (J.H.); (P.S.)
| | - Joerg Hueser
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; (M.G.); (J.H.); (P.S.)
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; (M.G.); (J.H.); (P.S.)
- Hannover Medical School, Institute for Pharmacology, 30625 Hannover, Germany
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (E.N.); (E.K.A.); (N.P.V.G.)
| |
Collapse
|
15
|
Yabluchanskiy A, Nyul-Toth A, Csiszar A, Gulej R, Saunders D, Towner R, Turner M, Zhao Y, Abdelkari D, Rypma B, Tarantini S. Age-related alterations in the cerebrovasculature affect neurovascular coupling and BOLD fMRI responses: Insights from animal models of aging. Psychophysiology 2021; 58:e13718. [PMID: 33141436 PMCID: PMC9166153 DOI: 10.1111/psyp.13718] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/10/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
The present and future research efforts in cognitive neuroscience and psychophysiology rely on the measurement, understanding, and interpretation of blood oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) to effectively investigate brain function. Aging and age-associated pathophysiological processes change the structural and functional integrity of the cerebrovasculature which can significantly alter how the BOLD signal is recorded and interpreted. In order to gain an improved understanding of the benefits, drawbacks, and methodological implications for BOLD fMRI in the context of cognitive neuroscience, it is crucial to understand the cellular and molecular mechanism of age-related vascular pathologies. This review discusses the multifaceted effects of aging and the contributions of age-related pathologies on structural and functional integrity of the cerebral microcirculation as they has been investigated in animal models of aging, including age-related alterations in neurovascular coupling responses, cellular and molecular mechanisms involved in microvascular damage, vascular rarefaction, blood-brain barrier disruption, senescence, humoral deficiencies as they relate to, and potentially introduce confounding factors in the interpretation of BOLD fMRI.
Collapse
Affiliation(s)
- Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Rheal Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Monroe Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Yuguang Zhao
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Dema Abdelkari
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary,Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
16
|
Mulica P, Grünewald A, Pereira SL. Astrocyte-Neuron Metabolic Crosstalk in Neurodegeneration: A Mitochondrial Perspective. Front Endocrinol (Lausanne) 2021; 12:668517. [PMID: 34025580 PMCID: PMC8138625 DOI: 10.3389/fendo.2021.668517] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Converging evidence made clear that declining brain energetics contribute to aging and are implicated in the initiation and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Indeed, both pathologies involve instances of hypometabolism of glucose and oxygen in the brain causing mitochondrial dysfunction, energetic failure and oxidative stress. Importantly, recent evidence suggests that astrocytes, which play a key role in supporting neuronal function and metabolism, might contribute to the development of neurodegenerative diseases. Therefore, exploring how the neuro-supportive role of astrocytes may be impaired in the context of these disorders has great therapeutic potential. In the following, we will discuss some of the so far identified features underlining the astrocyte-neuron metabolic crosstalk. Thereby, special focus will be given to the role of mitochondria. Furthermore, we will report on recent advancements concerning iPSC-derived models used to unravel the metabolic contribution of astrocytes to neuronal demise. Finally, we discuss how mitochondrial dysfunction in astrocytes could contribute to inflammatory signaling in neurodegenerative diseases.
Collapse
Affiliation(s)
- Patrycja Mulica
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
17
|
Olung NF, Aluko OM, Jeje SO, Adeagbo AS, Ijomone OM. Vascular Dysfunction in the Brain; Implications for Heavy Metal Exposures. Curr Hypertens Rev 2021; 17:5-13. [PMID: 33632106 DOI: 10.2174/1573402117666210225085528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/24/2020] [Accepted: 12/01/2020] [Indexed: 11/22/2022]
Abstract
Normal or diseased conditions that alter the brain's requirement for oxygen and nutrients via alterations to neurovascular coupling have an impact on the level of the neurovascular unit; comprising neuronal, glial and vascular components. The communications between the components of the neurovascular unit are precise and accurate for its functions; hence a minute disturbance can result in neurovascular dysfunction. Heavy metals such as cadmium, mercury, and lead have been identified to increase the vulnerability of the neurovascular unit to damage. This review examines the role of heavy metals in neurovascular dysfunctions and the possible mechanisms by which these metals act. Risk factors ranging from lifestyle, environment, genetics, infections, and physiologic ageing involved in neurological dysfunctions were highlighted, while stroke was discussed as the prevalent consequence of neurovascular dysfunctions. Furthermore, the role of these heavy metals in the pathogenesis of stroke consequently pinpoints the importance of understanding the mechanisms of neurovascular damage in a bid to curb the occurrence of neurovascular dysfunctions.
Collapse
Affiliation(s)
- Nzube F Olung
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Oritoke M Aluko
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Sikirullai O Jeje
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Ayotunde S Adeagbo
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Omamuyovwi M Ijomone
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
18
|
Weis SN, Souza JMF, Hoppe JB, Firmino M, Auer M, Ataii NN, da Silva LA, Gaelzer MM, Klein CP, Mól AR, de Lima CMR, Souza DO, Salbego CG, Ricart CAO, Fontes W, de Sousa MV. In-depth quantitative proteomic characterization of organotypic hippocampal slice culture reveals sex-specific differences in biochemical pathways. Sci Rep 2021; 11:2560. [PMID: 33510253 PMCID: PMC7844295 DOI: 10.1038/s41598-021-82016-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Sex differences in the brain of mammals range from neuroarchitecture through cognition to cellular metabolism. The hippocampus, a structure mostly associated with learning and memory, presents high vulnerability to neurodegeneration and aging. Therefore, we explored basal sex-related differences in the proteome of organotypic hippocampal slice culture, a major in vitro model for studying the cellular and molecular mechanisms related to neurodegenerative disorders. Results suggest a greater prevalence of astrocytic metabolism in females and significant neuronal metabolism in males. The preference for glucose use in glycolysis, pentose phosphate pathway and glycogen metabolism in females and high abundance of mitochondrial respiration subunits in males support this idea. An overall upregulation of lipid metabolism was observed in females. Upregulation of proteins responsible for neuronal glutamate and GABA synthesis, along with synaptic associated proteins, were observed in males. In general, the significant spectrum of pathways known to predominate in neurons or astrocytes, together with the well-known neuronal and glial markers observed, revealed sex-specific metabolic differences in the hippocampus. TEM qualitative analysis might indicate a greater presence of mitochondria at CA1 synapses in females. These findings are crucial to a better understanding of how sex chromosomes can influence the physiology of cultured hippocampal slices and allow us to gain insights into distinct responses of males and females on neurological diseases that present a sex-biased incidence.
Collapse
Affiliation(s)
- Simone Nardin Weis
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil.
| | - Jaques Miranda F Souza
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Juliana Bender Hoppe
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Marina Firmino
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Manfred Auer
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS Donner, Berkeley, CA, 94720, USA
| | - Nassim N Ataii
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS Donner, Berkeley, CA, 94720, USA
| | - Leonardo Assis da Silva
- Laboratory of Electron Microscopy, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | | | - Caroline Peres Klein
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Alan R Mól
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Consuelo M R de Lima
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Diogo Onofre Souza
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Christianne G Salbego
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Carlos André O Ricart
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Marcelo Valle de Sousa
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, 70910-900, Brazil
| |
Collapse
|
19
|
Luo YP, Zhang L, Wu XY, Hou WS, Chen L, Tian XL, Wen HZ. Cerebral blood microcirculation measurement in APP/PS1 double transgenic mice at the preclinical stage of Alzheimer's disease: preliminary data on the early intervention of anodal transcranial direct current stimulation .. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:3557-3560. [PMID: 33018771 DOI: 10.1109/embc44109.2020.9175875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anodal transcranial direct current stimulation (AtDCS) can improve memory and cognitive dysfunction in patients with Alzheimer's disease (AD), which has been proven in basic and clinical studies. Intervention of AD in preclinical stage is important to prevent progression of AD in the aging society. At the same time, there is increasing evidence that a close link exists between cerebrovascular dysfunction and AD disease. Here we investigated the changes of local cerebral blood microcirculation in preclinical AD mouse model after AtDCS based on the previous studies. Twenty-four 6-month-old male APP/PS1 double transgenic mice were randomly divided into three groups: a model group (AD), a model sham stimulation (ADST) group and a model stimulation group (ATD). Eight 6-month-old male C57 wild-type mice served as a control group (CTL). Mice in the ATD group received 10 AtDCS sessions. Two months after the end of AtDCS in the ATD group, the microcirculation parameters of the frontal cortex of the mice in each group, including cerebral blood flow (CBF), blood flow velocity (Velo), oxygen saturation (SO2) and relative hemoglobin content (rHb), were obtained by the non-invasive laser-Doppler spectrophotometry system "Oxygen-to-See (O2C)". The results showed that AtDCS increased CBF, Velo and SO2, and reduce rHb in APP/PS1 double transgenic mice at the preclinical stage of AD.Clinical Relevance-This shows the positive effect of AtDCS on preclinical AD in cerebrovascular function, and provides effective basic research facts for AtDCS to intervene and delay the clinical application of AD disease.
Collapse
|
20
|
Amtul Z, Randhawa J, Najdat AN, Hill DJ, Arany EJ. Role of Delayed Neuroglial Activation in Impaired Cerebral Blood Flow Restoration Following Comorbid Injury. Cell Mol Neurobiol 2020; 40:369-380. [PMID: 31522299 DOI: 10.1007/s10571-019-00735-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/04/2019] [Indexed: 11/27/2022]
Abstract
Besides other causes, ischemia and Alzheimer's disease pathology is also linked to decreased cerebral blood flow (CBF). There is little or no consensus about the role of neuroglial cells in maintaining CBF in various neuropathologies. This consensus becomes scarcer when it comes to clinical and experimental cases of comorbid Abeta-amyloid (Aβ) toxicity and ischemia. Here, a comorbid rat model of Aβ toxicity and endothelin-1 induced ischemia (ET1) not only demonstrated the appearance of axotomized phagocytosed pyknotic neurons (NeuN) immediately after the injury, but also showed a diversity of continuously changing neuroglia (MHC Class II/OX6, Iba1) and macrophage (Iba1/CD68) phenotypes with round, stout somas, and retracted processes. This is indicative of a response to a concomitant increase in large fluid-filled spaces due to the vascular leakage. Ironically 4 weeks after the injury despite a conclusive reduction in neurons, CBF restoration in ET1 rats was associated with a massive increase in neuroglial cell numbers, hypertrophy, ramification, and soma sizes bordering the continuously reducing lesion core and inflamed vasculature, possibly to shield their leaky phenotype. Astrocytes were also found to be releasing matrix metalloproteinase9 (MMP9), which stabilized matrix ligand β-dystroglycan (β-DG) in repaired or functional vessels. Changing neuroglia phenotypes, responses, motility, astrocytic recruitment of MMP9, and β-DG stabilization implies the role of communication between neuroglia and endothelium in recovering CBF, in the absence of neurons, in ET1 rats compared to Aβ+ET1 rats, which showed characteristics delayed neuroglial activation. Stimulation of timely neuroglial reactivity may serve as a viable strategy to compensate for the neuronal loss in restoring CBF in comorbid cases of ischemia and Aβ toxicity.
Collapse
Affiliation(s)
- Zareen Amtul
- Department of Anatomy and Cell Biology, University of Western Ontario, London, N6A 5C1, Canada.
| | - Jasmine Randhawa
- Department of Biology, University of Western Ontario, London, N6A 5B7, Canada
| | - Abdullah N Najdat
- Department of Biology, University of Western Ontario, London, N6A 5B7, Canada
| | - David J Hill
- Departments of Medicine, Physiology, and Pharmacology, and Pediatrics, University of Western Ontario, London, N6A 5C1, Canada
- Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Edith J Arany
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, N6A 5C1, Canada
| |
Collapse
|
21
|
Jafarian A, Litvak V, Cagnan H, Friston KJ, Zeidman P. Comparing dynamic causal models of neurovascular coupling with fMRI and EEG/MEG. Neuroimage 2020; 216:116734. [PMID: 32179105 PMCID: PMC7322559 DOI: 10.1016/j.neuroimage.2020.116734] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 01/09/2023] Open
Abstract
This technical note presents a dynamic causal modelling (DCM) procedure for evaluating different models of neurovascular coupling in the human brain - using combined electromagnetic (M/EEG) and functional magnetic resonance imaging (fMRI) data. This procedure compares the evidence for biologically informed models of neurovascular coupling using Bayesian model comparison. First, fMRI data are used to localise regionally specific neuronal responses. The coordinates of these responses are then used as the location priors in a DCM of electrophysiological responses elicited by the same paradigm. The ensuing estimates of model parameters are then used to generate neuronal drive functions, which model pre- or post-synaptic activity for each experimental condition. These functions form the input to a model of neurovascular coupling, whose parameters are estimated from the fMRI data. Crucially, this enables one to evaluate different models of neurovascular coupling, using Bayesian model comparison - asking, for example, whether instantaneous or delayed, pre- or post-synaptic signals mediate haemodynamic responses. We provide an illustrative application of the procedure using a single-subject auditory fMRI and MEG dataset. The code and exemplar data accompanying this technical note are available through the statistical parametric mapping (SPM) software.
Collapse
Affiliation(s)
| | - Vladimir Litvak
- The Wellcome Centre for Human Neuroimaging, University College London, UK
| | - Hayriye Cagnan
- MRC Brain Network Dynamics Unit (BNDU) at the University of Oxford, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Karl J Friston
- The Wellcome Centre for Human Neuroimaging, University College London, UK
| | - Peter Zeidman
- The Wellcome Centre for Human Neuroimaging, University College London, UK
| |
Collapse
|
22
|
Bell AH, Miller SL, Castillo-Melendez M, Malhotra A. The Neurovascular Unit: Effects of Brain Insults During the Perinatal Period. Front Neurosci 2020; 13:1452. [PMID: 32038147 PMCID: PMC6987380 DOI: 10.3389/fnins.2019.01452] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
The neurovascular unit (NVU) is a relatively recent concept in neuroscience that broadly describes the relationship between brain cells and their blood vessels. The NVU incorporates cellular and extracellular components involved in regulating cerebral blood flow and blood-brain barrier function. The NVU within the adult brain has attracted strong research interest and its structure and function is well described, however, the NVU in the developing brain over the fetal and neonatal period remains much less well known. One area of particular interest in perinatal brain development is the impact of known neuropathological insults on the NVU. The aim of this review is to synthesize existing literature to describe structure and function of the NVU in the developing brain, with a particular emphasis on exploring the effects of perinatal insults. Accordingly, a brief overview of NVU components and function is provided, before discussion of NVU development and how this may be affected by perinatal pathologies. We have focused this discussion around three common perinatal insults: prematurity, acute hypoxia, and chronic hypoxia. A greater understanding of processes affecting the NVU in the perinatal period may enable application of targeted therapies, as well as providing a useful basis for research as it expands further into this area.
Collapse
Affiliation(s)
- Alexander H. Bell
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Beiersdorfer A, Lohr C. AMPA Receptor-Mediated Ca 2+ Transients in Mouse Olfactory Ensheathing Cells. Front Cell Neurosci 2019; 13:451. [PMID: 31636544 PMCID: PMC6788192 DOI: 10.3389/fncel.2019.00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/20/2019] [Indexed: 11/13/2022] Open
Abstract
Ca2+ signaling in glial cells is primarily triggered by metabotropic pathways and the subsequent Ca2+ release from internal Ca2+ stores. However, there is upcoming evidence that various ion channels might also initiate Ca2+ rises in glial cells by Ca2+ influx. We investigated AMPA receptor-mediated inward currents and Ca2+ transients in olfactory ensheathing cells (OECs), a specialized glial cell population in the olfactory bulb (OB), using whole-cell voltage-clamp recordings and confocal Ca2+ imaging. By immunohistochemistry we showed immunoreactivity to the AMPA receptor subunits GluA1, GluA2 and GluA4 in OECs, suggesting the presence of AMPA receptors in OECs. Kainate-induced inward currents were mediated exclusively by AMPA receptors, as they were sensitive to the specific AMPA receptor antagonist, GYKI53655. Moreover, kainate-induced inward currents were reduced by the selective Ca2+-permeable AMPA receptor inhibitor, NASPM, suggesting the presence of functional Ca2+-permeable AMPA receptors in OECs. Additionally, kainate application evoked Ca2+ transients in OECs which were abolished in the absence of extracellular Ca2+, indicating that Ca2+ influx via Ca2+-permeable AMPA receptors contribute to kainate-induced Ca2+ transients. However, kainate-induced Ca2+ transients were partly reduced upon Ca2+ store depletion, leading to the conclusion that Ca2+ influx via AMPA receptor channels is essential to trigger Ca2+ transients in OECs, whereas Ca2+ release from internal stores contributes in part to the kainate-evoked Ca2+ response. Endogenous glutamate release by OSN axons initiated Ca2+ transients in OECs, equally mediated by metabotropic receptors (glutamatergic and purinergic) and AMPA receptors, suggesting a prominent role for AMPA receptor mediated Ca2+ signaling in axon-OEC communication.
Collapse
Affiliation(s)
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
24
|
Armstead WM, Vavilala MS. Improving Understanding and Outcomes of Traumatic Brain Injury Using Bidirectional Translational Research. J Neurotrauma 2019; 37:2372-2380. [PMID: 30834818 DOI: 10.1089/neu.2018.6119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent clinical trials in traumatic brain injury (TBI) have failed to demonstrate therapeutic effects even when there appears to be good evidence for efficacy in one or more appropriate pre-clinical models. While existing animal models mimic the injury, difficulties in translating promising therapeutics are exacerbated by the lack of alignment of discrete measures of the underlying injury pathology between the animal models and human subjects. To address this mismatch, we have incorporated reverse translation of bedside experience to inform pre-clinical studies in a large animal (pig) model of TBI that mirror practical clinical assessments. Cerebral autoregulation is impaired after TBI, contributing to poor outcome. Cerebral perfusion pressure (CPP) is often normalized by use of vasoactive agents to increase mean arterial pressure (MAP) and thereby limit impairment of cerebral autoregulation and neurological deficits. Vasoactive agents clinically used to elevate MAP to increase CPP after TBI, such as phenylephrine (Phe), dopamine (DA), norepinephrine (NE), and epinephrine (EPI), however, have not been compared sufficiently regarding effect on CPP, autoregulation, and survival after TBI, and clinically, current vasoactive agent use is variable. The cerebral effects of these clinically commonly used vasoactive agents are not known. This review will emphasize pediatric work and will describe bidirectional translational studies using a more human-like animal model of TBI to identify better therapeutic strategies to improve outcome post-injury. These studies in addition investigated the mechanism(s) involved in improvement of outcome in the setting of TBI.
Collapse
Affiliation(s)
- William M Armstead
- Department of Anesthesiology and Critical Care and University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Monica S Vavilala
- Department of Anesthesiology, Pediatrics, and Neurological Surgery, and Harborview Injury Prevention and Research Center, University of Washington, Seattle, Washington, USA
| |
Collapse
|
25
|
Turner MP, Hubbard NA, Sivakolundu DK, Himes LM, Hutchison JL, Hart J, Spence JS, Frohman EM, Frohman TC, Okuda DT, Rypma B. Preserved canonicality of the BOLD hemodynamic response reflects healthy cognition: Insights into the healthy brain through the window of Multiple Sclerosis. Neuroimage 2019; 190:46-55. [PMID: 29454932 DOI: 10.1016/j.neuroimage.2017.12.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022] Open
Abstract
The hemodynamic response function (HRF), a model of brain blood-flow changes in response to neural activity, reflects communication between neurons and the vasculature that supplies these neurons in part by means of glial cell intermediaries (e.g., astrocytes). Intact neural-vascular communication might play a central role in optimal cognitive performance. This hypothesis can be tested by comparing healthy individuals to those with known white-matter damage and impaired performance, as seen in Multiple Sclerosis (MS). Glial cell intermediaries facilitate the ability of neurons to adequately convey metabolic needs to cerebral vasculature for sufficient oxygen and nutrient perfusion. In this study, we isolated measurements of the HRF that could quantify the extent to which white-matter affects neural-vascular coupling and cognitive performance. HRFs were modeled from multiple brain regions during multiple cognitive tasks using piecewise cubic spline functions, an approach that minimized assumptions regarding HRF shape that may not be valid for diseased populations, and were characterized using two shape metrics (peak amplitude and time-to-peak). Peak amplitude was reduced, and time-to-peak was longer, in MS patients relative to healthy controls. Faster time-to-peak was predicted by faster reaction time, suggesting an important role for vasodilatory speed in the physiology underlying processing speed. These results support the hypothesis that intact neural-glial-vascular communication underlies optimal neural and cognitive functioning.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Nicholas A Hubbard
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Lyndahl M Himes
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - John Hart
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Elliot M Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Teresa C Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Darin T Okuda
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
26
|
Beiersdorfer A, Scheller A, Kirchhoff F, Lohr C. Panglial gap junctions between astrocytes and olfactory ensheathing cells mediate transmission of Ca 2+ transients and neurovascular coupling. Glia 2019; 67:1385-1400. [PMID: 30883940 DOI: 10.1002/glia.23613] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
Abstract
Astrocytes are arranged in highly organized gap junction-coupled networks, communicating via the propagation of Ca2+ waves. Astrocytes are gap junction-coupled not only to neighboring astrocytes, but also to oligodendrocytes, forming so-called panglial syncytia. It is not known, however, whether glial cells in panglial syncytia transmit information using Ca2+ signaling. We used confocal Ca2+ imaging to study intercellular communication between astrocytes and olfactory ensheathing glial cells (OECs) in in-toto preparations of the mouse olfactory bulb. Our results demonstrate that Ca2+ transients in juxtaglomerular astrocytes, evoked by local photolysis of "caged" ATP and "caged" tACPD, led to subsequent Ca2+ responses in OECs. This transmission of Ca2+ responses from astrocytes to OECs persisted in the presence of neuronal inhibition, but was absent when gap junctional coupling was suppressed with carbenoxolone. When Ca2+ transients were directly evoked in OECs by puff application of DHPG, they resulted in delayed Ca2+ responses in juxtaglomerular astrocytes, indicating that panglial transmission of Ca2+ signals occurred in a bidirectional manner. In addition, panglial transmission of Ca2+ signals from astrocytes to OECs resulted in vasoconstriction of OEC-associated blood vessels in the olfactory nerve layer. Our results demonstrate functional transmission of Ca2+ signals between different classes of glial cells within gap junction-coupled panglial networks and the resulting regulation of blood vessel diameter in the olfactory bulb.
Collapse
Affiliation(s)
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
27
|
Lu K, Xu G, Li W, Huo C, Liu Q, Lv Z, Wang Y, Li Z, Fan Y. Frequency-specific functional connectivity related to the rehabilitation task of stroke patients. Med Phys 2019; 46:1545-1560. [PMID: 30675729 DOI: 10.1002/mp.13398] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/21/2018] [Accepted: 01/17/2019] [Indexed: 01/15/2023] Open
Abstract
PURPOSES Stroke survivors suffering from deficits in motor control typically show persistent motor symptoms and limited functional abilities, which affect their functional independence in daily life. Active rehabilitation training is commonly applied for stroke patients to recover from motor dysfunction. The global connectivity reflects the synchronization of cardiac and respiratory activities in the cerebral regions. However, the understanding of the patterns of frequency-specific global connectivity (GC) and functional connectivity (FC) when performing active rehabilitation training is still far from comprehensive. This study was conducted to investigate the brain network patterns of stroke patients while performing a four-limb linkage rehabilitation training using the functional near-infrared spectroscopy (fNIRS) method. METHODS Two groups of stroke patients (LH, left hemiplegia; RH, right hemiplegia) and one healthy group were recruited to participate in this study. The wavelet phase coherence (WPCO) method was used to calculate the frequency-specific GC and FC of the brain network in four frequency bands: I, 0.6-2 Hz; II, 0.145-0.6 Hz; III, 0.052-0.145 Hz; and IV, 0.021-0.052 Hz. RESULTS Results showed that the healthy group exhibited lower WPCO in the four frequency bands during the task state than during the resting state (P < 0.05). Interestingly, the stroke groups showed increased WPCO in the frequency band II during the task state than during the resting state (P < 0.05). Moreover, significantly lower WPCO values in the frequency bands III (P < 0.05) were found during task state in the RH and LH groups compared with the healthy group. The RH group showed increased WPCO values in the frequency band II during the task state compared with the healthy group (P < 0.05). In addition, the RH group showed increased WPCO in the frequency bands I (P < 0.05) and II (P < 0.05) than the LH group. Notably, the rehabilitation task did not induce significant changes in stroke groups in the frequency band IV, which implied the neurogenic activity. CONCLUSIONS The reductions in FC suggested that the brain impairments caused a disturbed neurovascular coupling regulation in stroke patients. Results in frequency band IV suggested that the limb movement rehabilitation task intrinsically may not produce remarkable effect on the neurogenic activity of stroke patients. Significant differences in WPCO between the LH and RH groups suggested that the rehabilitation task should be specifically designed for individual rehabilitation. The frequency-specific FC methods based on WPCO would provide a potential approach to quantitatively assess the effect of rehabilitation tasks.
Collapse
Affiliation(s)
- Kuan Lu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 100086, Beijing, China
| | - Gongcheng Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 100086, Beijing, China
| | - Wenhao Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 100086, Beijing, China
| | - Congcong Huo
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids Beijing, Beijing, 100176, China
| | - Qianying Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 100086, Beijing, China
| | - Zeping Lv
- Rehabilitation Hospital, National Research Center for Rehabilitation Technical Aids, Beijing, 100176, China
| | - Yonghui Wang
- Qilu Hospital, ShanDong University, Jinan, 250061, China
| | - Zengyong Li
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids Beijing, Beijing, 100176, China.,Key Laboratory of Rehabilitation Aids Technology and System of the Ministry of Civil Affairs, Beijing, 100176, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 100086, Beijing, China.,Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids Beijing, Beijing, 100176, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| |
Collapse
|
28
|
Potokar M, Jorgačevski J, Zorec R. Astrocytes in Flavivirus Infections. Int J Mol Sci 2019; 20:ijms20030691. [PMID: 30736273 PMCID: PMC6386967 DOI: 10.3390/ijms20030691] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
Virus infections of the central nervous system (CNS) can manifest in various forms of inflammation, including that of the brain (encephalitis) and spinal cord (myelitis), all of which may have long-lasting deleterious consequences. Although the knowledge of how different viruses affect neural cells is increasing, understanding of the mechanisms by which cells respond to neurotropic viruses remains fragmented. Several virus types have the ability to infect neural tissue, and astrocytes, an abundant and heterogeneous neuroglial cell type and a key element providing CNS homeostasis, are one of the first CNS cell types to get infected. Astrocytes are morphologically closely aligned with neuronal synapses, blood vessels, and ventricle cavities, and thereby have the capacity to functionally interact with neurons and endothelial cells. In this review, we focus on the responses of astrocytes to infection by neurotropic flaviviruses, including tick-borne encephalitis virus (TBEV), Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV), which have all been confirmed to infect astrocytes and cause multiple CNS defects. Understanding these mechanisms may help design new strategies to better contain and mitigate virus- and astrocyte-dependent neuroinflammation.
Collapse
Affiliation(s)
- Maja Potokar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| |
Collapse
|
29
|
Zorec R, Županc TA, Verkhratsky A. Astrogliopathology in the infectious insults of the brain. Neurosci Lett 2018; 689:56-62. [PMID: 30096375 DOI: 10.1016/j.neulet.2018.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 12/28/2022]
Abstract
Astroglia, a heterogeneous type of neuroglia, play key homeostatic functions in the central nervous system (CNS) and represent an important defence system. Impaired homeostatic capacity of astrocytes manifests in diseases and this is mirrored in various astrocyte-based pathological features including reactive astrogliosis, astrodegeneration with astroglial atrophy and pathological remodelling of astrocytes. All of these manifestations are most prominently associated with infectious insults, mediated by bacteria, protozoa and viruses. Here we focus onto neurotropic viruses such as tick-borne encephalitis (TBEV) and Zika virus (ZIKV), both belonging to Flaviviridae and both causing severe neurological impairments. We argue that astrocytes provide a route through which neurotropic infectious agents attack the CNS, since they are anatomically associated with the blood-brain barrier and exhibit aerobic glycolysis, a metabolic specialisation of highly morphologically dynamic cells, which may provide a suitable metabolic milieu for proliferation of infectious agents, including viral bodies.
Collapse
Affiliation(s)
- Robert Zorec
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000, Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia
| | - Tatjana Avšič Županc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Alexei Verkhratsky
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000, Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
30
|
Manninen T, Havela R, Linne ML. Computational Models for Calcium-Mediated Astrocyte Functions. Front Comput Neurosci 2018; 12:14. [PMID: 29670517 PMCID: PMC5893839 DOI: 10.3389/fncom.2018.00014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/28/2018] [Indexed: 12/16/2022] Open
Abstract
The computational neuroscience field has heavily concentrated on the modeling of neuronal functions, largely ignoring other brain cells, including one type of glial cell, the astrocytes. Despite the short history of modeling astrocytic functions, we were delighted about the hundreds of models developed so far to study the role of astrocytes, most often in calcium dynamics, synchronization, information transfer, and plasticity in vitro, but also in vascular events, hyperexcitability, and homeostasis. Our goal here is to present the state-of-the-art in computational modeling of astrocytes in order to facilitate better understanding of the functions and dynamics of astrocytes in the brain. Due to the large number of models, we concentrated on a hundred models that include biophysical descriptions for calcium signaling and dynamics in astrocytes. We categorized the models into four groups: single astrocyte models, astrocyte network models, neuron-astrocyte synapse models, and neuron-astrocyte network models to ease their use in future modeling projects. We characterized the models based on which earlier models were used for building the models and which type of biological entities were described in the astrocyte models. Features of the models were compared and contrasted so that similarities and differences were more readily apparent. We discovered that most of the models were basically generated from a small set of previously published models with small variations. However, neither citations to all the previous models with similar core structure nor explanations of what was built on top of the previous models were provided, which made it possible, in some cases, to have the same models published several times without an explicit intention to make new predictions about the roles of astrocytes in brain functions. Furthermore, only a few of the models are available online which makes it difficult to reproduce the simulation results and further develop the models. Thus, we would like to emphasize that only via reproducible research are we able to build better computational models for astrocytes, which truly advance science. Our study is the first to characterize in detail the biophysical and biochemical mechanisms that have been modeled for astrocytes.
Collapse
Affiliation(s)
- Tiina Manninen
- Computational Neuroscience Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| | | | - Marja-Leena Linne
- Computational Neuroscience Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| |
Collapse
|
31
|
Romeiro FG, Ietsugu MDV, Franzoni LDC, Augusti L, Alvarez M, Santos LAA, Lima TB, Koga KH, Moriguchi SM, Caramori CA, Silva GF, Betting LEGG. Which of the branched-chain amino acids increases cerebral blood flow in hepatic encephalopathy? A double-blind randomized trial. Neuroimage Clin 2018; 19:302-310. [PMID: 30013913 PMCID: PMC6044187 DOI: 10.1016/j.nicl.2018.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/07/2018] [Accepted: 03/21/2018] [Indexed: 02/06/2023]
Abstract
Branched-chain amino acids increase the brain perfusion of patients with hepatic encephalopathy (HE), but the amino acid and the mechanisms involved are still unknown. This study compared brain perfusion and clinical improvement during leucine or isoleucine supplementation. After randomization, 27 subjects with cirrhosis and HE received leucine or isoleucine supplements for one year. Brain single Photon Emission Computed Tomography (SPECT) and dynamic brain scintigraphy (DBS) were performed pretreatment and at 1, 8 and 12 months of supplementation. Brain perfusion was increased only in the isoleucine group at 8 months of treatment by both SPECT and DBS (p < 0.001 and p = 0.05, respectively) and by SPECT at the 12th month (p < 0.05). This was associated with hepatic encephalopathy improvement at 8 and 12 months (p = 0.008 and 0.004, respectively), which was not observed in the leucine group (p = 0.313 and 0.055, respectively). Isoleucine supplementation achieved a better impact on brain perfusion restoration in HE.
Collapse
Key Words
- AC, arm circumference
- APMT, adductor pollicis muscle thickness
- BCAA, branched-chain amino acids
- BCKA, branched-chain ketoacids
- BMI, body mass index
- Branched-chain amino acids
- CAMA, corrected mid-arm muscle area
- CBF, cerebral blood flow
- Cerebral blood flow
- EEG, electroencephalogram
- FDR, false discovery rate
- GDH, glutamate dehydrogenase
- GLN, glutamine
- GLU, glutamate
- HE, hepatic encephalopathy
- HGS, handgrip strength
- HPLC, high-performance liquid chromatography
- HRQoL, health-related quality of life
- Hepatic encephalopathy
- Liver cirrhosis
- MAMC, mid-arm muscle circumference
- MELD, Model of End-Stage Liver Disease
- NH3, ammonia
- PDH, pyruvate dehydrogenase complex
- ROIs, regions of interest
- ROS, reactive oxygen species
- SF-36, 36-item Short-Form General Health Survey
- SPECT, Single Photon Emission Computed Tomography
- SPM12, Statistical Parametrical Mapping 12
- TCA, tricarboxylic acid
- TSF, triceps skinfold
- α-KG, α-ketoglutarate
- αKGDH, α-ketoglutarate dehydrogenase complex
Collapse
Affiliation(s)
- Fernando Gomes Romeiro
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil.
| | - Marjorie do Val Ietsugu
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Letícia de Campos Franzoni
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Laís Augusti
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Matheus Alvarez
- Department of Physics and Biophysics, Botucatu Biosciences Institute, UNESP - Univ Estadual Paulista, Rua Prof. Dr. Antonio Celso Wagner Zanin, s/n, Botucatu, São Paulo 18618-689, Brazil.
| | - Lívia Alves Amaral Santos
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Talles Bazeia Lima
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Kátia Hiromoto Koga
- Department of Tropical Diseases and Imaging Diagnosis, Botucatu Medical School, UNESP - Univ Estadual Paulista, Av. Prof. Mário Rubens Guimarães Montenegro, s/n. Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil.
| | - Sônia Marta Moriguchi
- Department of Tropical Diseases and Imaging Diagnosis, Botucatu Medical School, UNESP - Univ Estadual Paulista, Av. Prof. Mário Rubens Guimarães Montenegro, s/n. Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Carlos Antonio Caramori
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil
| | - Giovanni Faria Silva
- Department of Internal Medicine, Botucatu Medical School, UNESP - Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil.
| | - Luiz Eduardo Gomes Garcia Betting
- Department of Neurology, Psychology and Psychiatry, Botucatu Medical School, UNESP - Univ Estadual Paulista, Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, São Paulo 18618-687, Brazil.
| |
Collapse
|
32
|
Coucha M, Abdelsaid M, Ward R, Abdul Y, Ergul A. Impact of Metabolic Diseases on Cerebral Circulation: Structural and Functional Consequences. Compr Physiol 2018; 8:773-799. [PMID: 29687902 DOI: 10.1002/cphy.c170019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolic diseases including obesity, insulin resistance, and diabetes have profound effects on cerebral circulation. These diseases not only affect the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression but also alter the physiology of blood vessels resulting in compromised myogenic reactivity, neurovascular uncoupling, and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain rapidly occur. This overview is organized into sections describing cerebrovascular architecture, physiology, and BBB in these diseases. In each section, we review these properties starting with larger arteries moving into smaller vessels. Where information is available, we review in the order of obesity, insulin resistance, and diabetes. We also tried to include information on biological variables such as the sex of the animal models noted since most of the information summarized was obtained using male animals. © 2018 American Physiological Society. Compr Physiol 8:773-799, 2018.
Collapse
Affiliation(s)
- Maha Coucha
- South University, School of Pharmacy, Savannah, Georgia, USA
| | | | - Rebecca Ward
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yasir Abdul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
33
|
Zorec R, Parpura V, Verkhratsky A. Astroglial vesicular network: evolutionary trends, physiology and pathophysiology. Acta Physiol (Oxf) 2018; 222. [PMID: 28665546 DOI: 10.1111/apha.12915] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/17/2017] [Accepted: 06/24/2017] [Indexed: 12/13/2022]
Abstract
Intracellular organelles, including secretory vesicles, emerged when eukaryotic cells evolved some 3 billion years ago. The primordial organelles that evolved in Archaea were similar to endolysosomes, which developed, arguably, for specific metabolic tasks, including uptake, metabolic processing, storage and disposal of molecules. In comparison with prokaryotes, cell volume of eukaryotes increased by several orders of magnitude and vesicle traffic emerged to allow for communication between distant intracellular locations. Lysosomes, first described in 1955, a prominent intermediate of endo- and exocytotic pathways, operate virtually in all eukaryotic cells including astroglia, the most heterogeneous type of homeostatic glia in the central nervous system. Astrocytes support neuronal network activity in particular through elaborated secretion, based on a complex intracellular vesicle network dynamics. Deranged homeostasis underlies disease and astroglial vesicle traffic contributes to the pathophysiology of neurodegenerative (Alzheimer's disease, Huntington's disease), neurodevelopmental diseases (intellectual deficiency, Rett's disease) and neuroinfectious (Zika virus) disorders. This review addresses astroglial cell-autonomous vesicular traffic network, as well as its into primary and secondary vesicular network defects in diseases, and considers this network as a target for developing new therapies for neurological conditions.
Collapse
Affiliation(s)
- R. Zorec
- Laboratory of Neuroendocrinology and Molecular Cell Physiology; Institute of Pathophysiology; University of Ljubljana; Ljubljana Slovenia
- Celica; BIOMEDICAL; Ljubljana Slovenia
| | - V. Parpura
- Department of Neurobiology; Civitan International Research Center and Center for Glial Biology in Medicine; Evelyn F. McKnight Brain Institute; Atomic Force Microscopy and Nanotechnology Laboratories; University of Alabama; Birmingham AL USA
| | - A. Verkhratsky
- Laboratory of Neuroendocrinology and Molecular Cell Physiology; Institute of Pathophysiology; University of Ljubljana; Ljubljana Slovenia
- Celica; BIOMEDICAL; Ljubljana Slovenia
- Faculty of Biology; Medicine and Health; The University of Manchester; Manchester UK
- Achucarro Center for Neuroscience; IKERBASQUE; Basque Foundation for Science; Bilbao Spain
- Department of Neurosciences; University of the Basque Country UPV/EHU and CIBERNED; Leioa Spain
| |
Collapse
|
34
|
Verisokin AY, Verveyko DV, Postnov DE. Turing-like structures in a functional model of cortical spreading depression. Phys Rev E 2018; 96:062409. [PMID: 29347421 DOI: 10.1103/physreve.96.062409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Indexed: 11/07/2022]
Abstract
Cortical spreading depression (CSD) along with migraine waves and spreading depolarization events with stroke or injures are the front-line examples of extreme physiological behaviors of the brain cortex which manifest themselves via the onset and spreading of localized areas of neuronal hyperactivity followed by their depression. While much is known about the physiological pathways involved, the dynamical mechanisms of the formation and evolution of complex spatiotemporal patterns during CSD are still poorly understood, in spite of the number of modeling studies that have been already performed. Recently we have proposed a relatively simple mathematical model of cortical spreading depression which counts the effects of neurovascular coupling and cerebral blood flow redistribution during CSD. In the present study, we address the main dynamical consequences of newly included pathways, namely, the changes in the formation and propagation speed of the CSD front and the pattern formation features in two dimensions. Our most notable finding is that the combination of vascular-mediated spatial coupling with local regulatory mechanisms results in the formation of stationary Turing-like patterns during a CSD event.
Collapse
Affiliation(s)
- A Yu Verisokin
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, 305000, Kursk, Russia
| | - D V Verveyko
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, 305000, Kursk, Russia
| | - D E Postnov
- Saratov State University, Astrakhanskaya st., 83, 410012, Saratov, Russia
| |
Collapse
|
35
|
De Cicco V, Tramonti Fantozzi MP, Cataldo E, Barresi M, Bruschini L, Faraguna U, Manzoni D. Trigeminal, Visceral and Vestibular Inputs May Improve Cognitive Functions by Acting through the Locus Coeruleus and the Ascending Reticular Activating System: A New Hypothesis. Front Neuroanat 2018; 11:130. [PMID: 29358907 PMCID: PMC5766640 DOI: 10.3389/fnana.2017.00130] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
It is known that sensory signals sustain the background discharge of the ascending reticular activating system (ARAS) which includes the noradrenergic locus coeruleus (LC) neurons and controls the level of attention and alertness. Moreover, LC neurons influence brain metabolic activity, gene expression and brain inflammatory processes. As a consequence of the sensory control of ARAS/LC, stimulation of a sensory channel may potential influence neuronal activity and trophic state all over the brain, supporting cognitive functions and exerting a neuroprotective action. On the other hand, an imbalance of the same input on the two sides may lead to an asymmetric hemispheric excitability, leading to an impairment in cognitive functions. Among the inputs that may drive LC neurons and ARAS, those arising from the trigeminal region, from visceral organs and, possibly, from the vestibular system seem to be particularly relevant in regulating their activity. The trigeminal, visceral and vestibular control of ARAS/LC activity may explain why these input signals: (1) affect sensorimotor and cognitive functions which are not directly related to their specific informational content; and (2) are effective in relieving the symptoms of some brain pathologies, thus prompting peripheral activation of these input systems as a complementary approach for the treatment of cognitive impairments and neurodegenerative disorders.
Collapse
Affiliation(s)
- Vincenzo De Cicco
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Maria P Tramonti Fantozzi
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Massimo Barresi
- Institut des Maladie Neurodégénératives, University of Bordeaux, Bordeaux, France
| | - Luca Bruschini
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Ugo Faraguna
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy.,Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Diego Manzoni
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
36
|
Schellekens W, Ramsey NF, van Wezel RJA, Raemaekers M. Changes in fMRI BOLD dynamics reflect anticipation to moving objects. Neuroimage 2017; 161:188-195. [PMID: 27620983 DOI: 10.1016/j.neuroimage.2016.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/22/2016] [Accepted: 09/08/2016] [Indexed: 11/15/2022] Open
Abstract
The human brain is thought to respond differently to novel versus predictable neural input. In human visual cortex, neural response amplitude to visual input might be determined by the degree of predictability. We investigated how fMRI BOLD responses in human early visual cortex reflect the anticipation of a single moving bar's trajectory. We found that BOLD signals decreased linearly from onset to offset of the stimulus trajectory. Moreover, decreased amplitudes of BOLD responses coincided with an increased initial dip as the stimulus moved along its trajectory. Importantly, motion anticipation effects were absent, when motion coherence was disrupted by means of stimulus contrast reversals. These results show that human early visual cortex anticipates the trajectory of a coherently moving object at the initial stages of visual motion processing. The results can be explained by suppression of predictable input, plausibly underlying the formation of stable visual percepts.
Collapse
Affiliation(s)
- W Schellekens
- Brain Center Rudolf Magnus, Department of Neurology, UMC Utrecht, Utrecht, The Netherlands.
| | - N F Ramsey
- Brain Center Rudolf Magnus, Department of Neurology, UMC Utrecht, Utrecht, The Netherlands
| | - R J A van Wezel
- Department of Biophysics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands; Biomedical Signals and Systems, MIRA, University of Twente, Enschede, The Netherlands
| | - M Raemaekers
- Brain Center Rudolf Magnus, Department of Neurology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
37
|
Choi JH, Cho HY, Choi JW. Microdevice Platform for In Vitro Nervous System and Its Disease Model. Bioengineering (Basel) 2017; 4:E77. [PMID: 28952555 PMCID: PMC5615323 DOI: 10.3390/bioengineering4030077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 01/09/2023] Open
Abstract
The development of precise microdevices can be applied to the reconstruction of in vitro human microenvironmental systems with biomimetic physiological conditions that have highly tunable spatial and temporal features. Organ-on-a-chip can emulate human physiological functions, particularly at the organ level, as well as its specific roles in the body. Due to the complexity of the structure of the central nervous system and its intercellular interaction, there remains an urgent need for the development of human brain or nervous system models. Thus, various microdevice models have been proposed to mimic actual human brain physiology, which can be categorized as nervous system-on-a-chip. Nervous system-on-a-chip platforms can prove to be promising technologies, through the application of their biomimetic features to the etiology of neurodegenerative diseases. This article reviews the microdevices for nervous system-on-a-chip platform incorporated with neurobiology and microtechnology, including microfluidic designs that are biomimetic to the entire nervous system. The emulation of both neurodegenerative disorders and neural stem cell behavior patterns in micro-platforms is also provided, which can be used as a basis to construct nervous system-on-a-chip.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
| | - Hyeon-Yeol Cho
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 610 Taylor Road, Piscataway, NJ 08854, USA.
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
| |
Collapse
|
38
|
Glutamate-Mediated Blood-Brain Barrier Opening: Implications for Neuroprotection and Drug Delivery. J Neurosci 2017; 36:7727-39. [PMID: 27445149 DOI: 10.1523/jneurosci.0587-16.2016] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The blood-brain barrier is a highly selective anatomical and functional interface allowing a unique environment for neuro-glia networks. Blood-brain barrier dysfunction is common in most brain disorders and is associated with disease course and delayed complications. However, the mechanisms underlying blood-brain barrier opening are poorly understood. Here we demonstrate the role of the neurotransmitter glutamate in modulating early barrier permeability in vivo Using intravital microscopy, we show that recurrent seizures and the associated excessive glutamate release lead to increased vascular permeability in the rat cerebral cortex, through activation of NMDA receptors. NMDA receptor antagonists reduce barrier permeability in the peri-ischemic brain, whereas neuronal activation using high-intensity magnetic stimulation increases barrier permeability and facilitates drug delivery. Finally, we conducted a double-blind clinical trial in patients with malignant glial tumors, using contrast-enhanced magnetic resonance imaging to quantitatively assess blood-brain barrier permeability. We demonstrate the safety of stimulation that efficiently increased blood-brain barrier permeability in 10 of 15 patients with malignant glial tumors. We suggest a novel mechanism for the bidirectional modulation of brain vascular permeability toward increased drug delivery and prevention of delayed complications in brain disorders. SIGNIFICANCE STATEMENT In this study, we reveal a new mechanism that governs blood-brain barrier (BBB) function in the rat cerebral cortex, and, by using the discovered mechanism, we demonstrate bidirectional control over brain endothelial permeability. Obviously, the clinical potential of manipulating BBB permeability for neuroprotection and drug delivery is immense, as we show in preclinical and proof-of-concept clinical studies. This study addresses an unmet need to induce transient BBB opening for drug delivery in patients with malignant brain tumors and effectively facilitate BBB closure in neurological disorders.
Collapse
|
39
|
Papadopoulos P, Tong XK, Imboden H, Hamel E. Losartan improves cerebrovascular function in a mouse model of Alzheimer's disease with combined overproduction of amyloid-β and transforming growth factor-β1. J Cereb Blood Flow Metab 2017; 37:1959-1970. [PMID: 27389178 PMCID: PMC5464692 DOI: 10.1177/0271678x16658489] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alterations of the renin-angiotensin system have been implicated in the pathogenesis of Alzheimer's disease. We tested the efficacy of losartan (10 mg/kg/day for three months), a selective angiotensin II type 1 receptor antagonist, in alleviating cerebrovascular and cognitive deficits in double-transgenic mice (six months at endpoint) that overexpress a mutated form of the human amyloid precursor protein (APPSwe,Ind) and a constitutively active form of the transforming growth factor-β1, thereafter named A/T mice. Losartan rescued cerebrovascular reactivity, particularly the dilatory responses, but failed to attenuate astroglial activation and to normalize the neurovascular uncoupling response to sensory stimulation. The cognitive deficits of A/T mice were not restored by losartan nor were the increased brain levels of soluble and insoluble Aβ1-40 and Aβ1-42 peptides normalized. Our results are the first to demonstrate the capacity of losartan to improve cerebrovascular reactivity in an Alzheimer's disease mouse model of combined Aβ-induced vascular oxidative stress and transforming growth factor-β1-mediated vascular fibrosis. These data suggest that losartan may be promising for restoring cerebrovascular function in patients with vascular diseases at risk for vascular dementia or Alzheimer's disease. However, a combined therapy may be warranted for rescuing both vascular and cognitive deficits in a multifaceted pathology like Alzheimer's disease.
Collapse
Affiliation(s)
- Panayiota Papadopoulos
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Xin-Kang Tong
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Hans Imboden
- 2 Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Edith Hamel
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| |
Collapse
|
40
|
Occhieppo VB, Marchese NA, Rodríguez ID, Basmadjian OM, Baiardi G, Bregonzio C. Neurovascular unit alteration in somatosensory cortex and enhancement of thermal nociception induced by amphetamine involves central AT1receptor activation. Eur J Neurosci 2017; 45:1586-1593. [DOI: 10.1111/ejn.13594] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Victoria Belén Occhieppo
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Natalia Andrea Marchese
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Iara Diamela Rodríguez
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Osvaldo Martin Basmadjian
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Gustavo Baiardi
- Laboratorio de Neurofarmacología (IIBYT-CONICET); Universidad Nacional de Córdoba Facultad de Ciencias Químicas; Universidad Católica de Córdoba; Córdoba Argentina
| | - Claudia Bregonzio
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| |
Collapse
|
41
|
Abstract
This paper revisits the dynamic causal modelling of fMRI timeseries by replacing the usual (Taylor) approximation to neuronal dynamics with a neural mass model of the canonical microcircuit. This provides a generative or dynamic causal model of laminar specific responses that can generate haemodynamic and electrophysiological measurements. In principle, this allows the fusion of haemodynamic and (event related or induced) electrophysiological responses. Furthermore, it enables Bayesian model comparison of competing hypotheses about physiologically plausible synaptic effects; for example, does attentional modulation act on superficial or deep pyramidal cells – or both? In this technical note, we describe the resulting dynamic causal model and provide an illustrative application to the attention to visual motion dataset used in previous papers. Our focus here is on how to answer long-standing questions in fMRI; for example, do haemodynamic responses reflect extrinsic (afferent) input from distant cortical regions, or do they reflect intrinsic (recurrent) neuronal activity? To what extent do inhibitory interneurons contribute to neurovascular coupling? What is the relationship between haemodynamic responses and the frequency of induced neuronal activity? This paper does not pretend to answer these questions; rather it shows how they can be addressed using neural mass models of fMRI timeseries. This paper describes a DCM for fMRI based on neural mass models and canonical microcircuits. This enables the (Bayesian) fusion of EEG and fMRI data. That encompasses the formal modelling of neurovascular coupling. Offers a surprising insight into the relationship between haemodynamic and electrophysiological responses.
Collapse
|
42
|
McBryde FD, Malpas SC, Paton JFR. Intracranial mechanisms for preserving brain blood flow in health and disease. Acta Physiol (Oxf) 2017; 219:274-287. [PMID: 27172364 DOI: 10.1111/apha.12706] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/03/2016] [Accepted: 05/06/2016] [Indexed: 12/19/2022]
Abstract
The brain is an exceptionally energetically demanding organ with little metabolic reserve, and multiple systems operate to protect and preserve the brain blood supply. But how does the brain sense its own perfusion? In this review, we discuss how the brain may harness the cardiovascular system to counter threats to cerebral perfusion sensed via intracranial pressure (ICP), cerebral oxygenation and ischaemia. Since the work of Cushing over 100 years ago, the existence of brain baroreceptors capable of eliciting increases in sympathetic outflow and blood pressure has been hypothesized. In the clinic, this response has generally been thought to occur only in extremis, to perfuse the severely ischaemic brain as cerebral autoregulation fails. We review evidence that pressor responses may also occur with smaller, physiologically relevant increases in ICP. The incoming brain oxygen supply is closely monitored by the carotid chemoreceptors; however, hypoxia and other markers of ischaemia are also sensed intrinsically by astrocytes or other support cells within brain tissue itself and elicit reactive hyperaemia. Recent studies suggest that astrocytic oxygen signalling within the brainstem may directly affect sympathetic nerve activity and blood pressure. We speculate that local cerebral oxygen tension is a major determinant of the mean level of arterial pressure and discuss recent evidence that this may be the case. We conclude that intrinsic intra- and extra-cranial mechanisms sense and integrate information about hypoxia/ischaemia and ICP and play a major role in determining the long-term level of sympathetic outflow and arterial pressure, to optimize cerebral perfusion.
Collapse
Affiliation(s)
- F. D. McBryde
- Department of Physiology; Faculty of Medical and Health Sciences; University of Auckland; Auckland New Zealand
- School of Physiology, Pharmacology & Neuroscience; Biomedical Sciences; University of Bristol; Bristol UK
| | - S. C. Malpas
- Department of Physiology; Faculty of Medical and Health Sciences; University of Auckland; Auckland New Zealand
| | - J. F. R. Paton
- Department of Physiology; Faculty of Medical and Health Sciences; University of Auckland; Auckland New Zealand
- School of Physiology, Pharmacology & Neuroscience; Biomedical Sciences; University of Bristol; Bristol UK
| |
Collapse
|
43
|
Inflammatory Activation of Microglia and Astrocytes in Manganese Neurotoxicity. ADVANCES IN NEUROBIOLOGY 2017; 18:159-181. [PMID: 28889267 DOI: 10.1007/978-3-319-60189-2_8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotoxicity due to excessive exposure to manganese (Mn) has been described as early as 1837 (Couper, Br Ann Med Pharm Vital Stat Gen Sci 1:41-42, 1837). Extensive research over the past two decades has revealed that Mn-induced neurological injury involves complex pathophysiological signaling mechanisms between neurons and glial cells. Glial cells are an important target of Mn in the brain, both for sequestration of the metal, as well as for activating inflammatory signaling pathways that damage neurons through overproduction of numerous reactive oxygen and nitrogen species and inflammatory cytokines. Understanding how these pathways are regulated in glial cells during Mn exposure is critical to determining the mechanisms underlying permanent neurological dysfunction stemming from excess exposure. The subject of this review will be to delineate mechanisms by which Mn interacts with glial cells to perturb neuronal function, with a particular emphasis on neuroinflammation and neuroinflammatory signaling between distinct populations of glial cells.
Collapse
|
44
|
Hladky SB, Barrand MA. Fluid and ion transfer across the blood-brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS 2016; 13:19. [PMID: 27799072 PMCID: PMC5508927 DOI: 10.1186/s12987-016-0040-3] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/01/2016] [Indexed: 12/24/2022] Open
Abstract
The two major interfaces separating brain and blood have different primary roles. The choroid plexuses secrete cerebrospinal fluid into the ventricles, accounting for most net fluid entry to the brain. Aquaporin, AQP1, allows water transfer across the apical surface of the choroid epithelium; another protein, perhaps GLUT1, is important on the basolateral surface. Fluid secretion is driven by apical Na+-pumps. K+ secretion occurs via net paracellular influx through relatively leaky tight junctions partially offset by transcellular efflux. The blood-brain barrier lining brain microvasculature, allows passage of O2, CO2, and glucose as required for brain cell metabolism. Because of high resistance tight junctions between microvascular endothelial cells transport of most polar solutes is greatly restricted. Because solute permeability is low, hydrostatic pressure differences cannot account for net fluid movement; however, water permeability is sufficient for fluid secretion with water following net solute transport. The endothelial cells have ion transporters that, if appropriately arranged, could support fluid secretion. Evidence favours a rate smaller than, but not much smaller than, that of the choroid plexuses. At the blood-brain barrier Na+ tracer influx into the brain substantially exceeds any possible net flux. The tracer flux may occur primarily by a paracellular route. The blood-brain barrier is the most important interface for maintaining interstitial fluid (ISF) K+ concentration within tight limits. This is most likely because Na+-pumps vary the rate at which K+ is transported out of ISF in response to small changes in K+ concentration. There is also evidence for functional regulation of K+ transporters with chronic changes in plasma concentration. The blood-brain barrier is also important in regulating HCO3- and pH in ISF: the principles of this regulation are reviewed. Whether the rate of blood-brain barrier HCO3- transport is slow or fast is discussed critically: a slow transport rate comparable to those of other ions is favoured. In metabolic acidosis and alkalosis variations in HCO3- concentration and pH are much smaller in ISF than in plasma whereas in respiratory acidosis variations in pHISF and pHplasma are similar. The key similarities and differences of the two interfaces are summarized.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD UK
| |
Collapse
|
45
|
Lecrux C, Hamel E. Neuronal networks and mediators of cortical neurovascular coupling responses in normal and altered brain states. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150350. [PMID: 27574304 PMCID: PMC5003852 DOI: 10.1098/rstb.2015.0350] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 12/18/2022] Open
Abstract
Brain imaging techniques that use vascular signals to map changes in neuronal activity, such as blood oxygenation level-dependent functional magnetic resonance imaging, rely on the spatial and temporal coupling between changes in neurophysiology and haemodynamics, known as 'neurovascular coupling (NVC)'. Accordingly, NVC responses, mapped by changes in brain haemodynamics, have been validated for different stimuli under physiological conditions. In the cerebral cortex, the networks of excitatory pyramidal cells and inhibitory interneurons generating the changes in neural activity and the key mediators that signal to the vascular unit have been identified for some incoming afferent pathways. The neural circuits recruited by whisker glutamatergic-, basal forebrain cholinergic- or locus coeruleus noradrenergic pathway stimulation were found to be highly specific and discriminative, particularly when comparing the two modulatory systems to the sensory response. However, it is largely unknown whether or not NVC is still reliable when brain states are altered or in disease conditions. This lack of knowledge is surprising since brain imaging is broadly used in humans and, ultimately, in conditions that deviate from baseline brain function. Using the whisker-to-barrel pathway as a model of NVC, we can interrogate the reliability of NVC under enhanced cholinergic or noradrenergic modulation of cortical circuits that alters brain states.This article is part of the themed issue 'Interpreting BOLD: a dialogue between cognitive and cellular neuroscience'.
Collapse
Affiliation(s)
- C Lecrux
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, Quebec, Canada H3A 2B4
| | - E Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, Quebec, Canada H3A 2B4
| |
Collapse
|
46
|
Klingner CM, Brodoehl S, Witte OW. The importance of the negative blood-oxygenation-level-dependent (BOLD) response in the somatosensory cortex. Rev Neurosci 2016; 26:647-53. [PMID: 26057216 DOI: 10.1515/revneuro-2015-0002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/17/2015] [Indexed: 11/15/2022]
Abstract
In recent years, multiple studies have shown task-induced negative blood-oxygenation-level-dependent responses (NBRs) in multiple brain regions in humans and animals. Converging evidence suggests that task-induced NBRs can be interpreted in terms of decreased neuronal activity. However, the vascular and metabolic dynamics and functional importance of the NBR are highly debated. Here, we review studies investigating the origin and functional importance of the NBR, with special attention to the somatosensory cortex.
Collapse
|
47
|
Di Marco LY, Farkas E, Martin C, Venneri A, Frangi AF. Is Vasomotion in Cerebral Arteries Impaired in Alzheimer's Disease? J Alzheimers Dis 2016; 46:35-53. [PMID: 25720414 PMCID: PMC4878307 DOI: 10.3233/jad-142976] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A substantial body of evidence supports the hypothesis of a vascular component in the pathogenesis of Alzheimer’s disease (AD). Cerebral hypoperfusion and blood-brain barrier dysfunction have been indicated as key elements of this pathway. Cerebral amyloid angiopathy (CAA) is a cerebrovascular disorder, frequent in AD, characterized by the accumulation of amyloid-β (Aβ) peptide in cerebral blood vessel walls. CAA is associated with loss of vascular integrity, resulting in impaired regulation of cerebral circulation, and increased susceptibility to cerebral ischemia, microhemorrhages, and white matter damage. Vasomotion— the spontaneous rhythmic modulation of arterial diameter, typically observed in arteries/arterioles in various vascular beds including the brain— is thought to participate in tissue perfusion and oxygen delivery regulation. Vasomotion is impaired in adverse conditions such as hypoperfusion and hypoxia. The perivascular and glymphatic pathways of Aβ clearance are thought to be driven by the systolic pulse. Vasomotion produces diameter changes of comparable amplitude, however at lower rates, and could contribute to these mechanisms of Aβ clearance. In spite of potential clinical interest, studies addressing cerebral vasomotion in the context of AD/CAA are limited. This study reviews the current literature on vasomotion, and hypothesizes potential paths implicating impaired cerebral vasomotion in AD/CAA. Aβ and oxidative stress cause vascular tone dysregulation through direct effects on vascular cells, and indirect effects mediated by impaired neurovascular coupling. Vascular tone dysregulation is further aggravated by cholinergic deficit and results in depressed cerebrovascular reactivity and (possibly) impaired vasomotion, aggravating regional hypoperfusion and promoting further Aβ and oxidative stress accumulation.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Chris Martin
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Annalena Venneri
- Department of Neuroscience, University of Sheffield, Sheffield, UK.,IRCCS, Fondazione Ospedale S. Camillo, Venice, Italy
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
48
|
Logica T, Riviere S, Holubiec MI, Castilla R, Barreto GE, Capani F. Metabolic Changes Following Perinatal Asphyxia: Role of Astrocytes and Their Interaction with Neurons. Front Aging Neurosci 2016; 8:116. [PMID: 27445788 PMCID: PMC4921470 DOI: 10.3389/fnagi.2016.00116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/03/2016] [Indexed: 11/13/2022] Open
Abstract
Perinatal Asphyxia (PA) represents an important cause of severe neurological deficits including delayed mental and motor development, epilepsy, major cognitive deficits and blindness. The interaction between neurons, astrocytes and endothelial cells plays a central role coupling energy supply with changes in neuronal activity. Traditionally, experimental research focused on neurons, whereas astrocytes have been more related to the damage mechanisms of PA. Astrocytes carry out a number of functions that are critical to normal nervous system function, including uptake of neurotransmitters, regulation of pH and ion concentrations, and metabolic support for neurons. In this work, we aim to review metabolic neuron-astrocyte interactions with the purpose of encourage further research in this area in the context of PA, which is highly complex and its mechanisms and pathways have not been fully elucidated to this day.
Collapse
Affiliation(s)
- Tamara Logica
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Stephanie Riviere
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Mariana I Holubiec
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Rocío Castilla
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá Bogotá, Colombia
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABABuenos Aires, Argentina; Departamento de Biología, Universidad Argentina JF KennedyBuenos Aires, Argentina; Investigador Asociado, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|
49
|
Daulatzai MA. Dysfunctional Sensory Modalities, Locus Coeruleus, and Basal Forebrain: Early Determinants that Promote Neuropathogenesis of Cognitive and Memory Decline and Alzheimer’s Disease. Neurotox Res 2016; 30:295-337. [DOI: 10.1007/s12640-016-9643-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
50
|
Filosa JA, Morrison HW, Iddings JA, Du W, Kim KJ. Beyond neurovascular coupling, role of astrocytes in the regulation of vascular tone. Neuroscience 2016; 323:96-109. [PMID: 25843438 PMCID: PMC4592693 DOI: 10.1016/j.neuroscience.2015.03.064] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/24/2015] [Accepted: 03/27/2015] [Indexed: 12/22/2022]
Abstract
The brain possesses two intricate mechanisms that fulfill its continuous metabolic needs: cerebral autoregulation, which ensures constant cerebral blood flow over a wide range of arterial pressures and functional hyperemia, which ensures rapid delivery of oxygen and glucose to active neurons. Over the past decade, a number of important studies have identified astrocytes as key intermediaries in neurovascular coupling (NVC), the mechanism by which active neurons signal blood vessels to change their diameter. Activity-dependent increases in astrocytic Ca(2+) activity are thought to contribute to the release of vasoactive substances that facilitate arteriole vasodilation. A number of vasoactive signals have been identified and their role on vessel caliber assessed both in vitro and in vivo. In this review, we discuss mechanisms implicating astrocytes in NVC-mediated vascular responses, limitations encountered as a result of the challenges in maintaining all the constituents of the neurovascular unit intact and deliberate current controversial findings disputing a main role for astrocytes in NVC. Finally, we briefly discuss the potential role of pericytes and microglia in NVC-mediated processes.
Collapse
Affiliation(s)
- J A Filosa
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States.
| | - H W Morrison
- University of Arizona, 1305 N. Martin Avenue, P.O. Box 210203, Tucson, AZ 85721, United States
| | - J A Iddings
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| | - W Du
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| | - K J Kim
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| |
Collapse
|