1
|
Vachha BA, Kumar VA, Pillai JJ, Shimony J, Tanabe J, Sair HI. Resting-State Functional MRI: Current State, Controversies, Limitations, and Future Directions- AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2024. [PMID: 39660823 DOI: 10.2214/ajr.24.32163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Resting-state functional MRI (rs-fMRI), a promising method for interrogating different brain functional networks from a single MRI acquisition, is increasingly utilized in clinical presurgical and other pretherapeutic brain mapping. However, challenges in standardization of acquisition, preprocessing, and analysis methods across centers, and variability in results interpretation, complicate its clinical use. Additionally, inherent problems regarding reliability of language lateralization, interpatient variability of cognitive network representation, dynamic aspects of intranetwork and internetwork connectivity, and effects of neurovascular uncoupling on network detection still must be overcome. Although deep-learning solutions and further methodologic standardization will help address these issues, rs-fMRI remains generally considered an adjunct to task-based fMRI (tb-fMRI) for clinical presurgical mapping. Nonetheless, in many clinical instances, rs-fMRI may offer valuable additional information that supplements tb-fMRI, especially if tb-fMRI is inadequate due to patient performance or other limitations. Future growth in clinical applications of rs-fMRI is anticipated as challenges are increasingly addressed. In this AJR Expert Panel Narrative Review, we summarize the current state and emerging clinical utility of rs-fMRI, focusing on its role in presurgical mapping. We present ongoing controversies and limitations in clinical applicability and discuss future directions including the developing role of rs-fMRI in neuromodulation treatment for various neurologic disorders.
Collapse
Affiliation(s)
- Behroze A Vachha
- Department of Radiology, UMass Chan Medical School, UMass Memorial Health, Worcester, Massachusetts
| | - Vinodh A Kumar
- Department of Neuroradiology, University of Texas MD Anderson Cancer Center, Hou ston, Texas
| | - Jay J Pillai
- Department of Radiology, Division of Neuroradiology, Mayo Clinic, Rochester, Minnesota
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joshua Shimony
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Jody Tanabe
- Department of Radiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Haris I Sair
- Department of Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Malone Center for Engineering in Healthcare, The Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
2
|
Miao K, Xia X, Zou Y, Shi B. Small Scale, Big Impact: Nanotechnology-Enhanced Drug Delivery for Brain Diseases. Mol Pharm 2024; 21:3777-3799. [PMID: 39038108 DOI: 10.1021/acs.molpharmaceut.4c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Central nervous system (CNS) diseases, ranging from brain cancers to neurodegenerative disorders like dementia and acute conditions such as strokes, have been heavily burdening healthcare and have a direct impact on patient quality of life. A significant hurdle in developing effective treatments is the presence of the blood-brain barrier (BBB), a highly selective barrier that prevents most drugs from reaching the brain. The tight junctions and adherens junctions between the endothelial cells and various receptors expressed on the cells make the BBB form a nonfenestrated and highly selective structure that is crucial for brain homeostasis but complicates drug delivery. Nanotechnology offers a novel pathway to circumvent this barrier, with nanoparticles engineered to ferry drugs across the BBB, protect drugs from degradation, and deliver medications to the designated area. After years of development, nanoparticle optimization, including sizes, shapes, surface modifications, and targeting ligands, can enable nanomaterials tailored to specific brain drug delivery settings. Moreover, smart nano drug delivery systems can respond to endogenous and exogenous stimuli that control subsequent drug release. Here, we address the importance of the BBB in brain disease treatment, summarize different delivery routes for brain drug delivery, discuss the cutting-edge nanotechnology-based strategies for brain drug delivery, and further offer valuable insights into how these innovations in nanoparticle technology could revolutionize the treatment of CNS diseases, presenting a promising avenue for noninvasive, targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kaiting Miao
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xue Xia
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
3
|
Lindquist BE. Spreading depolarizations pose critical energy challenges in acute brain injury. J Neurochem 2024; 168:868-887. [PMID: 37787065 PMCID: PMC10987398 DOI: 10.1111/jnc.15966] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/08/2023] [Accepted: 09/10/2023] [Indexed: 10/04/2023]
Abstract
Spreading depolarization (SD) is an electrochemical wave of neuronal depolarization mediated by extracellular K+ and glutamate, interacting with voltage-gated and ligand-gated ion channels. SD is increasingly recognized as a major cause of injury progression in stroke and brain trauma, where the mechanisms of SD-induced neuronal injury are intimately linked to energetic status and metabolic impairment. Here, I review the established working model of SD initiation and propagation. Then, I summarize the historical and recent evidence for the metabolic impact of SD, transitioning from a descriptive to a mechanistic working model of metabolic signaling and its potential to promote neuronal survival and resilience. I quantify the energetic cost of restoring ionic gradients eroded during SD, and the extent to which ion pumping impacts high-energy phosphate pools and the energy charge of affected tissue. I link energy deficits to adaptive increases in the utilization of glucose and O2, and the resulting accumulation of lactic acid and CO2 downstream of catabolic metabolic activity. Finally, I discuss the neuromodulatory and vasoactive paracrine signaling mediated by adenosine and acidosis, highlighting these metabolites' potential to protect vulnerable tissue in the context of high-frequency SD clusters.
Collapse
Affiliation(s)
- Britta E Lindquist
- Department of Neurology, University of California, San Francisco, California, USA
- Gladstone Institute of Neurological Diseases, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
| |
Collapse
|
4
|
Oue H, Hatakeyama R, Ishida E, Yokoi M, Tsuga K. Experimental tooth loss affects spatial learning function and blood-brain barrier of mice. Oral Dis 2023; 29:2907-2916. [PMID: 36114741 DOI: 10.1111/odi.14379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE This study aims to investigate how experimental tooth loss affected learning, memory function, and brain pathophysiology in mice. MATERIALS AND METHODS The mice (C57BL/6 J, 2-month-old, male) were divided into tooth loss and control groups. The behavioral test battery was performed at 6 and 12 months after tooth extraction. The protein levels of the tight junctions in the brains of the mice were analyzed. Hippocampal astrocyte was measured using immunohistochemical staining. RESULTS The results of behavioral tests and biochemical analysis performed during the 6 months observation period did not show significant differences between the groups. However, the escape latency in the tooth loss group was significantly longer than that in the control group at the 12 months after tooth extraction. The level of claudin-5 decreased in the tooth loss group. Additionally, hippocampal astrogliosis was found in the tooth loss group. CONCLUSIONS Experimental tooth loss reduced the level of claudin-5 and caused astrogliosis in the brains of mice, which was accompanied by deterioration of learning functions. This study may provide a new insight about the association between tooth loss and cognitive dysfunction.
Collapse
Affiliation(s)
- Hiroshi Oue
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rie Hatakeyama
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eri Ishida
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Miyuki Yokoi
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Tsuga
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
5
|
Agarwal S, Welker KM, Black DF, Little JT, DeLone DR, Messina SA, Passe TJ, Bettegowda C, Pillai JJ. Detection and Mitigation of Neurovascular Uncoupling in Brain Gliomas. Cancers (Basel) 2023; 15:4473. [PMID: 37760443 PMCID: PMC10527022 DOI: 10.3390/cancers15184473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Functional magnetic resonance imaging (fMRI) with blood oxygen level-dependent (BOLD) technique is useful for preoperative mapping of brain functional networks in tumor patients, providing reliable in vivo detection of eloquent cortex to help reduce the risk of postsurgical morbidity. BOLD task-based fMRI (tb-fMRI) is the most often used noninvasive method that can reliably map cortical networks, including those associated with sensorimotor, language, and visual functions. BOLD resting-state fMRI (rs-fMRI) is emerging as a promising ancillary tool for visualization of diverse functional networks. Although fMRI is a powerful tool that can be used as an adjunct for brain tumor surgery planning, it has some constraints that should be taken into consideration for proper clinical interpretation. BOLD fMRI interpretation may be limited by neurovascular uncoupling (NVU) induced by brain tumors. Cerebrovascular reactivity (CVR) mapping obtained using breath-hold methods is an effective method for evaluating NVU potential.
Collapse
Affiliation(s)
- Shruti Agarwal
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Kirk M. Welker
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - David F. Black
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - Jason T. Little
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - David R. DeLone
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - Steven A. Messina
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - Theodore J. Passe
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Jay J. Pillai
- Division of Neuroradiology, Department of Radiology, Mayo Clinic Rochester & Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.M.W.); (D.F.B.); (J.T.L.); (D.R.D.); (S.A.M.); (T.J.P.)
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| |
Collapse
|
6
|
Tewari BP, Harshad PA, Singh M, Joshi NB, Joshi PG. Pilocarpine-induced acute seizure causes rapid area-specific astrogliosis and alters purinergic signaling in rat hippocampus. Brain Res 2023:148444. [PMID: 37290610 DOI: 10.1016/j.brainres.2023.148444] [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: 03/16/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023]
Abstract
The progressive nature of acquired epilepsy warrants a thorough examination of acute changes that occur immediately after an epileptogenic insult to better understand the cellular and molecular mechanisms that trigger epileptogenesis. Astrocytes are important regulators of neuronal functions and emerging evidence suggests an involvement of astrocytic purinergic signaling in the etiology of acquired epilepsies. However, how astrocytic purinergic signaling responds immediately after an acute seizure or an epileptogenic insult to impact epileptogenesis is not well studied. In the present study, we report area-specific rapid onset of astrocytic changes in morphology, as well as in expression and functional activity of the purinergic signaling in the hippocampus that occur immediately after pilocarpine-induced stage 5 seizure. After 3 hours of stage 5 acute seizure, hippocampal astrocytes show increased intrinsic calcium activity in stratum radiatum as well as reactive astrogliosis in the stratum lacunosum moleculare and hilus regions of the hippocampus. Hilar astrocytes also upregulated the expression of P2Y1 and P2Y2 metabotropic purinergic receptors. Subsequently, P2Y1 exhibited a functional upregulation by showing a significantly higher intracellular calcium rise in ex-vivo hippocampal slices on P2Y1 activation. Our results suggest that hippocampal astrocytes undergo rapid area-specific morphological and functional changes immediately after the commencement of the seizure activity and purinergic receptors upregulation is one of the earliest changes in response to seizure activity. These changes can be considered acute astrocytic responses to seizure activity which can potentially drive the epileptogenesis and can be explored further to identify astrocyte-specific targets for seizure therapy.
Collapse
Affiliation(s)
- Bhanu P Tewari
- Department of Biophysics, National Institute of mental health and Neuroscience (NIMHANS), Hosur Road, Bangalore, 560029, Karnataka, India.
| | - P A Harshad
- Department of Biophysics, National Institute of mental health and Neuroscience (NIMHANS), Hosur Road, Bangalore, 560029, Karnataka, India
| | - Mahendra Singh
- Department of Biophysics, National Institute of mental health and Neuroscience (NIMHANS), Hosur Road, Bangalore, 560029, Karnataka, India
| | - Nanda B Joshi
- Department of Biophysics, National Institute of mental health and Neuroscience (NIMHANS), Hosur Road, Bangalore, 560029, Karnataka, India
| | - Preeti G Joshi
- Department of Biophysics, National Institute of mental health and Neuroscience (NIMHANS), Hosur Road, Bangalore, 560029, Karnataka, India.
| |
Collapse
|
7
|
Affiliation(s)
- Kıvılcım Kılıç
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Anna Devor
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
8
|
Császár E, Lénárt N, Cserép C, Környei Z, Fekete R, Pósfai B, Balázsfi D, Hangya B, Schwarcz AD, Szabadits E, Szöllősi D, Szigeti K, Máthé D, West BL, Sviatkó K, Brás AR, Mariani JC, Kliewer A, Lenkei Z, Hricisák L, Benyó Z, Baranyi M, Sperlágh B, Menyhárt Á, Farkas E, Dénes Á. Microglia modulate blood flow, neurovascular coupling, and hypoperfusion via purinergic actions. J Exp Med 2022; 219:e20211071. [PMID: 35201268 PMCID: PMC8932534 DOI: 10.1084/jem.20211071] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/28/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Microglia, the main immunocompetent cells of the brain, regulate neuronal function, but their contribution to cerebral blood flow (CBF) regulation has remained elusive. Here, we identify microglia as important modulators of CBF both under physiological conditions and during hypoperfusion. Microglia establish direct, dynamic purinergic contacts with cells in the neurovascular unit that shape CBF in both mice and humans. Surprisingly, the absence of microglia or blockade of microglial P2Y12 receptor (P2Y12R) substantially impairs neurovascular coupling in mice, which is reiterated by chemogenetically induced microglial dysfunction associated with impaired ATP sensitivity. Hypercapnia induces rapid microglial calcium changes, P2Y12R-mediated formation of perivascular phylopodia, and microglial adenosine production, while depletion of microglia reduces brain pH and impairs hypercapnia-induced vasodilation. Microglial actions modulate vascular cyclic GMP levels but are partially independent of nitric oxide. Finally, microglial dysfunction markedly impairs P2Y12R-mediated cerebrovascular adaptation to common carotid artery occlusion resulting in hypoperfusion. Thus, our data reveal a previously unrecognized role for microglia in CBF regulation, with broad implications for common neurological diseases.
Collapse
Affiliation(s)
- Eszter Császár
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zsuzsanna Környei
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Rebeka Fekete
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Diána Balázsfi
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Anett D. Schwarcz
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Szabadits
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Dávid Szöllősi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | | | - Katalin Sviatkó
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Ana Rita Brás
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Jean-Charles Mariani
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - Andrea Kliewer
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - Zsolt Lenkei
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Mária Baranyi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine, University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine, University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
9
|
Stokum JA, Shim B, Huang W, Kane M, Smith JA, Gerzanich V, Simard JM. A large portion of the astrocyte proteome is dedicated to perivascular endfeet, including critical components of the electron transport chain. J Cereb Blood Flow Metab 2021; 41:2546-2560. [PMID: 33818185 PMCID: PMC8504955 DOI: 10.1177/0271678x211004182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The perivascular astrocyte endfoot is a specialized and diffusion-limited subcellular compartment that fully ensheathes the cerebral vasculature. Despite their ubiquitous presence, a detailed understanding of endfoot physiology remains elusive, in part due to a limited understanding of the proteins that distinguish the endfoot from the greater astrocyte body. Here, we developed a technique to isolate astrocyte endfeet from brain tissue, which was used to study the endfoot proteome in comparison to the astrocyte somata. In our approach, brain microvessels, which retain their endfoot processes, were isolated from mouse brain and dissociated, whereupon endfeet were recovered using an antibody-based column astrocyte isolation kit. Our findings expand the known set of proteins enriched at the endfoot from 10 to 516, which comprised more than 1/5th of the entire detected astrocyte proteome. Numerous critical electron transport chain proteins were expressed only at the endfeet, while enzymes involved in glycogen storage were distributed to the somata, indicating subcellular metabolic compartmentalization. The endfoot proteome also included numerous proteins that, while known to have important contributions to blood-brain barrier function, were not previously known to localize to the endfoot. Our findings highlight the importance of the endfoot and suggest new routes of investigation into endfoot function.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bosung Shim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Maureen Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Jesse A Smith
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Potential of Multiscale Astrocyte Imaging for Revealing Mechanisms Underlying Neurodevelopmental Disorders. Int J Mol Sci 2021; 22:ijms221910312. [PMID: 34638653 PMCID: PMC8508625 DOI: 10.3390/ijms221910312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/18/2023] Open
Abstract
Astrocytes provide trophic and metabolic support to neurons and modulate circuit formation during development. In addition, astrocytes help maintain neuronal homeostasis through neurovascular coupling, blood-brain barrier maintenance, clearance of metabolites and nonfunctional proteins via the glymphatic system, extracellular potassium buffering, and regulation of synaptic activity. Thus, astrocyte dysfunction may contribute to a myriad of neurological disorders. Indeed, astrocyte dysfunction during development has been implicated in Rett disease, Alexander's disease, epilepsy, and autism, among other disorders. Numerous disease model mice have been established to investigate these diseases, but important preclinical findings on etiology and pathophysiology have not translated into clinical interventions. A multidisciplinary approach is required to elucidate the mechanism of these diseases because astrocyte dysfunction can result in altered neuronal connectivity, morphology, and activity. Recent progress in neuroimaging techniques has enabled noninvasive investigations of brain structure and function at multiple spatiotemporal scales, and these technologies are expected to facilitate the translation of preclinical findings to clinical studies and ultimately to clinical trials. Here, we review recent progress on astrocyte contributions to neurodevelopmental and neuropsychiatric disorders revealed using novel imaging techniques, from microscopy scale to mesoscopic scale.
Collapse
|
11
|
Yamazaki Y, Liu CC, Yamazaki A, Shue F, Martens YA, Chen Y, Qiao W, Kurti A, Oue H, Ren Y, Li Y, Aikawa T, Cherukuri Y, Fryer JD, Asmann YW, Kim BYS, Kanekiyo T, Bu G. Vascular ApoE4 Impairs Behavior by Modulating Gliovascular Function. Neuron 2021; 109:438-447.e6. [PMID: 33321072 PMCID: PMC7864888 DOI: 10.1016/j.neuron.2020.11.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 09/22/2020] [Accepted: 11/16/2020] [Indexed: 01/19/2023]
Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and multiple vascular conditions. ApoE is abundantly expressed in multiple brain cell types, including astrocytes, microglia, and vascular mural cells (VMCs). Here, we show that VMC-specific expression of apoE4 in mice impairs behavior and cerebrovascular function. Expression of either apoE3 or apoE4 in VMCs was sufficient to rescue the hypercholesterolemia and atherosclerosis phenotypes seen in Apoe knockout mice. Intriguingly, vascular expression of apoE4, but not apoE3, reduced arteriole blood flow, impaired spatial learning, and increased anxiety-like phenotypes. Single-cell RNA sequencing of vascular and glial cells revealed that apoE4 in VMCs was associated with astrocyte activation, while apoE3 was linked to angiogenic signature in pericytes. Together, our data support cell-autonomous effects of vascular apoE on brain homeostasis in an isoform-dependent manner, suggesting a critical contribution of vascular apoE to AD pathogenesis.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuanxin Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hiroshi Oue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yingxue Ren
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ying Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55902, USA
| | - Tomonori Aikawa
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yesesri Cherukuri
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - John D Fryer
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
12
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
13
|
Abstract
Neurovascular uncoupling (NVU) is one of the most important confounds of blood oxygen level dependent (BOLD) functional magnetic resonance imaging (fMR imaging) in the setting of focal brain lesions such as brain tumors. This article reviews the assessment of NVU related to focal brain lesions with emphasis on the use of cerebrovascular reactivity mapping measurement methods and resting state BOLD fMR imaging metrics in the detection of NVU, as well as the use of amplitude of low-frequency fluctuation metrics to mitigate the effects of NVU on clinical fMR imaging activation.
Collapse
Affiliation(s)
- Shruti Agarwal
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Haris I Sair
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA; The Malone Center for Engineering in Healthcare, The Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jay J Pillai
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD 21287, USA.
| |
Collapse
|
14
|
Laranjinha J, Nunes C, Ledo A, Lourenço C, Rocha B, Barbosa RM. The Peculiar Facets of Nitric Oxide as a Cellular Messenger: From Disease-Associated Signaling to the Regulation of Brain Bioenergetics and Neurovascular Coupling. Neurochem Res 2020; 46:64-76. [PMID: 32193753 DOI: 10.1007/s11064-020-03015-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
In this review, we address the regulatory and toxic role of ·NO along several pathways, from the gut to the brain. Initially, we address the role on ·NO in the regulation of mitochondrial respiration with emphasis on the possible contribution to Parkinson's disease via mechanisms that involve its interaction with a major dopamine metabolite, DOPAC. In parallel with initial discoveries of the inhibition of mitochondrial respiration by ·NO, it became clear the potential for toxic ·NO-mediated mechanisms involving the production of more reactive species and the post-translational modification of mitochondrial proteins. Accordingly, we have proposed a novel mechanism potentially leading to dopaminergic cell death, providing evidence that NO synergistically interact with DOPAC in promoting cell death via mechanisms that involve GSH depletion. The modulatory role of NO will be then briefly discussed as a master regulator on brain energy metabolism. The energy metabolism in the brain is central to the understanding of brain function and disease. The core role of ·NO in the regulation of brain metabolism and vascular responses is further substantiated by discussing its role as a mediator of neurovascular coupling, the increase in local microvessels blood flow in response to spatially restricted increase of neuronal activity. The many facets of NO as intracellular and intercellular messenger, conveying information associated with its spatial and temporal concentration dynamics, involve not only the discussion of its reactions and potential targets on a defined biological environment but also the regulation of its synthesis by the family of nitric oxide synthases. More recently, a novel pathway, out of control of NOS, has been the subject of a great deal of controversy, the nitrate:nitrite:NO pathway, adding new perspectives to ·NO biology. Thus, finally, this novel pathway will be addressed in connection with nitrate consumption in the diet and the beneficial effects of protein nitration by reactive nitrogen species.
Collapse
Affiliation(s)
- João Laranjinha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal. .,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal.
| | - Carla Nunes
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Cátia Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Bárbara Rocha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Rui M Barbosa
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| |
Collapse
|
15
|
Abstract
The blood-brain barrier (BBB) protects the vertebrate central nervous system from harmful blood-borne, endogenous and exogenous substances to ensure proper neuronal function. The BBB describes a function that is established by endothelial cells of CNS vessels in conjunction with pericytes, astrocytes, neurons and microglia, together forming the neurovascular unit (NVU). Endothelial barrier function is crucially induced and maintained by the Wnt/β-catenin pathway and requires intact NVU for proper functionality. The BBB and the NVU are characterized by a specialized assortment of molecular specializations, providing the basis for tightening, transport and immune response functionality.The present chapter introduces state-of-the-art knowledge of BBB structure and function and highlights current research topics, aiming to understanding in more depth the cellular and molecular interactions at the NVU, determining functionality of the BBB in health and disease, and providing novel potential targets for therapeutic BBB modulation. Moreover, we highlight recent advances in understanding BBB and NVU heterogeneity within the CNS as well as their contribution to CNS physiology, such as neurovascular coupling, and pathophysiology, is discussed. Finally, we give an outlook onto new avenues of BBB research.
Collapse
Affiliation(s)
- Fabienne Benz
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio Pulmonary System (CPI), Partner Site Frankfurt, Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
16
|
Wang Y, Venton BJ. Caffeine Modulates Spontaneous Adenosine and Oxygen Changes during Ischemia and Reperfusion. ACS Chem Neurosci 2019; 10:1941-1949. [PMID: 30252436 PMCID: PMC7003050 DOI: 10.1021/acschemneuro.8b00251] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Adenosine is an endogenous neuroprotectant that modulates vasodilation in the central nervous system. Oxygen changes occur when there is an increase in local cerebral blood flow and thus are a measure of vasodilation. Transient oxygen events following rapid adenosine events have been recently discovered, but the relationship between adenosine and blood flow change during ischemia/reperfusion (I/R) has not been characterized. Caffeine is a nonselective adenosine receptor antagonist that can modulate the effects of adenosine in the brain, but how it affects adenosine and oxygen levels during I/R is also unknown. In this study, extracellular changes in adenosine and oxygen were simultaneously monitored using fast-scan cyclic voltammetry during bilateral common carotid artery occlusion (BCCAO) and the effects of a specific A2A antagonist, SCH 442416, or general antagonist, caffeine, were studied. Measurements were made in the caudate-putamen for 1 h of normoxia, followed by 30 min of BCCAO and 30 min of reperfusion. The frequency and number of both adenosine and oxygen transient events significantly increased during I/R. The specific A2A antagonist, SCH 442416 (3 mg/kg, i.p.), eliminated the increase in adenosine and oxygen events caused by I/R. The general adenosine receptor antagonist, caffeine (100 mg/kg, i.p.), decreased the frequency of adenosine and oxygen transient events during I/R. These results demonstrate that, during BCCAO, there are more rapid release events of the neuromodulator adenosine and correlated local oxygen changes, and these rapid, local effects are dampened by caffeine and other A2A antagonists.
Collapse
Affiliation(s)
- Ying Wang
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| | - B Jill Venton
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| |
Collapse
|
17
|
Rotermund N, Schulz K, Hirnet D, Lohr C. Purinergic Signaling in the Vertebrate Olfactory System. Front Cell Neurosci 2019; 13:112. [PMID: 31057369 PMCID: PMC6477478 DOI: 10.3389/fncel.2019.00112] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/07/2019] [Indexed: 12/15/2022] Open
Abstract
Adenosine 5'-triphosphate (ATP) is an ubiquitous co-transmitter in the vertebrate brain. ATP itself, as well as its breakdown products ADP and adenosine are involved in synaptic transmission and plasticity, neuron-glia communication and neural development. Although purinoceptors have been demonstrated in the vertebrate olfactory system by means of histological techniques for many years, detailed insights into physiological properties and functional significance of purinergic signaling in olfaction have been published only recently. We review the current literature on purinergic neuromodulation, neuron-glia interactions and neurogenesis in the vertebrate olfactory system.
Collapse
Affiliation(s)
- Natalie Rotermund
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Kristina Schulz
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Daniela Hirnet
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
18
|
Hackett TA. Adenosine A 1 Receptor mRNA Expression by Neurons and Glia in the Auditory Forebrain. Anat Rec (Hoboken) 2018; 301:1882-1905. [PMID: 30315630 PMCID: PMC6282551 DOI: 10.1002/ar.23907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 12/30/2022]
Abstract
In the brain, purines such as ATP and adenosine can function as neurotransmitters and co‐transmitters, or serve as signals in neuron–glial interactions. In thalamocortical (TC) projections to sensory cortex, adenosine functions as a negative regulator of glutamate release via activation of the presynaptic adenosine A1 receptor (A1R). In the auditory forebrain, restriction of A1R‐adenosine signaling in medial geniculate (MG) neurons is sufficient to extend LTP, LTD, and tonotopic map plasticity in adult mice for months beyond the critical period. Interfering with adenosine signaling in primary auditory cortex (A1) does not contribute to these forms of plasticity, suggesting regional differences in the roles of A1R‐mediated adenosine signaling in the forebrain. To advance understanding of the circuitry, in situ hybridization was used to localize neuronal and glial cell types in the auditory forebrain that express A1R transcripts (Adora1), based on co‐expression with cell‐specific markers for neuronal and glial subtypes. In A1, Adora1 transcripts were concentrated in L3/4 and L6 of glutamatergic neurons. Subpopulations of GABAergic neurons, astrocytes, oligodendrocytes, and microglia expressed lower levels of Adora1. In MG, Adora1 was expressed by glutamatergic neurons in all divisions, and subpopulations of all glial classes. The collective findings imply that A1R‐mediated signaling broadly extends to all subdivisions of auditory cortex and MG. Selective expression by neuronal and glial subpopulations suggests that experimental manipulations of A1R‐adenosine signaling could impact several cell types, depending on their location. Strategies to target Adora1 in specific cell types can be developed from the data generated here. Anat Rec, 301:1882–1905, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Agarwal S, Sair HI, Pillai JJ. Limitations of Resting-State Functional MR Imaging in the Setting of Focal Brain Lesions. Neuroimaging Clin N Am 2018; 27:645-661. [PMID: 28985935 DOI: 10.1016/j.nic.2017.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Methods of image acquisition and analysis for resting-state functional MR imaging (rsfMR imaging) are still evolving. Neurovascular uncoupling and susceptibility artifact are important confounds of rsfMR imaging in the setting of focal brain lesions such as brain tumors. This article reviews the detection of these confounds using rsfMR imaging metrics in the setting of focal brain lesions. In the near future, with the wide range of ongoing research in rsfMR imaging, these issues likely will be overcome and will open new windows into brain function and connectivity.
Collapse
Affiliation(s)
- Shruti Agarwal
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Phipps B-100, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Haris I Sair
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Phipps B-100, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Jay J Pillai
- Division of Neuroradiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Phipps B-100, 1800 Orleans Street, Baltimore, MD 21287, USA.
| |
Collapse
|
20
|
Agarwal S, Lu H, Pillai JJ. Value of Frequency Domain Resting-State Functional Magnetic Resonance Imaging Metrics Amplitude of Low-Frequency Fluctuation and Fractional Amplitude of Low-Frequency Fluctuation in the Assessment of Brain Tumor-Induced Neurovascular Uncoupling. Brain Connect 2018; 7:382-389. [PMID: 28657344 DOI: 10.1089/brain.2016.0480] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of this study was to explore whether the phenomenon of brain tumor-related neurovascular uncoupling (NVU) in resting-state blood oxygen level-dependent functional magnetic resonance imaging (BOLD fMRI) (rsfMRI) may also affect the resting-state fMRI (rsfMRI) frequency domain metrics the amplitude of low-frequency fluctuation (ALFF) and fractional ALFF (fALFF). Twelve de novo brain tumor patients, who underwent clinical fMRI examinations, including task-based fMRI (tbfMRI) and rsfMRI, were included in this Institutional Review Board-approved study. Each patient displayed decreased/absent tbfMRI activation in the primary ipsilesional (IL) sensorimotor cortex in the absence of a corresponding motor deficit or suboptimal task performance, consistent with NVU. Z-score maps for the motor tasks were obtained from general linear model analysis (reflecting motor activation vs. rest). Seed-based correlation analysis (SCA) maps of sensorimotor network, ALFF, and fALFF were calculated from rsfMRI data. Precentral and postcentral gyri in contralesional (CL) and IL hemispheres were parcellated using an automated anatomical labeling template for each patient. Region of interest (ROI) analysis was performed on four maps: tbfMRI, SCA, ALFF, and fALFF. Voxel values in the CL and IL ROIs of each map were divided by the corresponding global mean of ALFF and fALFF in the cortical brain tissue. Group analysis revealed significantly decreased IL ALFF (p = 0.02) and fALFF (p = 0.03) metrics compared with CL ROIs, consistent with similar findings of significantly decreased IL BOLD signal for tbfMRI (p = 0.0005) and SCA maps (p = 0.0004). The frequency domain metrics ALFF and fALFF may be markers of lesion-induced NVU in rsfMRI similar to previously reported alterations in tbfMRI activation and SCA-derived resting-state functional connectivity maps.
Collapse
Affiliation(s)
- Shruti Agarwal
- 1 Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Hanzhang Lu
- 1 Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Jay J Pillai
- 1 Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Agarwal S, Sair HI, Pillai JJ. The Resting-State Functional Magnetic Resonance Imaging Regional Homogeneity Metrics-Kendall's Coefficient of Concordance-Regional Homogeneity and Coherence-Regional Homogeneity-Are Valid Indicators of Tumor-Related Neurovascular Uncoupling. Brain Connect 2018; 7:228-235. [PMID: 28363248 DOI: 10.1089/brain.2016.0482] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of this study is to determine whether regional homogeneity (ReHo) of resting-state blood oxygenation level-dependent (BOLD) functional magnetic resonance imaging (rsfMRI) data based on Kendall's coefficient of concordance (KCC-ReHo) and coherence (Cohe-ReHo) metrics may allow detection of brain tumor-induced neurovascular uncoupling (NVU) in the sensorimotor network similar to findings in standard motor task-based BOLD fMRI (tbfMRI) activation. Twelve de novo brain tumor patients undergoing clinical fMRI exams (tbfMRI and rsfMRI) were included in this Institutional Review Board (IRB)-approved study. Each patient displayed decreased/absent tbfMRI activation in the primary ipsilesional sensorimotor cortex in the absence of corresponding motor deficit or suboptimal task performance, consistent with NVU. Z-score maps for motor tasks were obtained from the general linear model (GLM) analysis (reflecting motor activation vs. rest). KCC-ReHo and Cohe-ReHo maps were calculated from rsfMRI data. Precentral and postcentral gyri in contralesional (CL) and ipsilesional (IL) hemispheres were parcellated using an automated anatomical labeling (AAL) template for each patient. Similar region of interest (ROI) analysis was performed on tbfMRI, KCC-ReHo, and Cohe-ReHo maps to allow direct comparison of results. Voxel values in CL and IL ROIs of each map were divided by the corresponding global mean of KCC-ReHo and Cohe-ReHo in bihemispheric cortical brain tissue. Group analysis revealed significantly decreased IL mean KCC-ReHo (p = 0.02) and Cohe-ReHo (p = 0.04) metrics compared with respective values in the CL ROIs, consistent with similar findings of significantly decreased ipsilesional BOLD signal for tbfMRI (p = 0.0005). Ipsilesional abnormalities in ReHo derived from rsfMRI may serve as potential indicators of NVU in patients with brain tumors and other resectable brain lesions; as such, ReHo findings may complement findings on tbfMRI used for presurgical planning.
Collapse
Affiliation(s)
- Shruti Agarwal
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Haris I Sair
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Jay J Pillai
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
22
|
Antipurinergic therapy for autism-An in-depth review. Mitochondrion 2017; 43:1-15. [PMID: 29253638 DOI: 10.1016/j.mito.2017.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022]
Abstract
Are the symptoms of autism caused by a treatable metabolic syndrome that traces to the abnormal persistence of a normal, alternative functional state of mitochondria? A small clinical trial published in 2017 suggests this is possible. Based on a new unifying theory of pathogenesis for autism called the cell danger response (CDR) hypothesis, this study of 10 boys, ages 5-14years, showed that all 5 boys who received antipurinergic therapy (APT) with a single intravenous dose of suramin experienced improvements in all the core symptoms of autism that lasted for 5-8weeks. Language, social interaction, restricted interests, and repetitive movements all improved. Two children who were non-verbal spoke their first sentences. None of these improvements were observed in the placebo group. Larger and longer studies are needed to confirm this promising discovery. This review introduces the concept of M2 (anti-inflammatory) and M1 (pro-inflammatory) mitochondria that are polarized along a functional continuum according to cell stress. The pathophysiology of the CDR, the complementary functions of M1 and M2 mitochondria, relevant gene-environment interactions, and the metabolic underpinnings of behavior are discussed as foundation stones for understanding the improvements in ASD behaviors produced by antipurinergic therapy in this small clinical trial.
Collapse
|
23
|
Shestopalov VI, Panchin Y, Tarasova OS, Gaynullina D, Kovalzon VM. Pannexins Are Potential New Players in the Regulation of Cerebral Homeostasis during Sleep-Wake Cycle. Front Cell Neurosci 2017; 11:210. [PMID: 28769767 PMCID: PMC5511838 DOI: 10.3389/fncel.2017.00210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
During brain homeostasis, both neurons and astroglia release ATP that is rapidly converted to adenosine in the extracellular space. Pannexin-1 (Panx1) hemichannels represent a major conduit of non-vesicular ATP release from brain cells. Previous studies have shown that Panx1−/− mice possess severe disruption of the sleep-wake cycle. Here, we review experimental data supporting the involvement of pannexins (Panx) in the coordination of fundamental sleep-associated brain processes, such as neuronal activity and regulation of cerebrovascular tone. Panx1 hemichannels are likely implicated in the regulation of the sleep-wake cycle via an indirect effect of released ATP on adenosine receptors and through interaction with other somnogens, such as IL-1β, TNFα and prostaglandin D2. In addition to the recently established role of Panx1 in the regulation of endothelium-dependent arterial dilation, similar signaling pathways are the major cellular component of neurovascular coupling. The new discovered role of Panx in sleep regulation may have broad implications in coordinating neuronal activity and homeostatic housekeeping processes during the sleep-wake cycle.
Collapse
Affiliation(s)
- Valery I Shestopalov
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of MedicineMiami, FL, United States.,Microbiology and Bioengineering Laboratory, Department of Genomics and Biotechnology, Vavilov Institute of General Genetics, Russian Academy of SciencesMoscow, Russia
| | - Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Mathematical Methods in Biology, Belozersky Institute, M.V. Lomonosov Moscow State UniversityMoscow, Russia
| | - Olga S Tarasova
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,State Research Center of the Russian Federation, Institute for Biomedical Problems, Russian Academy of SciencesMoscow, Russia
| | - Dina Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,Department of Physiology, Russian National Research Medical UniversityMoscow, Russia
| | - Vladimir M Kovalzon
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Severtsov Institute Ecology and Evolution, Russian Academy of SciencesMoscow, Russia
| |
Collapse
|
24
|
Lourenço CF, Ledo A, Barbosa RM, Laranjinha J. Neurovascular-neuroenergetic coupling axis in the brain: master regulation by nitric oxide and consequences in aging and neurodegeneration. Free Radic Biol Med 2017; 108:668-682. [PMID: 28435052 DOI: 10.1016/j.freeradbiomed.2017.04.026] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 02/21/2017] [Accepted: 04/18/2017] [Indexed: 02/08/2023]
Abstract
The strict energetic demands of the brain require that nutrient supply and usage be fine-tuned in accordance with the specific temporal and spatial patterns of ever-changing levels of neuronal activity. This is achieved by adjusting local cerebral blood flow (CBF) as a function of activity level - neurovascular coupling - and by changing how energy substrates are metabolized and shuttled amongst astrocytes and neurons - neuroenergetic coupling. Both activity-dependent increase of CBF and O2 and glucose utilization by active neural cells are inextricably linked, establishing a functional metabolic axis in the brain, the neurovascular-neuroenergetic coupling axis. This axis incorporates and links previously independent processes that need to be coordinated in the normal brain. We here review evidence supporting the role of neuronal-derived nitric oxide (•NO) as the master regulator of this axis. Nitric oxide is produced in tight association with glutamatergic activation and, diffusing several cell diameters, may interact with different molecular targets within each cell type. Hemeproteins such as soluble guanylate cyclase, cytochrome c oxidase and hemoglobin, with which •NO reacts at relatively fast rates, are but a few of the key in determinants of the regulatory role of •NO in the neurovascular-neuroenergetic coupling axis. Accordingly, critical literature supporting this concept is discussed. Moreover, in view of the controversy regarding the regulation of catabolism of different neural cells, we further discuss key aspects of the pathways through which •NO specifically up-regulates glycolysis in astrocytes, supporting lactate shuttling to neurons for oxidative breakdown. From a biomedical viewpoint, derailment of neurovascular-neuroenergetic axis is precociously linked to aberrant brain aging, cognitive impairment and neurodegeneration. Thus, we summarize current knowledge of how both neurovascular and neuroenergetic coupling are compromised in aging, traumatic brain injury, epilepsy and age-associated neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, suggesting that a shift in cellular redox balance may contribute to divert •NO bioactivity from regulation to dysfunction.
Collapse
Affiliation(s)
- Cátia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Rui M Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
25
|
Howarth C, Sutherland B, Choi HB, Martin C, Lind BL, Khennouf L, LeDue JM, Pakan JMP, Ko RWY, Ellis-Davies G, Lauritzen M, Sibson NR, Buchan AM, MacVicar BA. A Critical Role for Astrocytes in Hypercapnic Vasodilation in Brain. J Neurosci 2017; 37:2403-2414. [PMID: 28137973 PMCID: PMC5354350 DOI: 10.1523/jneurosci.0005-16.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 11/21/2016] [Accepted: 12/14/2016] [Indexed: 11/21/2022] Open
Abstract
Cerebral blood flow (CBF) is controlled by arterial blood pressure, arterial CO2, arterial O2, and brain activity and is largely constant in the awake state. Although small changes in arterial CO2 are particularly potent to change CBF (1 mmHg variation in arterial CO2 changes CBF by 3%-4%), the coupling mechanism is incompletely understood. We tested the hypothesis that astrocytic prostaglandin E2 (PgE2) plays a key role for cerebrovascular CO2 reactivity, and that preserved synthesis of glutathione is essential for the full development of this response. We combined two-photon imaging microscopy in brain slices with in vivo work in rats and C57BL/6J mice to examine the hemodynamic responses to CO2 and somatosensory stimulation before and after inhibition of astrocytic glutathione and PgE2 synthesis. We demonstrate that hypercapnia (increased CO2) evokes an increase in astrocyte [Ca2+]i and stimulates COX-1 activity. The enzyme downstream of COX-1 that synthesizes PgE2 (microsomal prostaglandin E synthase-1) depends critically for its vasodilator activity on the level of glutathione in the brain. We show that, when glutathione levels are reduced, astrocyte calcium-evoked release of PgE2 is decreased and vasodilation triggered by increased astrocyte [Ca2+]iin vitro and by hypercapnia in vivo is inhibited. Astrocyte synthetic pathways, dependent on glutathione, are involved in cerebrovascular reactivity to CO2 Reductions in glutathione levels in aging, stroke, or schizophrenia could lead to dysfunctional regulation of CBF and subsequent neuronal damage.SIGNIFICANCE STATEMENT Neuronal activity leads to the generation of CO2, which has previously been shown to evoke cerebral blood flow (CBF) increases via the release of the vasodilator PgE2 We demonstrate that hypercapnia (increased CO2) evokes increases in astrocyte calcium signaling, which in turn stimulates COX-1 activity and generates downstream PgE2 production. We demonstrate that astrocyte calcium-evoked production of the vasodilator PgE2 is critically dependent on brain levels of the antioxidant glutathione. These data suggest a novel role for astrocytes in the regulation of CO2-evoked CBF responses. Furthermore, these results suggest that depleted glutathione levels, which occur in aging and stroke, will give rise to dysfunctional CBF regulation and may result in subsequent neuronal damage.
Collapse
Affiliation(s)
- Clare Howarth
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Cancer Research United Kingdom and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
- Department of Psychology, University of Sheffield, Sheffield, S10 2TP, United Kingdom
| | - Brad Sutherland
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Hyun B Choi
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Chris Martin
- Cancer Research United Kingdom and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
- Department of Psychology, University of Sheffield, Sheffield, S10 2TP, United Kingdom
| | - Barbara Lykke Lind
- Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Lila Khennouf
- Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jeffrey M LeDue
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Janelle M P Pakan
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Rebecca W Y Ko
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Graham Ellis-Davies
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10028, and
| | - Martin Lauritzen
- Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Department of Clinical Neurophysiology, Rigshospitalet, DK-2600 Glostrup, Denmark
| | - Nicola R Sibson
- Cancer Research United Kingdom and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom,
| | - Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada,
| |
Collapse
|
26
|
Wang Y, Venton BJ. Correlation of transient adenosine release and oxygen changes in the caudate-putamen. J Neurochem 2016; 140:13-23. [PMID: 27314215 DOI: 10.1111/jnc.13705] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/19/2016] [Accepted: 06/10/2016] [Indexed: 12/01/2022]
Abstract
Adenosine is an endogenous nucleoside that modulates important physiological processes, such as vasodilation, in the central nervous system. A rapid, 2-4 s, mode of adenosine signaling has been recently discovered, but the relationship between this type of adenosine and blood flow change has not been characterized. In this study, adenosine and oxygen changes were simultaneously measured using fast-scan cyclic voltammetry. Oxygen changes occur when there is an increase in local cerebral blood flow and thus are a measure of vasodilation. About 34% of adenosine transients in the rat caudate-putamen are correlated with a subsequent transient change in oxygen. The amount of oxygen was correlated with the concentration of adenosine release and larger adenosine transients (over 0.4 μM) always had subsequent oxygen changes. The average duration of adenosine and oxygen transients was 3.2 and 3.5 s, respectively. On average, the adenosine release starts and peaks 0.2 s prior to the oxygen. The A2a antagonist, SCH442416, decreased the number of both adenosine and oxygen transient events by about 32%. However, the A1 antagonist, DPCPX, did not significantly affect simultaneous adenosine and oxygen release. The nitric oxide (NO) synthase inhibitor l-NAME also did not affect the concentration or number of adenosine and oxygen release events. These results demonstrate that both adenosine and oxygen release are modulated via A2a receptors. The correlation of transient concentrations, time delay between adenosine and oxygen peaks, and effect of A2a receptors suggests that adenosine modulates blood flow on a rapid, sub-second time scale. Read the Editorial Highlight for this article on page 10.
Collapse
Affiliation(s)
- Ying Wang
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
27
|
Vasculo-Neuronal Coupling: Retrograde Vascular Communication to Brain Neurons. J Neurosci 2016; 36:12624-12639. [PMID: 27821575 DOI: 10.1523/jneurosci.1300-16.2016] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 12/25/2022] Open
Abstract
Continuous cerebral blood flow is essential for neuronal survival, but whether vascular tone influences resting neuronal function is not known. Using a multidisciplinary approach in both rat and mice brain slices, we determined whether flow/pressure-evoked increases or decreases in parenchymal arteriole vascular tone, which result in arteriole constriction and dilation, respectively, altered resting cortical pyramidal neuron activity. We present evidence for intercellular communication in the brain involving a flow of information from vessel to astrocyte to neuron, a direction opposite to that of classic neurovascular coupling and referred to here as vasculo-neuronal coupling (VNC). Flow/pressure increases within parenchymal arterioles increased vascular tone and simultaneously decreased resting pyramidal neuron firing activity. On the other hand, flow/pressure decreases evoke parenchymal arteriole dilation and increased resting pyramidal neuron firing activity. In GLAST-CreERT2; R26-lsl-GCaMP3 mice, we demonstrate that increased parenchymal arteriole tone significantly increased intracellular calcium in perivascular astrocyte processes, the onset of astrocyte calcium changes preceded the inhibition of cortical pyramidal neuronal firing activity. During increases in parenchymal arteriole tone, the pyramidal neuron response was unaffected by blockers of nitric oxide, GABAA, glutamate, or ecto-ATPase. However, VNC was abrogated by TRPV4 channel, GABAB, as well as an adenosine A1 receptor blocker. Differently to pyramidal neuron responses, increases in flow/pressure within parenchymal arterioles increased the firing activity of a subtype of interneuron. Together, these data suggest that VNC is a complex constitutive active process that enables neurons to efficiently adjust their resting activity according to brain perfusion levels, thus safeguarding cellular homeostasis by preventing mismatches between energy supply and demand. SIGNIFICANCE STATEMENT We present evidence for vessel-to-neuron communication in the brain slice defined here as vasculo-neuronal coupling. We showed that, in response to increases in parenchymal arteriole tone, astrocyte intracellular Ca2+ increased and cortical neuronal activity decreased. On the other hand, decreasing parenchymal arteriole tone increased resting cortical pyramidal neuron activity. Vasculo-neuronal coupling was partly mediated by TRPV4 channels as genetic ablation, or pharmacological blockade impaired increased flow/pressure-evoked neuronal inhibition. Increased flow/pressure-evoked neuronal inhibition was blocked in the presence of adenosine A1 receptor and GABAB receptor blockade. Results provide evidence for the concept of vasculo-neuronal coupling and highlight the importance of understanding the interplay between basal CBF and resting neuronal activity.
Collapse
|
28
|
Abstract
Sleep and its disorders are known to affect the functions of essential organs and systems in the body. However, very little is known about how the blood-brain barrier (BBB) is regulated. A few years ago, we launched a project to determine the impact of sleep fragmentation and chronic sleep restriction on BBB functions, including permeability to fluorescent tracers, tight junction protein expression and distribution, glucose and other solute transporter activities, and mediation of cellular mechanisms. Recent publications and relevant literature allow us to summarize here the sleep-BBB interactions in five sections: (1) the structural basis enabling the BBB to serve as a huge regulatory interface; (2) BBB transport and permeation of substances participating in sleep-wake regulation; (3) the circadian rhythm of BBB function; (4) the effect of experimental sleep disruption maneuvers on BBB activities, including regional heterogeneity, possible threshold effect, and reversibility; and (5) implications of sleep disruption-induced BBB dysfunction in neurodegeneration and CNS autoimmune diseases. After reading the review, the general audience should be convinced that the BBB is an important mediating interface for sleep-wake regulation and a crucial relay station of mind-body crosstalk. The pharmaceutical industry should take into consideration that sleep disruption alters the pharmacokinetics of BBB permeation and CNS drug delivery, being attentive to the chrono timing and activation of co-transporters in subjects with sleep disorders.
Collapse
Affiliation(s)
- Weihong Pan
- 1 Biopotentials Sleep Center, Baton Rouge, LA 70809
| | - Abba J Kastin
- 2 Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
29
|
Busija DW, Rutkai I, Dutta S, Katakam PV. Role of Mitochondria in Cerebral Vascular Function: Energy Production, Cellular Protection, and Regulation of Vascular Tone. Compr Physiol 2016; 6:1529-48. [PMID: 27347901 DOI: 10.1002/cphy.c150051] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria not only produce energy in the form of ATP to support the activities of cells comprising the neurovascular unit, but mitochondrial events, such as depolarization and/or ROS release, also initiate signaling events which protect the endothelium and neurons against lethal stresses via pre-/postconditioning as well as promote changes in cerebral vascular tone. Mitochondrial depolarization in vascular smooth muscle (VSM), via pharmacological activation of the ATP-dependent potassium channels on the inner mitochondrial membrane (mitoKATP channels), leads to vasorelaxation through generation of calcium sparks by the sarcoplasmic reticulum and subsequent downstream signaling mechanisms. Increased release of ROS by mitochondria has similar effects. Relaxation of VSM can also be indirectly achieved via actions of nitric oxide (NO) and other vasoactive agents produced by endothelium, perivascular and parenchymal nerves, and astroglia following mitochondrial activation. Additionally, NO production following mitochondrial activation is involved in neuronal preconditioning. Cerebral arteries from female rats have greater mitochondrial mass and respiration and enhanced cerebral arterial dilation to mitochondrial activators. Preexisting chronic conditions such as insulin resistance and/or diabetes impair mitoKATP channel relaxation of cerebral arteries and preconditioning. Surprisingly, mitoKATP channel function after transient ischemia appears to be retained in the endothelium of large cerebral arteries despite generalized cerebral vascular dysfunction. Thus, mitochondrial mechanisms may represent the elusive signaling link between metabolic rate and blood flow as well as mediators of vascular change according to physiological status. Mitochondrial mechanisms are an important, but underutilized target for improving vascular function and decreasing brain injury in stroke patients. © 2016 American Physiological Society. Compr Physiol 6:1529-1548, 2016.
Collapse
Affiliation(s)
- David W Busija
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Ibolya Rutkai
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Somhrita Dutta
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Prasad V Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
30
|
Toth P, Tarantini S, Davila A, Valcarcel-Ares MN, Tucsek Z, Varamini B, Ballabh P, Sonntag WE, Baur JA, Csiszar A, Ungvari Z. Purinergic glio-endothelial coupling during neuronal activity: role of P2Y1 receptors and eNOS in functional hyperemia in the mouse somatosensory cortex. Am J Physiol Heart Circ Physiol 2015; 309:H1837-45. [PMID: 26453330 DOI: 10.1152/ajpheart.00463.2015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Abstract
Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) via neurovascular coupling is thought to play a critical role in the genesis of cognitive impairment associated with aging and pathological conditions associated with accelerated cerebromicrovascular aging (e.g., hypertension, obesity). Although previous studies demonstrate that endothelial dysfunction plays a critical role in neurovascular uncoupling in these conditions, the role of endothelial NO mediation in neurovascular coupling responses is not well understood. To establish the link between endothelial function and functional hyperemia, neurovascular coupling responses were studied in mutant mice overexpressing or deficient in endothelial NO synthase (eNOS), and the role of P2Y1 receptors in purinergic glioendothelial coupling was assessed. We found that genetic depletion of eNOS (eNOS(-/-)) and pharmacological inhibition of NO synthesis significantly decreased the CBF responses in the somatosensory cortex evoked by whisker stimulation and by administration of ATP. Overexpression of eNOS enhanced NO mediation of functional hyperemia. In control mice, the selective and potent P2Y1 receptor antagonist MRS2179 attenuated both whisker stimulation-induced and ATP-mediated CBF responses, whereas, in eNOS(-/-) mice, the inhibitory effects of MRS2179 were blunted. Collectively, our findings provide additional evidence for purinergic glio-endothelial coupling during neuronal activity, highlighting the role of ATP-mediated activation of eNOS via P2Y1 receptors in functional hyperemia.
Collapse
Affiliation(s)
- Peter Toth
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Neurosurgery, University of Pecs, Pecs, Hungary; Szentagothai Research Center, Medical School, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Antonio Davila
- Institute for Diabetes, Obesity, and Metabolism and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - M Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Zsuzsanna Tucsek
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Szentagothai Research Center, Medical School, University of Pecs, Pecs, Hungary
| | - Behzad Varamini
- Biological Sciences Department, Biola University, La Mirada, California
| | - Praveen Ballabh
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York; Department of Pediatrics, Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, New York
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Joseph A Baur
- Institute for Diabetes, Obesity, and Metabolism and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Szentagothai Research Center, Medical School, University of Pecs, Pecs, Hungary; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Szentagothai Research Center, Medical School, University of Pecs, Pecs, Hungary; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
31
|
Agarwal S, Sair HI, Yahyavi-Firouz-Abadi N, Airan R, Pillai JJ. Neurovascular uncoupling in resting state fMRI demonstrated in patients with primary brain gliomas. J Magn Reson Imaging 2015. [PMID: 26201672 DOI: 10.1002/jmri.25012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND To demonstrate that the problem of brain tumor-related neurovascular uncoupling (NVU) is a significant issue with respect to resting state blood oxygen level dependent (BOLD) functional MRI (rsfMRI) similar to task-based BOLD fMRI, in which signal detectability can be compromised by breakdown of normal neurovascular coupling. METHODS We evaluated seven de novo brain tumor patients who underwent resting state fMRI as part of comprehensive clinical fMRI exams at 3 Tesla. For each of the seven patients who demonstrated evidence of NVU on task-based motor fMRI, we performed both an independent component analysis (ICA) and an atlas-based parcellation-based seed correlation analysis (SCA) of the resting state fMRI data. For each patient, ipsilesional (IL) and contralesional (CL) regions of interest (ROIs) comprising primary motor and somatosensory cortices were used to evaluate BOLD signal changes on Z score maps derived from both ICA and SCA analysis for evidence of NVU. A subsequent two-tailed t-test was performed to determine whether statistically significant differences between the two sides were present that were consistent with NVU. RESULTS In seven patients, overall decreased BOLD signal (based on suprathreshold voxels in ICA and SCA-derived Z-score maps) was noted in IL compared with CL ROIs (P < 0.01), consistent with NVU. CONCLUSION We have demonstrated that NVU can result in false negative BOLD signal changes on rsfMRI comparable to previously published findings on standard motor task-based fMRI.
Collapse
Affiliation(s)
- Shruti Agarwal
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haris I Sair
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Noushin Yahyavi-Firouz-Abadi
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raag Airan
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jay J Pillai
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Muñoz MF, Puebla M, Figueroa XF. Control of the neurovascular coupling by nitric oxide-dependent regulation of astrocytic Ca(2+) signaling. Front Cell Neurosci 2015; 9:59. [PMID: 25805969 PMCID: PMC4354411 DOI: 10.3389/fncel.2015.00059] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/07/2015] [Indexed: 12/28/2022] Open
Abstract
Neuronal activity must be tightly coordinated with blood flow to keep proper brain function, which is achieved by a mechanism known as neurovascular coupling. Then, an increase in synaptic activity leads to a dilation of local parenchymal arterioles that matches the enhanced metabolic demand. Neurovascular coupling is orchestrated by astrocytes. These glial cells are located between neurons and the microvasculature, with the astrocytic endfeet ensheathing the vessels, which allows fine intercellular communication. The neurotransmitters released during neuronal activity reach astrocytic receptors and trigger a Ca2+ signaling that propagates to the endfeet, activating the release of vasoactive factors and arteriolar dilation. The astrocyte Ca2+ signaling is coordinated by gap junction channels and hemichannels formed by connexins (Cx43 and Cx30) and channels formed by pannexins (Panx-1). The neuronal activity-initiated Ca2+ waves are propagated among neighboring astrocytes directly via gap junctions or through ATP release via connexin hemichannels or pannexin channels. In addition, Ca2+ entry via connexin hemichannels or pannexin channels may participate in the regulation of the astrocyte signaling-mediated neurovascular coupling. Interestingly, nitric oxide (NO) can activate connexin hemichannel by S-nitrosylation and the Ca2+-dependent NO-synthesizing enzymes endothelial NO synthase (eNOS) and neuronal NOS (nNOS) are expressed in astrocytes. Therefore, the astrocytic Ca2+ signaling triggered in neurovascular coupling may activate NO production, which, in turn, may lead to Ca2+ influx through hemichannel activation. Furthermore, NO release from the hemichannels located at astrocytic endfeet may contribute to the vasodilation of parenchymal arterioles. In this review, we discuss the mechanisms involved in the regulation of the astrocytic Ca2+ signaling that mediates neurovascular coupling, with a special emphasis in the possible participation of NO in this process.
Collapse
Affiliation(s)
- Manuel F Muñoz
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mariela Puebla
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Xavier F Figueroa
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
33
|
Cheung G, Chever O, Rouach N. Connexons and pannexons: newcomers in neurophysiology. Front Cell Neurosci 2014; 8:348. [PMID: 25408635 PMCID: PMC4219455 DOI: 10.3389/fncel.2014.00348] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/06/2014] [Indexed: 11/14/2022] Open
Abstract
Connexin hemichannels are single membrane channels which have been traditionally thought to work in pairs to form gap junction channels across two opposing cells. In astrocytes, gap junction channels allow direct intercellular communication and greatly facilitate the transmission of signals. Recently, there has been growing evidence demonstrating that connexin hemichannels, as well as pannexin channels, on their own are open in various conditions. They allow bidirectional flow of ions and signaling molecules and act as release sites for transmitters like ATP and glutamate into the extracellular space. While much attention has focused on the function of connexin hemichannels and pannexons during pathological situations like epilepsy, inflammation, neurodegeneration or ischemia, their potential roles in physiology is often ignored. In order to fully understand the dynamic properties and roles of connexin hemichannels and pannexons in the brain, it is essential to decipher whether they also have some physiological functions and contribute to normal cerebral processes. Here, we present recent studies in the CNS suggesting emerging physiological functions of connexin hemichannels and pannexons in normal neuronal activity and behavior. We also discuss how these pioneer studies pave the way for future research to extend the physiological relevance of connexons and pannexons, and some fundamental issues yet to be addressed.
Collapse
Affiliation(s)
- Giselle Cheung
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| |
Collapse
|
34
|
Pathological potential of astroglial purinergic receptors. ADVANCES IN NEUROBIOLOGY 2014; 11:213-56. [PMID: 25236731 DOI: 10.1007/978-3-319-08894-5_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute brain injury and neurodegenerative disorders may result in astroglial activation. Astrocytes are able to determine the progression and outcome of these neuropathologies in a beneficial or detrimental way. Nucleotides, e.g. adenosine 5'-triphosphate (ATP), released after acute or chronic neuronal injury, are important mediators of glial activation and astrogliosis.Acute injury may cause significant changes in ATP balance, resulting in (1) a decline of intracellular ATP levels and (2) an increase in extracellular ATP concentrations via efflux from the intracellular space. The released ATP may have trophic effects, but can also act as a proinflammatory mediator or cytotoxic factor, inducing necrosis/apoptosis as a universal "danger" signal. Furthermore, ATP, primarily released from astrocytes, is a means of communication between neurons, glial cells, and intracerebral blood vessels.Astrocytes express a heterogeneous battery of purinergic ionotropic and metabotropic receptors (P2XRs and P2YRs, respectively) to respond to extracellular nucleotides.In this chapter, we summarize the contemporary knowledge on the pathological potential of P2Rs in relation to changes of astrocytic functions, determined by distinct molecular signaling cascades, in a variety of diseases. We discuss specific aspects of reactive astrogliosis, with respect to the involvement of prominent receptor subtypes, such as the P2X7 and P2Y1/2Rs. Examples of purinergic signaling of microglia, oligodendrocytes, and blood vessels under pathophysiological conditions will also be presented.The understanding of the pathological potential of purinergic signaling in "controlling and fine-tuning" of astrocytic responses is important for identifying possible therapeutic principles to treat acute and chronic central nervous system diseases.
Collapse
|
35
|
Busija DW, Katakam PV. Mitochondrial mechanisms in cerebral vascular control: shared signaling pathways with preconditioning. J Vasc Res 2014; 51:175-89. [PMID: 24862206 PMCID: PMC4149841 DOI: 10.1159/000360765] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 02/19/2014] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial-initiated events protect the neurovascular unit against lethal stress via a process called preconditioning, which independently promotes changes in cerebrovascular tone through shared signaling pathways. Activation of adenosine triphosphate (ATP)-dependent potassium channels on the inner mitochondrial membrane (mitoKATP channels) is a specific and dependable way to induce protection of neurons, astroglia, and cerebral vascular endothelium. Through the opening of mitoKATP channels, mitochondrial depolarization leads to activation of protein kinases and transient increases in cytosolic calcium (Ca(2+)) levels that activate terminal mechanisms that protect the neurovascular unit against lethal stress. The release of reactive oxygen species from mitochondria has similar protective effects. Signaling elements of the preconditioning pathways also are involved in the regulation of vascular tone. Activation of mitoKATP channels in cerebral arteries causes vasodilation, with cell-specific contributions from the endothelium, vascular smooth muscles, and nerves. Preexisting chronic conditions, such as insulin resistance and/or diabetes, prevent preconditioning and impair relaxation to mitochondrial-centered responses in cerebral arteries. Surprisingly, mitochondrial activation after anoxic or ischemic stress appears to protect cerebral vascular endothelium and promotes the restoration of blood flow; therefore, mitochondria may represent an important, but underutilized target in attenuating vascular dysfunction and brain injury in stroke patients.
Collapse
Affiliation(s)
- David W Busija
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, La., USA
| | | |
Collapse
|
36
|
Muoio V, Persson PB, Sendeski MM. The neurovascular unit - concept review. Acta Physiol (Oxf) 2014; 210:790-8. [PMID: 24629161 DOI: 10.1111/apha.12250] [Citation(s) in RCA: 364] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/01/2013] [Accepted: 01/27/2014] [Indexed: 01/01/2023]
Abstract
The cerebral hyperaemia is one of the fundamental mechanisms for the central nervous system homeostasis. Due also to this mechanism, oxygen and nutrients are maintained in satisfactory levels, through vasodilation and vasoconstriction. The brain hyperaemia, or coupling, is accomplished by a group of cells, closely related to each other; called neurovascular unit (NVU). The neurovascular unit is composed by neurones, astrocytes, endothelial cells of blood-brain barrier (BBB), myocytes, pericytes and extracellular matrix components. These cells, through their intimate anatomical and chemical relationship, detect the needs of neuronal supply and trigger necessary responses (vasodilation or vasoconstriction) for such demands. Here, we review the concepts of NVU, the coupling mechanisms and research strategies.
Collapse
Affiliation(s)
- V. Muoio
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| | - P. B. Persson
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| | - M. M. Sendeski
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| |
Collapse
|
37
|
Papa M, De Luca C, Petta F, Alberghina L, Cirillo G. Astrocyte-neuron interplay in maladaptive plasticity. Neurosci Biobehav Rev 2014; 42:35-54. [PMID: 24509064 DOI: 10.1016/j.neubiorev.2014.01.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/03/2014] [Accepted: 01/28/2014] [Indexed: 12/21/2022]
Abstract
The complexity of neuronal networks cannot only be explained by neuronal activity so neurobiological research in the last decade has focused on different components of the central nervous system: the glia. Glial cells are fundamental elements for development and maintenance of physiological brain work. New data confirm that glia significantly influences neuronal communication through specific molecules, named "gliotransmitters", and their related receptors. This new approach to the traditional model of the way synapses work is also supported by changes occurring in pathological conditions, such as neurodegenerative diseases or toxic/traumatic injury to nervous system. Experimental models have revealed that glial cells are the starting point of damage progression that subsequently involves neurons. The "bedside to bench" approach has demonstrated that clinical phenotypes are strictly related to neuronal death, however it is conceivable that the disease begins earlier, years before clinical onset. This temporal gap is necessary to determine complex changes in the neuro-glial network organization and produce a "maladaptive plasticity". We review the function of glial cells in health and disease, pointing the putative mechanisms of maladaptive plasticity, suggesting that glial cells may represent a fascinating therapeutic target to prevent irreversible neuronal cell death.
Collapse
Affiliation(s)
- Michele Papa
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, Second University of Naples, 80138 Naples, Italy; SYSBIO, Centre of Systems Biology, University of Milano-Bicocca, Milano, Italy.
| | - Ciro De Luca
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, Second University of Naples, 80138 Naples, Italy
| | - Federica Petta
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, Second University of Naples, 80138 Naples, Italy
| | - Lilia Alberghina
- Laboratory of Neuroscience "R. Levi-Montalcini", Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy; SYSBIO, Centre of Systems Biology, University of Milano-Bicocca, Milano, Italy
| | - Giovanni Cirillo
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, Second University of Naples, 80138 Naples, Italy
| |
Collapse
|
38
|
Chaitanya GV, Minagar A, Alexander JS. Neuronal and astrocytic interactions modulate brain endothelial properties during metabolic stresses of in vitro cerebral ischemia. Cell Commun Signal 2014; 12:7. [PMID: 24438487 PMCID: PMC3927849 DOI: 10.1186/1478-811x-12-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/25/2013] [Indexed: 01/25/2023] Open
Abstract
Neurovascular and gliovascular interactions significantly affect endothelial phenotype. Physiologically, brain endothelium attains several of its properties by its intimate association with neurons and astrocytes. However, during cerebrovascular pathologies such as cerebral ischemia, the uncoupling of neurovascular and gliovascular units can result in several phenotypical changes in brain endothelium. The role of neurovascular and gliovascular uncoupling in modulating brain endothelial properties during cerebral ischemia is not clear. Specifically, the roles of metabolic stresses involved in cerebral ischemia, including aglycemia, hypoxia and combined aglycemia and hypoxia (oxygen glucose deprivation and re-oxygenation, OGDR) in modulating neurovascular and gliovascular interactions are not known. The complex intimate interactions in neurovascular and gliovascular units are highly difficult to recapitulate in vitro. However, in the present study, we used a 3D co-culture model of brain endothelium with neurons and astrocytes in vitro reflecting an intimate neurovascular and gliovascular interactions in vivo. While the cellular signaling interactions in neurovascular and gliovascular units in vivo are much more complex than the 3D co-culture models in vitro, we were still able to observe several important phenotypical changes in brain endothelial properties by metabolically stressed neurons and astrocytes including changes in barrier, lymphocyte adhesive properties, endothelial cell adhesion molecule expression and in vitro angiogenic potential.
Collapse
Affiliation(s)
| | | | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health-Shreveport, Louisiana 71103, USA.
| |
Collapse
|
39
|
Fredholm BB. Adenosine--a physiological or pathophysiological agent? J Mol Med (Berl) 2013; 92:201-6. [PMID: 24362516 DOI: 10.1007/s00109-013-1101-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/26/2013] [Accepted: 04/29/2013] [Indexed: 01/25/2023]
Abstract
This minireview briefly summarizes the evidence that adenosine, acting on four G-protein coupled receptors, can play physiological roles, but is also critically involved in pathological processes. The factors that decide which of these is the more important in a specific cell or organ are briefly summarized. The fact that drugs that target adenosine receptors in disease will also hit the physiological processes will make drug development more tricky.
Collapse
Affiliation(s)
- Bertil B Fredholm
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177, Stockholm, Sweden,
| |
Collapse
|
40
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
41
|
Shen HY, Sun H, Hanthorn MM, Zhi Z, Lan JQ, Poulsen DJ, Wang RK, Boison D. Overexpression of adenosine kinase in cortical astrocytes and focal neocortical epilepsy in mice. J Neurosurg 2013; 120:628-38. [PMID: 24266544 DOI: 10.3171/2013.10.jns13918] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECT New experimental models and diagnostic methods are needed to better understand the pathophysiology of focal neocortical epilepsies in a search for improved epilepsy treatment options. The authors hypothesized that a focal disruption of adenosine homeostasis in the neocortex might be sufficient to trigger electrographic seizures. They further hypothesized that a focal disruption of adenosine homeostasis might affect microcirculation and thus offer a diagnostic opportunity for the detection of a seizure focus located in the neocortex. METHODS Focal disruption of adenosine homeostasis was achieved by injecting an adeno-associated virus (AAV) engineered to overexpress adenosine kinase (ADK), the major metabolic clearance enzyme for the brain's endogenous anticonvulsant adenosine, into the neocortex of mice. Eight weeks following virus injection, the affected brain area was imaged via optical microangiography (OMAG) to detect changes in microcirculation. After completion of imaging, cortical electroencephalography (EEG) recordings were obtained from the imaged brain area. RESULTS Viral expression of the Adk cDNA in astrocytes generated a focal area (~ 2 mm in diameter) of ADK overexpression within the neocortex. OMAG scanning revealed a reduction in vessel density within the affected brain area of approximately 23% and 29% compared with control animals and the contralateral hemisphere, respectively. EEG recordings revealed electrographic seizures within the focal area of ADK overexpression at a rate of 1.3 ± 0.2 seizures per hour (mean ± SEM). CONCLUSIONS The findings of this study suggest that focal adenosine deficiency is sufficient to generate a neocortical focus of hyperexcitability, which is also characterized by reduced vessel density. The authors conclude that their model constitutes a useful tool to study neocortical epilepsies and that OMAG constitutes a noninvasive diagnostic tool for the imaging of seizure foci with disrupted adenosine homeostasis.
Collapse
Affiliation(s)
- Hai-Ying Shen
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, Oregon
| | | | | | | | | | | | | | | |
Collapse
|
42
|
zur Nedden S, Doney AS, Frenguelli BG. Modulation of intracellular ATP determines adenosine release and functional outcome in response to metabolic stress in rat hippocampal slices and cerebellar granule cells. J Neurochem 2013; 128:111-24. [PMID: 23937448 DOI: 10.1111/jnc.12397] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/30/2013] [Accepted: 08/06/2013] [Indexed: 11/24/2022]
Abstract
Cerebral ischaemia rapidly depletes cellular ATP. Whilst this deprives brain tissue of a valuable energy source, the concomitant production of adenosine mitigates the damaging effects of energy failure by suppressing neuronal activity. However, the production of adenosine and other metabolites, and their loss across the blood-brain barrier, deprives the brain of substrates for the purine salvage pathway, the primary means by which the brain makes ATP. Because of this, cerebral ATP levels remain depressed after brain injury. To test whether manipulating cellular ATP levels in brain tissue could affect functional neuronal outcomes in response to oxygen/glucose deprivation (OGD), we examined the effects of creatine and d-ribose and adenine (RibAde). In hippocampal slices creatine delayed ATP breakdown, reduced adenosine release, retarded both the depression of synaptic transmission and the anoxic depolarization caused by OGD, and improved the recovery of transmission. In contrast, RibAde increased cellular ATP, caused increased OGD-induced adenosine release and accelerated the depression of synaptic transmission, but did not improve functional recovery. However, RibAde improved the viability of cerebellar granule cells when administered after OGD. Our data indicate that RibAde may be effective in promoting recovery of brain tissue after injury, potentially via enhancement of salvage-mediated ATP production.
Collapse
|
43
|
Boison D. Glowing feet control the blood of seizures. Epilepsy Curr 2013; 13:122-3. [PMID: 23840169 PMCID: PMC3697877 DOI: 10.5698/1535-7511-13.3.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2024] Open
|
44
|
Moreno A, Jego P, de la Cruz F, Canals S. Neurophysiological, metabolic and cellular compartments that drive neurovascular coupling and neuroimaging signals. FRONTIERS IN NEUROENERGETICS 2013; 5:3. [PMID: 23543907 PMCID: PMC3610078 DOI: 10.3389/fnene.2013.00003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/13/2013] [Indexed: 11/22/2022]
Abstract
Complete understanding of the mechanisms that coordinate work and energy supply of the brain, the so called neurovascular coupling, is fundamental to interpreting brain energetics and their influence on neuronal coding strategies, but also to interpreting signals obtained from brain imaging techniques such as functional magnetic resonance imaging. Interactions between neuronal activity and cerebral blood flow regulation are largely compartmentalized. First, there exists a functional compartmentalization in which glutamatergic peri-synaptic activity and its electrophysiological events occur in close proximity to vascular responses. Second, the metabolic processes that fuel peri-synaptic activity are partially segregated between glycolytic and oxidative compartments. Finally, there is cellular segregation between astrocytic and neuronal compartments, which has potentially important implications on neurovascular coupling. Experimental data is progressively showing a tight interaction between the products of energy consumption and neurotransmission-driven signaling molecules that regulate blood flow. Here, we review some of these issues in light of recent findings with special attention to the neuron-glia interplay on the generation of neuroimaging signals.
Collapse
Affiliation(s)
- Andrea Moreno
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezSan Juan de Alicante, Spain
| | - Pierrick Jego
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezSan Juan de Alicante, Spain
| | - Feliberto de la Cruz
- Centro de Estudios Avanzados de Cuba, Ministerio de Ciencia Tecnología y Medio AmbienteHabana, Cuba
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezSan Juan de Alicante, Spain
| |
Collapse
|
45
|
The astrocytic contribution to neurovascular coupling – Still more questions than answers? Neurosci Res 2013; 75:171-83. [DOI: 10.1016/j.neures.2013.01.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/15/2012] [Accepted: 12/30/2012] [Indexed: 01/03/2023]
|
46
|
Ronaldson PT, Davis TP. Targeted drug delivery to treat pain and cerebral hypoxia. Pharmacol Rev 2013; 65:291-314. [PMID: 23343976 DOI: 10.1124/pr.112.005991] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Limited drug penetration is an obstacle that is often encountered in treatment of central nervous system (CNS) diseases including pain and cerebral hypoxia. Over the past several years, biochemical characteristics of the brain (i.e., tight junction protein complexes at brain barrier sites, expression of influx and efflux transporters) have been shown to be directly involved in determining CNS permeation of therapeutic agents; however, the vast majority of these studies have focused on understanding those mechanisms that prevent drugs from entering the CNS. Recently, this paradigm has shifted toward identifying and characterizing brain targets that facilitate CNS drug delivery. Such targets include the organic anion-transporting polypeptides (OATPs in humans; Oatps in rodents), a family of sodium-independent transporters that are endogenously expressed in the brain and are involved in drug uptake. OATP/Oatp substrates include drugs that are efficacious in treatment of pain and/or cerebral hypoxia (i.e., opioid analgesic peptides, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors). This clearly suggests that OATP/Oatp isoforms are viable transporter targets that can be exploited for optimization of drug delivery to the brain and, therefore, improved treatment of CNS diseases. This review summarizes recent knowledge in this area and emphasizes the potential that therapeutic targeting of OATP/Oatp isoforms may have in facilitating CNS drug delivery and distribution. Additionally, information presented in this review will point to novel strategies that can be used for treatment of pain and cerebral hypoxia.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Medical Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Avenue, P.O. Box 245050, Tucson, AZ 85724-5050, USA.
| | | |
Collapse
|
47
|
Neural progenitor cells regulate capillary blood flow in the postnatal subventricular zone. J Neurosci 2013; 32:16435-48. [PMID: 23152626 DOI: 10.1523/jneurosci.1457-12.2012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the postnatal subventricular zone (SVZ), S phase entry of neural progenitor cells (NPCs) correlates with a local increase in blood flow. However, the cellular mechanism controlling this hemodynamic response remains unknown. We show that a subpopulation of SVZ cells, astrocyte-like cells or B-cells, sends projections ensheathing pericytes on SVZ capillaries in young mice. We examined whether calcium increases in pericytes or B-cells led to a vascular response in acute slices using the P2Y(2/4) receptor (P2Y(2/4)R) agonist UTP, electrical stimulation, or transgenic mice expressing exogenous Gq-coupled receptors (MrgA1) in B-cells. UTP increased calcium in pericytes leading to capillary constrictions. Electrical stimulation induced calcium propagation in SVZ cells followed by capillary constrictions involving purinergic receptors. In transgenic mice, selective calcium increases in B-cells induced P2Y(2/4)R-dependent capillary constrictions, suggesting that B-cells release ATP activating purinergic receptors on pericytes. Interestingly, in the presence of a P2Y(2/4)R blocker, dilation was observed. Intraventricular UTP injection transiently decreased blood flow monitored in vivo using laser Doppler flowmetry. Using neonatal electroporation, we expressed MrgA1 in slow cycling radial glia-derived B1 cells, i.e., NPCs. Intraventricular injection of an MrgA1 ligand increased blood flow in the SVZ. Thus, upon intracellular calcium increases B-cells/NPCs release ATP and vasodilating factors that activate purinergic receptors on pericytes triggering a vascular response and blood flow increase in vivo. Considering that NPCs receive signals from other SVZ cells, these findings further suggest that NPCs act as transducers of neurometabolic coupling in the SVZ.
Collapse
|
48
|
Central α-adrenoceptors contribute to mustard oil-induced central sensitization in the rat medullary dorsal horn. Neuroscience 2013; 236:244-52. [PMID: 23333675 DOI: 10.1016/j.neuroscience.2013.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/22/2012] [Accepted: 01/03/2013] [Indexed: 11/22/2022]
Abstract
Our previous studies have demonstrated that application of the inflammatory irritant mustard oil (MO) to the tooth pulp produces trigeminal central sensitization that includes increases in mechanoreceptive field size and responses to noxious stimuli and decrease in activation threshold in brainstem nociceptive neurons of trigeminal subnucleus caudalis (the medullary dorsal horn, MDH). The aim of the present study was to test if central noradrenergic processes are involved in the central sensitization of MDH neurons and if α1-adrenoceptors or α2-adrenoceptors or both are involved. In urethane/α-chloralose-anesthetized rats, the activity of extracellularly recorded and functionally identified single nociceptive neurons in the MDH was studied. Continuous intrathecal (i.t.) superfusion of the adrenergic modulator guanethidine and α-adrenoceptor blocker phentolamine or selective α1-adrenoceptor antagonist prazosin over the medulla strongly attenuated all three MO-induced parameters of central sensitization in the MDH nociceptive neurons, compared to phosphate-buffered saline (as vehicle control). In contrast, i.t. superfusion of the selective α2-adrenoceptor antagonist yohimbine had little effect on the mechanoreceptive field expansion and the decreased mechanical activation threshold, and indeed facilitated responses to noxious stimuli of sensitized nociceptive neurons. Superfusion of each of the four chemicals alone did not affect baseline nociceptive neuronal properties. These findings provide the first documentation of the involvement of central noradrenergic processes in MDH in the development of the central sensitization, and that α1- and α2-adrenoceptors may be differentially involved.
Collapse
|
49
|
Ronaldson PT, Davis TP. Blood-brain barrier integrity and glial support: mechanisms that can be targeted for novel therapeutic approaches in stroke. Curr Pharm Des 2012; 18:3624-44. [PMID: 22574987 DOI: 10.2174/138161212802002625] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/06/2012] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is a critical regulator of brain homeostasis. Additionally, the BBB is the most significant obstacle to effective CNS drug delivery. It possesses specific charcteristics (i.e., tight junction protein complexes, influx and efflux transporters) that control permeation of circulating solutes including therapeutic agents. In order to form this "barrier," brain microvascular endothelial cells require support of adjacent astrocytes and microglia. This intricate relationship also occurs between endothelial cells and other cell types and structures of the CNS (i.e., pericytes, neurons, extracellular matrix), which implies existence of a "neurovascular unit." Ischemic stroke can disrupt the neurovascular unit at both the structural and functional level, which leads to an increase in leak across the BBB. Recent studies have identified several pathophysiological mechanisms (i.e., oxidative stress, activation of cytokine-mediated intracellular signaling systems) that mediate changes in the neurovascular unit during ischemic stroke. This review summarizes current knowledge in this area and emphasizes pathways (i.e., oxidative stress, cytokine-mediated intracellular signaling, glial-expressed receptors/targets) that can be manipulated pharmacologically for i) preservation of BBB and glial integrity during ischemic stroke and ii) control of drug permeation and/or transport across the BBB. Targeting these pathways present a novel opportunity for optimization of CNS delivery of therapeutics in the setting of ischemic stroke.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Medical Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Avenue, P.O. Box 245050, Tucson, AZ 85724-5050, USA.
| | | |
Collapse
|
50
|
Franke H, Verkhratsky A, Burnstock G, Illes P. Pathophysiology of astroglial purinergic signalling. Purinergic Signal 2012; 8:629-57. [PMID: 22544529 DOI: 10.1007/s11302-012-9300-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/01/2012] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are fundamental for central nervous system (CNS) physiology and are the fulcrum of neurological diseases. Astroglial cells control development of the nervous system, regulate synaptogenesis, maturation, maintenance and plasticity of synapses and are central for nervous system homeostasis. Astroglial reactions determine progression and outcome of many neuropathologies and are critical for regeneration and remodelling of neural circuits following trauma, stroke, ischaemia or neurodegenerative disorders. They secrete multiple neurotransmitters and neurohormones to communicate with neurones, microglia and the vascular walls of capillaries. Signalling through release of ATP is the most widespread mean of communication between astrocytes and other types of neural cells. ATP serves as a fast excitatory neurotransmitter and has pronounced long-term (trophic) roles in cell proliferation, growth, and development. During pathology, ATP is released from damaged cells and acts both as a cytotoxic factor and a proinflammatory mediator, being a universal "danger" signal. In this review, we summarise contemporary knowledge on the role of purinergic receptors (P2Rs) in a variety of diseases in relation to changes of astrocytic functions and nucleotide signalling. We have focussed on the role of the ionotropic P2X and metabotropic P2YRs working alone or in concert to modify the release of neurotransmitters, to activate signalling cascades and to change the expression levels of ion channels and protein kinases. All these effects are of great importance for the initiation, progression and maintenance of astrogliosis-the conserved and ubiquitous glial defensive reaction to CNS pathologies. We highlighted specific aspects of reactive astrogliosis, especially with respect to the involvement of the P2X(7) and P2Y(1)R subtypes. Reactive astrogliosis exerts both beneficial and detrimental effects in a context-specific manner determined by distinct molecular signalling cascades. Understanding the role of purinergic signalling in astrocytes is critical to identifying new therapeutic principles to treat acute and chronic neurological diseases.
Collapse
Affiliation(s)
- Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstrasse 16-18, 04107, Leipzig, Germany.
| | | | | | | |
Collapse
|