1
|
Li WG, Li LX, Song RX, Wang XP, Jia SY, Ma XY, Zhang JY, Yin GF, Li XM, Zhang LM. Enhancement of Ca 2+ Signal Strength in Astrocytes in the Lateral Septum Improves Cognitive Disorders in Mice After Hemorrhagic Shock and Resuscitation. Neurosci Bull 2025:10.1007/s12264-025-01404-5. [PMID: 40279023 DOI: 10.1007/s12264-025-01404-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/19/2025] [Indexed: 04/26/2025] Open
Abstract
Hemorrhagic shock is a common clinical emergency that can aggravate cell injury after resuscitation. Astrocytes are crucial for the survival of neurons because they regulate the surrounding ionic microenvironment of neurons. Although hemorrhagic shock and resuscitation (HSR) injury can impair cognition, it remains unclear how this insult directly affects astrocytes. In this study, we established an HSR model by bleeding and re-transfusion in mice. The social interaction test and new object recognition test were applied to evaluate post-operative cognitive changes, and the results suggest that mice experience cognitive impairment following exposure to HSR. In the HSR group, the power spectral density of β and γ oscillations decreased, and the coupling of the θ oscillation phase and γ oscillation amplitude was abnormal, which indicated abnormal neuronal oscillation and cognitive impairment after HSR exposure. In brief, cognitive impairment in mice is strongly correlated with Ca2+ signal strength in lateral septum astrocytes following HSR.
Collapse
Affiliation(s)
- Wen-Guang Li
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, 061000, China
- Graduate School, Hebei Medical University, Shijiazhuang, 050011, China
| | - Lan-Xin Li
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, 061000, China
| | - Rong-Xin Song
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, 061000, China
| | - Xu-Peng Wang
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, 061000, China
| | - Xiao-Yi Ma
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, 061000, China
- Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Hebei University of Chinese Medicine, Cangzhou, 061000, China
| | - Jing-Yu Zhang
- Clinical Lab, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, 050200, China
| | - Gang-Feng Yin
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Xiao-Ming Li
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, 061000, China
| | - Li-Min Zhang
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, 061000, China.
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, 061000, China.
| |
Collapse
|
2
|
Phillips CD, DeFazio RA, Moenter SM. Sex and Time of Day Alter the Interactions Between Hypothalamic Glia and the Neural Circuits Controlling Reproduction. Endocrinology 2025; 166:bqaf057. [PMID: 40111184 PMCID: PMC11968336 DOI: 10.1210/endocr/bqaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
An upstream network, including glia and arcuate nucleus (ARC) kisspeptin neurons, controls hormone secretion from preoptic area (POA) gonadotropin-releasing hormone (GnRH) neurons, which form the final common pathway for the central control of fertility. In males, chemogenetic activation of Gq-mediated signaling in POA glia activated GnRH neurons and downstream luteinizing hormone (LH) release, while chemogenetic activation of ARC glia had no effect on ARC kisspeptin neurons. We characterized sex differences and time-of-day effects in these critical circuits to understand their effects on reproduction. Chemogenetic activation of glial fibrillary acidic protein (GFAP)-expressing cells increased intracellular calcium concentrations regardless of sex, brain region, or time of day. Activation of POA glia or treatment with the gliotransmitter analog dimethyl prostaglandin E2 (dmPGE2) increased GnRH neuron firing rate, and these responses were dependent upon sex and time of day. In contrast, ARC kisspeptin neuron firing rate was unresponsive to ARC glia activation or dmPGE2 regardless of sex or time of day. POA glial activation increased LH levels in males and females but the response in males was more rapid. ARC glia activation had no effect on LH in females and the response in males was delayed compared to POA glia activation. A similar LH response persisted after ARC kisspeptin neuron ablation, suggesting it is not mediated by those neurons. GnRH neurons, rather than arcuate kisspeptin neurons, are thus the main target of glial regulation of reproductive neuroendocrine output and this regulation is dependent on sex and time of day.
Collapse
Affiliation(s)
- Chrystian D Phillips
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - R Anthony DeFazio
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M Moenter
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Bree D, Zhao J, Stratton J, Levy D. Cortical astrocyte activation triggers meningeal nociception and migraine-like pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637109. [PMID: 39975142 PMCID: PMC11839082 DOI: 10.1101/2025.02.08.637109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Migraine attacks are believed to originate in the brain, but the exact mechanisms by which the brain generates peripheral nociceptive signals that drive migraine pain remain unclear. Sensory cortex hyperexcitability has been observed consistently across different migraine subtypes. Astrocytes detect aberrant increases in cortical activity via their Gq-coupled receptors and respond by releasing gliotransmitters and other factors with proinflammatory and nociceptive properties. In the present study, we used a rat model to investigate whether heightened cortical astrocyte Gq-coupled signaling is sufficient to drive peripheral trigeminal meningeal nociceptive responses linked to the generation of migraine headaches. We used an AAV-based chemogenetic approach that allows selective activation of cortical astrocyte Gq-GPCR signaling. We targeted astrocytes in the visual cortex as hyperexcitability in this region has been implicated in migraine. Furthermore, the meninges overlying the visual cortex are densely innervated by nociceptive fibers. We combined this chemogenetic approach with in vivo single-unit recording of meningeal nociceptors to assess changes in their ongoing activity and mechanosensitivity, along with testing of migraine-like behaviors. We further targeted calcitonin gene-related peptide (CGRP), using a monoclonal antibody (anti-CGRP mAb), to assess the relevance of cortical astrocyte activation to migraine. We discovered that heightened activation of Gq-coupled signaling in visual cortex astrocytes drives persistent discharge and increased mechanosensitivity of meningeal nociceptors. Cortical astrocytic activation also generated cephalic mechanical pain hypersensitivity, reduced exploratory behavior, and anxiety-like behaviors linked to migraine headaches. Blocking calcitonin gene-related peptide signaling suppressed astrocyte-mediated increases in meningeal nociceptor discharge and alleviated associated migraine-related behaviors. Our findings reveal a previously unappreciated role for augmented visual cortex astrocyte signaling as a triggering factor sufficient to generate meningeal nociception and migraine pain and greatly expand our understanding of migraine pathophysiology.
Collapse
Affiliation(s)
- Dara Bree
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Jun Zhao
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | | | - Dan Levy
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
- Lead Contact
| |
Collapse
|
4
|
Al-Jaf S, Soliman AY, El-Yazbi AF, Abd-Elrahman KS. Unveiling the Interplay: Neurovascular Coupling, Astrocytes and G Protein-Coupled Receptors in Alzheimer's Disease. ACS Pharmacol Transl Sci 2025; 8:271-285. [PMID: 39974631 PMCID: PMC11833731 DOI: 10.1021/acsptsci.4c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025]
Abstract
Astrocytes are a type of glial cell that are involved in actively modulating synaptic plasticity, neurotransmitter homeostasis, and neuroinflammatory responses. More importantly, they coordinate neuronal activity and cerebral blood flow (CBF) in what is known as neurovascular coupling (NVC). NVC is an essential mechanism that maintains the high energy demand the brain requires by supplying continuous and rapid supply of oxygen and nutrients through CBF. Impairment in NVC is one of the key events that triggers a spiral of occurrences that lead to the clinical advancement of Alzheimer's disease (AD). It is yet to be determined what the molecular manifestations of NVC impairment relate to; nonetheless, it is believed that alterations in G protein-coupled receptors (GPCRs) are responsible for exacerbating these effects. In this review, we summarize the current evidence supporting the involvement of GPCRs on astrocytes in NVC and the pathophysiology of AD. Additionally, we propose potential research directions to further elucidate the underlying mechanisms and evaluate the feasibility of targeting specific GPCRs as a therapeutic strategy to correct brain blood flow and memory impairments associated with AD.
Collapse
Affiliation(s)
- Sanarya Al-Jaf
- Department
of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian
Centre for Brain Health, The University
of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Alaa Y. Soliman
- Faculty
of Pharmacy and Research and Innovation Hub, Alamein International University, Alamein, Matrouh 51718, Egypt
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed F. El-Yazbi
- Faculty
of Pharmacy and Research and Innovation Hub, Alamein International University, Alamein, Matrouh 51718, Egypt
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Khaled S. Abd-Elrahman
- Department
of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian
Centre for Brain Health, The University
of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department
of Medical Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
5
|
Hernández-Martín N, Martínez MG, Bascuñana P, Fernández de la Rosa R, García-García L, Gómez F, Solas M, Martín ED, Pozo MA. Astrocytic Ca 2+ activation by chemogenetics mitigates the effect of kainic acid-induced excitotoxicity on the hippocampus. Glia 2024; 72:2217-2230. [PMID: 39188024 DOI: 10.1002/glia.24607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Astrocytes play a multifaceted role regulating brain glucose metabolism, ion homeostasis, neurotransmitters clearance, and water dynamics being essential in supporting synaptic function. Under different pathological conditions such as brain stroke, epilepsy, and neurodegenerative disorders, excitotoxicity plays a crucial role, however, the contribution of astrocytic activity in protecting neurons from excitotoxicity-induced damage is yet to be fully understood. In this work, we evaluated the effect of astrocytic activation by Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) on brain glucose metabolism in wild-type (WT) mice, and we investigated the effects of sustained astrocyte activation following an insult induced by intrahippocampal (iHPC) kainic acid (KA) injection using 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) imaging, along with behavioral test, nuclear magnetic resonance (NMR) spectroscopy and histochemistry. Astrocytic Ca2+ activation increased the 18F-FDG uptake, but this effect was not found when the study was performed in knock out mice for type-2 inositol 1,4,5-trisphosphate receptor (Ip3r2-/-) nor in floxed mice to abolish glucose transporter 1 (GLUT1) expression in hippocampal astrocytes (GLUT1ΔGFAP). Sustained astrocyte activation after KA injection reversed the brain glucose hypometabolism, restored hippocampal function, prevented neuronal death, and increased hippocampal GABA levels. The findings of our study indicate that astrocytic GLUT1 function is crucial for regulating brain glucose metabolism. Astrocytic Ca2+ activation has been shown to promote adaptive changes that significantly contribute to mitigating the effects of KA-induced damage. This evidence suggests a protective role of activated astrocytes against KA-induced excitotoxicity.
Collapse
Affiliation(s)
- Nira Hernández-Martín
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | | | - Pablo Bascuñana
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Rubén Fernández de la Rosa
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Bioimac, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis García-García
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisca Gómez
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Maite Solas
- Facultad de Farmacia, Universidad de Navarra, Pamplona, Spain
| | | | - Miguel A Pozo
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
6
|
Natsubori A, Kwon S, Honda Y, Kojima T, Karashima A, Masamoto K, Honda M. Serotonergic regulation of cortical neurovascular coupling and hemodynamics upon awakening from sleep in mice. J Cereb Blood Flow Metab 2024; 44:1591-1607. [PMID: 38477254 PMCID: PMC11418750 DOI: 10.1177/0271678x241238843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Neurovascular coupling (NVC) is the functional hyperemia of the brain responding to local neuronal activity. It is mediated by astrocytes and affected by subcortical ascending pathways in the cortex that convey information, such as sensory stimuli and the animal condition. Here, we investigate the influence of the raphe serotonergic system, a subcortical ascending arousal system in animals, on the modulation of cortical NVC and cerebral blood flow (CBF). Raphe serotonergic neurons were optogenically activated for 30 s, which immediately awakened the mice from non-rapid eye movement sleep. This caused a biphasic cortical hemodynamic change: a transient increase for a few seconds immediately after photostimulation onset, followed by a large progressive decrease during the stimulation period. Serotonergic neuron activation increased intracellular Ca2+ levels in cortical pyramidal neurons and astrocytes, demonstrating its effect on the NVC components. Pharmacological inhibition of cortical neuronal firing activity and astrocyte metabolic activity had small hypovolemic effects on serotonin-induced biphasic CBF changes, while blocking 5-HT1B receptors expressed primarily in cerebral vasculature attenuated the decreasing CBF phase. This suggests that serotonergic neuron activation leading to animal awakening could allow the NVC to exert a hyperemic function during a biphasic CBF response, with a predominant decrease in the cortex.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoshiko Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai, Japan
| | - Kazuto Masamoto
- Dept. Mechanical and Intelligent Systems Engineering, Univ. of Electro-Communications, Tokyo, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
7
|
Ogino T, Agetsuma M, Sawada M, Inada H, Nabekura J, Sawamoto K. Astrocytic activation increases blood flow in the adult olfactory bulb. Mol Brain 2024; 17:52. [PMID: 39107815 PMCID: PMC11301997 DOI: 10.1186/s13041-024-01126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Activation of astrocytes after sensory stimulation has been reported to be involved in increased blood flow in the central nervous system. In the present study, using a chemogenetic method to induce astrocyte activation in mice without sensory stimulation, we found that astrocytic activation led to increased blood flow in the olfactory bulb, suggesting that astrocyte activation is sufficient for increasing blood flow in the olfactory bulb. The technique established here will be useful for studying the mechanisms underlying sensory input-dependent blood flow increases.
Collapse
Affiliation(s)
- Takashi Ogino
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Masakazu Agetsuma
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Chiba Inage-ku, Chiba, 263-8555, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Hiroyuki Inada
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan.
| |
Collapse
|
8
|
Feng L, Gao L. The role of neurovascular coupling dysfunction in cognitive decline of diabetes patients. Front Neurosci 2024; 18:1375908. [PMID: 38576869 PMCID: PMC10991808 DOI: 10.3389/fnins.2024.1375908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Neurovascular coupling (NVC) is an important mechanism to ensure adequate blood supply to active neurons in the brain. NVC damage can lead to chronic impairment of neuronal function. Diabetes is characterized by high blood sugar and is considered an important risk factor for cognitive impairment. In this review, we provide fMRI evidence of NVC damage in diabetic patients with cognitive decline. Combined with the exploration of the major mechanisms and signaling pathways of NVC, we discuss the effects of chronic hyperglycemia on the cellular structure of NVC signaling, including key receptors, ion channels, and intercellular connections. Studying these diabetes-related changes in cell structure will help us understand the underlying causes behind diabetes-induced NVC damage and early cognitive decline, ultimately helping to identify the most effective drug targets for treatment.
Collapse
Affiliation(s)
| | - Ling Gao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Owens CD, Bonin Pinto C, Detwiler S, Olay L, Pinaffi-Langley ACDC, Mukli P, Peterfi A, Szarvas Z, James JA, Galvan V, Tarantini S, Csiszar A, Ungvari Z, Kirkpatrick AC, Prodan CI, Yabluchanskiy A. Neurovascular coupling impairment as a mechanism for cognitive deficits in COVID-19. Brain Commun 2024; 6:fcae080. [PMID: 38495306 PMCID: PMC10943572 DOI: 10.1093/braincomms/fcae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Components that comprise our brain parenchymal and cerebrovascular structures provide a homeostatic environment for proper neuronal function to ensure normal cognition. Cerebral insults (e.g. ischaemia, microbleeds and infection) alter cellular structures and physiologic processes within the neurovascular unit and contribute to cognitive dysfunction. COVID-19 has posed significant complications during acute and convalescent stages in multiple organ systems, including the brain. Cognitive impairment is a prevalent complication in COVID-19 patients, irrespective of severity of acute SARS-CoV-2 infection. Moreover, overwhelming evidence from in vitro, preclinical and clinical studies has reported SARS-CoV-2-induced pathologies in components of the neurovascular unit that are associated with cognitive impairment. Neurovascular unit disruption alters the neurovascular coupling response, a critical mechanism that regulates cerebromicrovascular blood flow to meet the energetic demands of locally active neurons. Normal cognitive processing is achieved through the neurovascular coupling response and involves the coordinated action of brain parenchymal cells (i.e. neurons and glia) and cerebrovascular cell types (i.e. endothelia, smooth muscle cells and pericytes). However, current work on COVID-19-induced cognitive impairment has yet to investigate disruption of neurovascular coupling as a causal factor. Hence, in this review, we aim to describe SARS-CoV-2's effects on the neurovascular unit and how they can impact neurovascular coupling and contribute to cognitive decline in acute and convalescent stages of the disease. Additionally, we explore potential therapeutic interventions to mitigate COVID-19-induced cognitive impairment. Given the great impact of cognitive impairment associated with COVID-19 on both individuals and public health, the necessity for a coordinated effort from fundamental scientific research to clinical application becomes imperative. This integrated endeavour is crucial for mitigating the cognitive deficits induced by COVID-19 and its subsequent burden in this especially vulnerable population.
Collapse
Affiliation(s)
- Cameron D Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Ana Clara da C Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Judith A James
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Veronica Galvan
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Angelia C Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
10
|
Ozawa K, Nagao M, Konno A, Iwai Y, Vittani M, Kusk P, Mishima T, Hirai H, Nedergaard M, Hirase H. Astrocytic GPCR-Induced Ca 2+ Signaling Is Not Causally Related to Local Cerebral Blood Flow Changes. Int J Mol Sci 2023; 24:13590. [PMID: 37686396 PMCID: PMC10487464 DOI: 10.3390/ijms241713590] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/17/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Activation of Gq-type G protein-coupled receptors (GPCRs) gives rise to large cytosolic Ca2+ elevations in astrocytes. Previous in vitro and in vivo studies have indicated that astrocytic Ca2+ elevations are closely associated with diameter changes in the nearby blood vessels, which astrocytes enwrap with their endfeet. However, the causal relationship between astrocytic Ca2+ elevations and blood vessel diameter changes has been questioned, as mice with diminished astrocytic Ca2+ signaling show normal sensory hyperemia. We addressed this controversy by imaging cortical vasculature while optogenetically elevating astrocyte Ca2+ in a novel transgenic mouse line, expressing Opto-Gq-type GPCR Optoα1AR (Astro-Optoα1AR) in astrocytes. Blue light illumination on the surface of the somatosensory cortex induced Ca2+ elevations in cortical astrocytes and their endfeet in mice under anesthesia. Blood vessel diameter did not change significantly with Optoα1AR-induced Ca2+ elevations in astrocytes, while it was increased by forelimb stimulation. Next, we labeled blood plasma with red fluorescence using AAV8-P3-Alb-mScarlet in Astro-Optoα1AR mice. We were able to identify arterioles that display diameter changes in superficial areas of the somatosensory cortex through the thinned skull. Photo-stimulation of astrocytes in the cortical area did not result in noticeable changes in the arteriole diameters compared with their background strain C57BL/6. Together, compelling evidence for astrocytic Gq pathway-induced vasodiameter changes was not observed. Our results support the notion that short-term (<10 s) hyperemia is not mediated by GPCR-induced astrocytic Ca2+ signaling.
Collapse
Affiliation(s)
- Katsuya Ozawa
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako 351-0106, Saitama, Japan; (K.O.)
| | - Masaki Nagao
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan
- Viral Vector Core, Gunma University, Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Youichi Iwai
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako 351-0106, Saitama, Japan; (K.O.)
| | - Marta Vittani
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Peter Kusk
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Tsuneko Mishima
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan
- Viral Vector Core, Gunma University, Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
11
|
Pereira MJ, Ayana R, Holt MG, Arckens L. Chemogenetic manipulation of astrocyte activity at the synapse- a gateway to manage brain disease. Front Cell Dev Biol 2023; 11:1193130. [PMID: 37534103 PMCID: PMC10393042 DOI: 10.3389/fcell.2023.1193130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Astrocytes are the major glial cell type in the central nervous system (CNS). Initially regarded as supportive cells, it is now recognized that this highly heterogeneous cell population is an indispensable modulator of brain development and function. Astrocytes secrete neuroactive molecules that regulate synapse formation and maturation. They also express hundreds of G protein-coupled receptors (GPCRs) that, once activated by neurotransmitters, trigger intracellular signalling pathways that can trigger the release of gliotransmitters which, in turn, modulate synaptic transmission and neuroplasticity. Considering this, it is not surprising that astrocytic dysfunction, leading to synaptic impairment, is consistently described as a factor in brain diseases, whether they emerge early or late in life due to genetic or environmental factors. Here, we provide an overview of the literature showing that activation of genetically engineered GPCRs, known as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to specifically modulate astrocyte activity partially mimics endogenous signalling pathways in astrocytes and improves neuronal function and behavior in normal animals and disease models. Therefore, we propose that expressing these genetically engineered GPCRs in astrocytes could be a promising strategy to explore (new) signalling pathways which can be used to manage brain disorders. The precise molecular, functional and behavioral effects of this type of manipulation, however, differ depending on the DREADD receptor used, targeted brain region and timing of the intervention, between healthy and disease conditions. This is likely a reflection of regional and disease/disease progression-associated astrocyte heterogeneity. Therefore, a thorough investigation of the effects of such astrocyte manipulation(s) must be conducted considering the specific cellular and molecular environment characteristic of each disease and disease stage before this has therapeutic applicability.
Collapse
Affiliation(s)
- Maria João Pereira
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Rajagopal Ayana
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Instituto de Investigação e Inovação em Saúde (i3S), Laboratory of Synapse Biology, Universidade do Porto, Porto, Portugal
| | - Lutgarde Arckens
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
12
|
Lia A, Di Spiezio A, Speggiorin M, Zonta M. Two decades of astrocytes in neurovascular coupling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1162757. [PMID: 37078069 PMCID: PMC10106690 DOI: 10.3389/fnetp.2023.1162757] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
The brain is a highly energy demanding organ, which accounts in humans for the 20% of total energy consumption at resting state although comprising only 2% of the body mass. The necessary delivery of nutrients to brain parenchyma is ensured by the cerebral circulatory system, through the exchange of glucose and oxygen (O2) at the capillary level. Notably, a tight spatial and temporal correlation exists between local increases in neuronal activity and the subsequent changes in regional cerebral blood flow. The recognized concept of neurovascular coupling (NVC), also named functional hyperemia, expresses this close relationship and stands at the basis of the modern functional brain imaging techniques. Different cellular and molecular mechanisms have been proposed to mediate this tight coupling. In this context, astrocytes are ideally positioned to act as relay elements that sense neuronal activity through their perisynaptic processes and release vasodilator agents at their endfeet in contact with brain parenchymal vessels. Two decades after the astrocyte involvement in neurovascular coupling has been proposed, we here review the experimental evidence that contributed to unraveling the molecular and cellular mechanisms underlying cerebral blood flow regulation. While traveling through the different controversies that moved the research in this field, we keep a peculiar focus on those exploring the role of astrocytes in neurovascular coupling and conclude with two sections related to methodological aspects in neurovascular research and to some pathological conditions resulting in altered neurovascular coupling.
Collapse
Affiliation(s)
- Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
| | - Alessandro Di Spiezio
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
| |
Collapse
|
13
|
Chen X, Lu W, Lu C, Zhang L, Xu F, Dong H. The CaSR/TRPV4 coupling mediates pro-inflammatory macrophage function. Acta Physiol (Oxf) 2023; 237:e13926. [PMID: 36606511 DOI: 10.1111/apha.13926] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023]
Abstract
AIM Although calcium-sensing receptor (CaSR) and transient receptor potential vanilloid 4 (TRPV4) channels are functionally expressed on macrophages, it is unclear if they work coordinately to mediate macrophage function. The present study investigates whether CaSR couples to TRPV4 channels and mediates macrophage polarization via Ca2+ signaling. METHODS The role of CaSR/TRPV4/Ca2+ signaling was assessed in lipopolysaccharide (LPS)-treated peritoneal macrophages (PMs) from wild-type (WT) and TRPV4 knockout (TRPV4 KO) mice. The expression and function of CaSR and TRPV4 in PMs were analyzed by immunofluorescence and digital Ca2+ imaging. The correlation factors of M1 polarization, CCR7, IL-1β, and TNFα were detected using q-PCR, western blot, and ELISA. RESULTS We found that PMs expressed CaSR and TRPV4, and CaSR activation-induced marked Ca2+ signaling predominately through extracellular Ca2+ entry, which was inhibited by selective pharmacological blockers of CaSR and TRPV4 channels. The CaSR activation-induced Ca2+ signaling was significantly attenuated in PMs from TRPV4 KO mice compared to those from WT mice. Moreover, the CaSR activation-induced Ca2+ entry via TRPV4 channels was inhibited by blocking phospholipases A2 (PLA2)/cytochromeP450 (CYP450) and phospholipase C (PLC)/Protein kinase C (PKC) pathways. Finally, CaSR activation promoted the expression and release of M1-associated cytokines IL-1β and TNFɑ, which were attenuated in PMs from TRPV4 KO mice. CONCLUSION We reveal a novel coupling of the CaSR and TRPV4 channels via PLA2/CYP450 and PLC/PKC pathways, promoting a Ca2+ -dependent M1 macrophage polarization. Modulation of this coupling and downstream pathways may become a potential strategy for the prevention/treatment of immune-related disease.
Collapse
Affiliation(s)
- Xiongying Chen
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Lu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luyun Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
14
|
Lee SH, Mak A, Verheijen MHG. Comparative assessment of the effects of DREADDs and endogenously expressed GPCRs in hippocampal astrocytes on synaptic activity and memory. Front Cell Neurosci 2023; 17:1159756. [PMID: 37051110 PMCID: PMC10083367 DOI: 10.3389/fncel.2023.1159756] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) have proven themselves as one of the key in vivo techniques of modern neuroscience, allowing for unprecedented access to cellular manipulations in living animals. With respect to astrocyte research, DREADDs have become a popular method to examine the functional aspects of astrocyte activity, particularly G-protein coupled receptor (GPCR)-mediated intracellular calcium (Ca2+) and cyclic adenosine monophosphate (cAMP) dynamics. With this method it has become possible to directly link the physiological aspects of astrocytic function to cognitive processes such as memory. As a result, a multitude of studies have explored the impact of DREADD activation in astrocytes on synaptic activity and memory. However, the emergence of varying results prompts us to reconsider the degree to which DREADDs expressed in astrocytes accurately mimic endogenous GPCR activity. Here we compare the major downstream signaling mechanisms, synaptic, and behavioral effects of stimulating Gq-, Gs-, and Gi-DREADDs in hippocampal astrocytes of adult mice to those of endogenously expressed GPCRs.
Collapse
Affiliation(s)
- Sophie H. Lee
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Research Master’s Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Aline Mak
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Mark Verheijen,
| |
Collapse
|
15
|
Mughal A, Nelson MT, Hill-Eubanks D. The post-arteriole transitional zone: a specialized capillary region that regulates blood flow within the CNS microvasculature. J Physiol 2023; 601:889-901. [PMID: 36751860 PMCID: PMC9992301 DOI: 10.1113/jp282246] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
The brain is an energy hog, consuming available energy supplies at a rate out of all proportion to its relatively small size. This outsized demand, largely reflecting the unique computational activity of the brain, is met by an ensemble of neurovascular coupling mechanisms that link neuronal activity with local increases in blood delivery. This just-in-time replenishment strategy, made necessary by the limited energy-storage capacity of neurons, complicates the nutrient-delivery task of the cerebral vasculature, layering on a temporo-spatial requirement that invites - and challenges - mechanistic interpretation. The centre of gravity of research efforts to disentangle these mechanisms has shifted from an initial emphasis on astrocyte-arteriole-level processes to mechanisms that operate on the capillary level, a shift that has brought into sharp focus questions regarding the fine control of blood distribution to active neurons. As these investigations have drilled down into finer reaches of the microvasculature, they have revealed an arteriole-proximate subregion of CNS capillary networks that serves a regulatory function in directing blood flow into and within downstream capillaries. They have also illuminated differences in researchers' perspectives on the vascular structures and identity of mural cells in this region that impart the vasomodulatory effects that control blood distribution. In this review, we highlight the regulatory role of a variably named region of the microvasculature, referred to here as the post-arteriole transition zone, in channeling blood flow within CNS capillary networks, and underscore the contribution of dynamically contractile perivascular mural cell - generally, but not universally, recognized as pericytes - to this function.
Collapse
Affiliation(s)
- Amreen Mughal
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Mark T. Nelson
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | | |
Collapse
|
16
|
Shiratori-Hayashi M, Tsuda M. IP 3R1-dependent astrocyte calcium signaling in chronic itch. Neurosci Res 2023; 187:40-44. [PMID: 36181909 DOI: 10.1016/j.neures.2022.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 10/14/2022]
Abstract
Astrocytes, the most abundant type of glial cell, are electrically non-excitable cells that use intracellular calcium (Ca2+) for functional regulation. Changes in intracellular Ca2+ concentration play important roles in the central nervous system (CNS), as they are involved in the release of gliotransmitters and the control of extracellular ion concentrations, thereby affecting the regulation of neuronal excitability, CNS homeostasis, and behavior. Intracellular calcium mobilization in astrocytes is known to be mediated via inositol 1,4,5-trisphosphate receptors (IP3Rs), particularly IP3R2, and its association with CNS pathogenesis has been widely reported. In addition, the existence of IP3R2-independent calcium signaling has recently been postulated; however, the detailed mechanisms and its role in astrocyte functions and CNS pathogenesis are still poorly understood. In this paper, we describe the putative mechanisms underlying IP3R1-dependent calcium signaling in astrocytes and its effects on the reactive state, compare this signaling with IP3R2-dependent calcium signaling, and discuss its contribution to chronic itch-like behavior.
Collapse
Affiliation(s)
- Miho Shiratori-Hayashi
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; Kyushu University Institute for Advanced Study, Fukuoka, Japan.
| |
Collapse
|
17
|
Institoris A, Vandal M, Peringod G, Catalano C, Tran CH, Yu X, Visser F, Breiteneder C, Molina L, Khakh BS, Nguyen MD, Thompson RJ, Gordon GR. Astrocytes amplify neurovascular coupling to sustained activation of neocortex in awake mice. Nat Commun 2022; 13:7872. [PMID: 36550102 PMCID: PMC9780254 DOI: 10.1038/s41467-022-35383-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Functional hyperemia occurs when enhanced neuronal activity signals to increase local cerebral blood flow (CBF) to satisfy regional energy demand. Ca2+ elevation in astrocytes can drive arteriole dilation to increase CBF, yet affirmative evidence for the necessity of astrocytes in functional hyperemia in vivo is lacking. In awake mice, we discovered that functional hyperemia is bimodal with a distinct early and late component whereby arteriole dilation progresses as sensory stimulation is sustained. Clamping astrocyte Ca2+ signaling in vivo by expressing a plasma membrane Ca2+ ATPase (CalEx) reduces sustained but not brief sensory-evoked arteriole dilation. Elevating astrocyte free Ca2+ using chemogenetics selectively augments sustained hyperemia. Antagonizing NMDA-receptors or epoxyeicosatrienoic acid production reduces only the late component of functional hyperemia, leaving brief increases in CBF to sensory stimulation intact. We propose that a fundamental role of astrocyte Ca2+ is to amplify functional hyperemia when neuronal activation is prolonged.
Collapse
Affiliation(s)
- Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Milène Vandal
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Govind Peringod
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Christy Catalano
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Cam Ha Tran
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557-352, USA
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Molecular and Integrative Physiology, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Frank Visser
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Cheryl Breiteneder
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Leonardo Molina
- Hotchkiss Brain Institute, Department of Clinical Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Department of Clinical Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
18
|
Ding F, Liang S, Li R, Yang Z, He Y, Yang S, Duan Q, Zhang J, Lyu J, Zhou Z, Huang M, Wang H, Li J, Yang C, Wang Y, Gong M, Chen S, Jia H, Chen X, Liao X, Fu L, Zhang K. Astrocytes exhibit diverse Ca2+ changes at subcellular domains during brain aging. Front Aging Neurosci 2022; 14:1029533. [PMID: 36389078 PMCID: PMC9650392 DOI: 10.3389/fnagi.2022.1029533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Astrocytic Ca2+ transients are essential for astrocyte integration into neural circuits. These Ca2+ transients are primarily sequestered in subcellular domains, including primary branches, branchlets and leaflets, and endfeet. In previous studies, it suggests that aging causes functional defects in astrocytes. Until now, it was unclear whether and how aging affects astrocytic Ca2+ transients at subcellular domains. In this study, we combined a genetically encoded Ca2+ sensor (GCaMP6f) and in vivo two-photon Ca2+ imaging to determine changes in Ca2+ transients within astrocytic subcellular domains during brain aging. We showed that aging increased Ca2+ transients in astrocytic primary branches, higher-order branchlets, and terminal leaflets. However, Ca2+ transients decreased within astrocytic endfeet during brain aging, which could be caused by the decreased expressions of Aquaporin-4 (AQP4). In addition, aging-induced changes of Ca2+ transient types were heterogeneous within astrocytic subcellular domains. These results demonstrate that the astrocytic Ca2+ transients within subcellular domains are affected by aging differently. This finding contributes to a better understanding of the physiological role of astrocytes in aging-induced neural circuit degeneration.
Collapse
Affiliation(s)
- Fusheng Ding
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shanshan Liang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Ruijie Li
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Yong He
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shaofan Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Qingtian Duan
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jianxiong Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jing Lyu
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Zhenqiao Zhou
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Mingzhu Huang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Haoyu Wang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jin Li
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Yuxia Wang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shangbin Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Jia
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, China
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Guangyang Bay Laboratory, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
- *Correspondence: Xiang Liao,
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
- Ling Fu,
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Kuan Zhang,
| |
Collapse
|
19
|
Butcher JB, Sims RE, Ngum NM, Bazzari AH, Jenkins SI, King M, Hill EJ, Nagel DA, Fox K, Parri HR, Glazewski S. A requirement for astrocyte IP3R2 signaling for whisker experience-dependent depression and homeostatic upregulation in the mouse barrel cortex. Front Cell Neurosci 2022; 16:905285. [PMID: 36090792 PMCID: PMC9452848 DOI: 10.3389/fncel.2022.905285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022] Open
Abstract
Changes to sensory experience result in plasticity of synapses in the cortex. This experience-dependent plasticity (EDP) is a fundamental property of the brain. Yet, while much is known about neuronal roles in EDP, very little is known about the role of astrocytes. To address this issue, we used the well-described mouse whiskers-to-barrel cortex system, which expresses a number of forms of EDP. We found that all-whisker deprivation induced characteristic experience-dependent Hebbian depression (EDHD) followed by homeostatic upregulation in L2/3 barrel cortex of wild type mice. However, these changes were not seen in mutant animals (IP3R2–/–) that lack the astrocyte-expressed IP3 receptor subtype. A separate paradigm, the single-whisker experience, induced potentiation of whisker-induced response in both wild-type (WT) mice and IP3R2–/– mice. Recordings in ex vivo barrel cortex slices reflected the in vivo results so that long-term depression (LTD) could not be elicited in slices from IP3R2–/– mice, but long-term potentiation (LTP) could. Interestingly, 1 Hz stimulation inducing LTD in WT paradoxically resulted in NMDAR-dependent LTP in slices from IP3R2–/– animals. The LTD to LTP switch was mimicked by acute buffering astrocytic [Ca2+]i in WT slices. Both WT LTD and IP3R2–/– 1 Hz LTP were mediated by non-ionotropic NMDAR signaling, but only WT LTD was P38 MAPK dependent, indicating an underlying mechanistic switch. These results demonstrate a critical role for astrocytic [Ca2+]i in several EDP mechanisms in neocortex.
Collapse
Affiliation(s)
- John B. Butcher
- School of Life Sciences, Keele University, Keele, United Kingdom
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Robert E. Sims
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Neville M. Ngum
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Amjad H. Bazzari
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Stuart I. Jenkins
- Neural Tissue Engineering Group, Institute for Science and Technology in Medicine (ISTM), Keele University, Keele, United Kingdom
| | - Marianne King
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Eric J. Hill
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - David A. Nagel
- Aston Medical School, Aston Medical Research Institute, Aston University, Birmingham, United Kingdom
| | - Kevin Fox
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - H. Rheinallt Parri
- College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
- *Correspondence: H. Rheinallt Parri,
| | - Stanislaw Glazewski
- School of Life Sciences, Keele University, Keele, United Kingdom
- Stanislaw Glazewski,
| |
Collapse
|
20
|
Nippert AR, Chiang PP, Del Franco AP, Newman EA. Astrocyte regulation of cerebral blood flow during hypoglycemia. J Cereb Blood Flow Metab 2022; 42:1534-1546. [PMID: 35296178 PMCID: PMC9274859 DOI: 10.1177/0271678x221089091] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022]
Abstract
Hypoglycemia triggers increases in cerebral blood flow (CBF), augmenting glucose supply to the brain. We have tested whether astrocytes, which can regulate vessel tone, contribute to this CBF increase. We hypothesized that hypoglycemia-induced adenosine signaling acts to increase astrocyte Ca2+ activity, which then causes the release of prostaglandins (PGs) and epoxyeicosatrienoic acids (EETs), leading to the dilation of brain arterioles and blood flow increases. We used an awake mouse model to investigate the effects of insulin-induced hypoglycemia on arterioles and astrocytes in the somatosensory cortex. During insulin-induced hypoglycemia, penetrating arterioles dilated and astrocyte Ca2+ signaling increased when blood glucose dropped below a threshold of ∼50 mg/dL. Application of the A2A adenosine receptor antagonist ZM-241385 eliminated hypoglycemia-evoked astrocyte Ca2+ increases and reduced arteriole dilations by 44% (p < 0.05). SC-560 and miconazole, which block the production of the astrocyte vasodilators PGs and EETs respectively, reduced arteriole dilations in response to hypoglycemia by 89% (p < 0.001) and 76% (p < 0.001). Hypoglycemia-induced arteriole dilations were decreased by 65% (p < 0.001) in IP3R2 knockout mice, which have reduced astrocyte Ca2+ signaling compared to wild-type. These results support the hypothesis that astrocytes contribute to hypoglycemia-induced increases in CBF by releasing vasodilators in a Ca2+-dependent manner.
Collapse
Affiliation(s)
- Amy R Nippert
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | | | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Zhu WM, Neuhaus A, Beard DJ, Sutherland BA, DeLuca GC. Neurovascular coupling mechanisms in health and neurovascular uncoupling in Alzheimer's disease. Brain 2022; 145:2276-2292. [PMID: 35551356 PMCID: PMC9337814 DOI: 10.1093/brain/awac174] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
To match the metabolic demands of the brain, mechanisms have evolved to couple neuronal activity to vasodilation, thus increasing local cerebral blood flow and delivery of oxygen and glucose to active neurons. Rather than relying on metabolic feedback signals such as the consumption of oxygen or glucose, the main signalling pathways rely on the release of vasoactive molecules by neurons and astrocytes, which act on contractile cells. Vascular smooth muscle cells and pericytes are the contractile cells associated with arterioles and capillaries, respectively, which relax and induce vasodilation. Much progress has been made in understanding the complex signalling pathways of neurovascular coupling, but issues such as the contributions of capillary pericytes and astrocyte calcium signal remain contentious. Study of neurovascular coupling mechanisms is especially important as cerebral blood flow dysregulation is a prominent feature of Alzheimer’s disease. In this article we will discuss developments and controversies in the understanding of neurovascular coupling and finish by discussing current knowledge concerning neurovascular uncoupling in Alzheimer’s disease.
Collapse
Affiliation(s)
- Winston M Zhu
- Oxford Medical School, University of Oxford, Oxford, UK
| | - Ain Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Tran CHT. Toolbox for studying neurovascular coupling in vivo, with a focus on vascular activity and calcium dynamics in astrocytes. NEUROPHOTONICS 2022; 9:021909. [PMID: 35295714 PMCID: PMC8920490 DOI: 10.1117/1.nph.9.2.021909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/23/2022] [Indexed: 05/14/2023]
Abstract
Significance: Insights into the cellular activity of each member of the neurovascular unit (NVU) is critical for understanding their contributions to neurovascular coupling (NVC)-one of the key control mechanisms in cerebral blood flow regulation. Advances in imaging and genetic tools have enhanced our ability to observe, manipulate and understand the cellular activity of NVU components, namely neurons, astrocytes, microglia, endothelial cells, vascular smooth muscle cells, and pericytes. However, there are still many unresolved questions. Since astrocytes are considered electrically unexcitable,Ca 2 + signaling is the main parameter used to monitor their activity. It is therefore imperative to study astrocyticCa 2 + dynamics simultaneously with vascular activity using tools appropriate for the question of interest. Aim: To highlight currently available genetic and imaging tools for studying the NVU-and thus NVC-with a focus on astrocyteCa 2 + dynamics and vascular activity, and discuss the utility, technical advantages, and limitations of these tools for elucidating NVC mechanisms. Approach: We draw attention to some outstanding questions regarding the mechanistic basis of NVC and emphasize the role of astrocyticCa 2 + elevations in functional hyperemia. We further discuss commonly used genetic, and optical imaging tools, as well as some newly developed imaging modalities for studying NVC at the cellular level, highlighting their advantages and limitations. Results: We provide an overview of the current state of NVC research, focusing on the role of astrocyticCa 2 + elevations in functional hyperemia; summarize recent advances in genetically engineeredCa 2 + indicators, fluorescence microscopy techniques for studying NVC; and discuss the unmet challenges for future imaging development. Conclusions: Advances in imaging techniques together with improvements in genetic tools have significantly contributed to our understanding of NVC. Many pieces of the puzzle have been revealed, but many more remain to be discovered. Ultimately, optimizing NVC research will require a concerted effort to improve imaging techniques, available genetic tools, and analytical software.
Collapse
Affiliation(s)
- Cam Ha T. Tran
- University of Nevada, Reno School of Medicine, Department of Physiology and Cell Biology, Reno, Nevada, United States
| |
Collapse
|
23
|
Del Franco AP, Chiang PP, Newman EA. Dilation of cortical capillaries is not related to astrocyte calcium signaling. Glia 2022; 70:508-521. [PMID: 34767261 PMCID: PMC8732319 DOI: 10.1002/glia.24119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 12/30/2022]
Abstract
The brain requires an adequate supply of oxygen and nutrients to maintain proper function as neuronal activity varies. This is achieved, in part, through neurovascular coupling mechanisms that mediate local increases in blood flow through the dilation of arterioles and capillaries. The role of astrocytes in mediating this functional hyperemia response is controversial. Specifically, the function of astrocyte Ca2+ signaling is unclear. Cortical arterioles dilate in the absence of astrocyte Ca2+ signaling, but previous work suggests that Ca2+ increases are necessary for capillary dilation. This question has not been fully addressed in vivo, however, and we have reexamined the role of astrocyte Ca2+ signaling in vessel dilation in the barrel cortex of awake, behaving mice. We recorded evoked vessel dilations and astrocyte Ca2+ signaling in response to whisker stimulation. Experiments were carried out on WT and IP3R2 KO mice, a transgenic model where astrocyte Ca2+ signaling is substantially reduced. Compared to WT mice at rest, Ca2+ signaling in astrocyte endfeet contacting capillaries increased by 240% when whisker stimulation evoked running. In contrast, Ca2+ signaling was reduced to 9% of WT values in IP3R2 KO mice. In all three conditions, however, the amplitude of capillary dilation was largely unchanged. In addition, the latency to the onset of astrocyte Ca2+ signaling lagged behind dilation onset in most trials, although a subset of rapid onset Ca2+ events with latencies as short as 0.15 s occurred. In summary, we found that whisker stimulation-evoked capillary dilations occurred independent of astrocyte Ca2+ increases in the cerebral cortex.
Collapse
Affiliation(s)
- Armani P Del Franco
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
24
|
Georgiou L, Echeverría A, Georgiou A, Kuhn B. Ca + activity maps of astrocytes tagged by axoastrocytic AAV transfer. SCIENCE ADVANCES 2022; 8:eabe5371. [PMID: 35138891 PMCID: PMC8827655 DOI: 10.1126/sciadv.abe5371] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/17/2021] [Indexed: 05/27/2023]
Abstract
Astrocytes exhibit localized Ca2+ microdomain (MD) activity thought to be actively involved in information processing in the brain. However, functional organization of Ca2+ MDs in space and time in relationship to behavior and neuronal activity is poorly understood. Here, we first show that adeno-associated virus (AAV) particles transfer anterogradely from axons to astrocytes. Then, we use this axoastrocytic AAV transfer to express genetically encoded Ca2+ indicators at high-contrast circuit specifically. In combination with two-photon microscopy and unbiased, event-based analysis, we investigated cortical astrocytes embedded in the vibrissal thalamocortical circuit. We found a wide range of Ca2+ MD signals, some of which were ultrafast (≤300 ms). Frequency and size of signals were extensively increased by locomotion but only subtly with sensory stimulation. The overlay of these signals resulted in behavior-dependent maps with characteristic Ca2+ activity hotspots, maybe representing memory engrams. These functional subdomains are stable over days, suggesting subcellular specialization.
Collapse
|
25
|
Xie AX, Iguchi N, Clarkson TC, Malykhina AP. Pharmacogenetic inhibition of lumbosacral sensory neurons alleviates visceral hypersensitivity in a mouse model of chronic pelvic pain. PLoS One 2022; 17:e0262769. [PMID: 35077502 PMCID: PMC8789164 DOI: 10.1371/journal.pone.0262769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
The study investigated the cellular and molecular mechanisms in the peripheral nervous system (PNS) underlying the symptoms of urologic chronic pelvic pain syndrome (UCPPS) in mice. This work also aimed to test the feasibility of reversing peripheral sensitization in vivo in alleviating UCPPS symptoms. Intravesical instillation of vascular endothelial growth factor A (VEGFA) was used to induce UCPPS-like symptoms in mice. Spontaneous voiding spot assays and manual Von Frey tests were used to evaluate the severity of lower urinary tract symptoms (LUTS) and visceral hypersensitivity in VEGFA-instilled mice. Bladder smooth muscle strip contractility recordings (BSMSC) were used to identify the potential changes in myogenic and neurogenic detrusor muscle contractility at the tissue-level. Quantitative real-time PCR (qPCR) and fluorescent immunohistochemistry were performed to compare the expression levels of VEGF receptors and nociceptors in lumbosacral dorsal root ganglia (DRG) between VEGFA-instilled mice and saline-instilled controls. To manipulate primary afferent activity, Gi-coupled Designer Receptors Exclusively Activated by Designer Drugs (Gi-DREADD) were expressed in lumbosacral DRG neurons of TRPV1-Cre-ZGreen mice via targeted adeno-associated viral vector (AAVs) injections. A small molecule agonist of Gi-DREADD, clozapine-N-oxide (CNO), was injected into the peritoneum (i. p.) in awake animals to silence TRPV1 expressing sensory neurons in vivo during physiological and behavioral recordings of bladder function. Intravesical instillation of VEGFA in the urinary bladders increased visceral mechanical sensitivity and enhanced RTX-sensitive detrusor contractility. Sex differences were identified in the baseline detrusor contractility responses and VEGF-induced visceral hypersensitivity. VEGFA instillations in the urinary bladder led to significant increases in the mRNA and protein expression of transient receptor potential cation channel subfamily A member 1 (TRPA1) in lumbosacral DRG, whereas the expression levels of transient receptor potential cation channel subfamily V member 1 (TRPV1) and VEGF receptors (VEGFR1 and VEGFR2) remained unchanged when compared to saline-instilled animals. Importantly, the VEGFA-induced visceral hypersensitivity was reversed by Gi-DREADD-mediated neuronal silencing in lumbosacral sensory neurons. Activation of bladder VEGF signaling causes sensory neural plasticity and visceral hypersensitivity in mice, confirming its role of an UCPPS biomarker as identified by the Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) research studies. Pharmacogenetic inhibition of lumbosacral sensory neurons in vivo completely reversed VEGFA-induced pelvic hypersensitivity in mice, suggesting the strong therapeutic potential for decreasing primary afferent activity in the treatment of pain severity in UCPPS patients.
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Nao Iguchi
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Taylor C. Clarkson
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Anna P. Malykhina
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| |
Collapse
|
26
|
Akther S, Hirase H. Assessment of astrocytes as a mediator of memory and learning in rodents. Glia 2021; 70:1484-1505. [PMID: 34582594 DOI: 10.1002/glia.24099] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/26/2022]
Abstract
The classical view of astrocytes is that they provide supportive functions for neurons, transporting metabolites and maintaining the homeostasis of the extracellular milieu. This view is gradually changing with the advent of molecular genetics and optical methods allowing interrogation of selected cell types in live experimental animals. An emerging view that astrocytes additionally act as a mediator of synaptic plasticity and contribute to learning processes has gained in vitro and in vivo experimental support. Here we focus on the literature published in the past two decades to review the roles of astrocytes in brain plasticity in rodents, whereby the roles of neurotransmitters and neuromodulators are considered to be comparable to those in humans. We outline established inputs and outputs of astrocytes and discuss how manipulations of astrocytes have impacted the behavior in various learning paradigms. Multiple studies suggest that the contribution of astrocytes has a considerably longer time course than neuronal activation, indicating metabolic roles of astrocytes. We advocate that exploring upstream and downstream mechanisms of astrocytic activation will further provide insight into brain plasticity and memory/learning impairment.
Collapse
Affiliation(s)
- Sonam Akther
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Van Steenbergen V, Bareyre FM. Chemogenetic approaches to unravel circuit wiring and related behavior after spinal cord injury. Exp Neurol 2021; 345:113839. [PMID: 34389362 DOI: 10.1016/j.expneurol.2021.113839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 01/21/2023]
Abstract
A critical shortcoming of the central nervous system is its limited ability to repair injured nerve connections. Trying to overcome this limitation is not only relevant to understand basic neurobiological principles but also holds great promise to advance therapeutic strategies related, in particular, to spinal cord injury (SCI). With barely any SCI patients re-gaining complete neurological function, there is a high need to understand how we could target and improve spinal plasticity to re-establish neuronal connections into a functional network. The development of chemogenetic tools has proven to be of great value to understand functional circuit wiring before and after injury and to correlate novel circuit formation with behavioral outcomes. This review covers commonly used chemogenetic approaches based on metabotropic receptors and their use to improve our understanding of circuit wiring following spinal cord injury.
Collapse
Affiliation(s)
- Valérie Van Steenbergen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany; Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg-Martinsried, Germany.
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany; Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg-Martinsried, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
28
|
Haidey JN, Peringod G, Institoris A, Gorzo KA, Nicola W, Vandal M, Ito K, Liu S, Fielding C, Visser F, Nguyen MD, Gordon GR. Astrocytes regulate ultra-slow arteriole oscillations via stretch-mediated TRPV4-COX-1 feedback. Cell Rep 2021; 36:109405. [PMID: 34348138 DOI: 10.1016/j.celrep.2021.109405] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/22/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022] Open
Abstract
Very-low-frequency oscillations in microvascular diameter cause fluctuations in oxygen delivery that are important for fueling the brain and for functional imaging. However, little is known about how the brain regulates ongoing oscillations in cerebral blood flow. In mouse and rat cortical brain slice arterioles, we find that selectively enhancing tone is sufficient to recruit a TRPV4-mediated Ca2+ elevation in adjacent astrocyte endfeet. This endfoot Ca2+ signal triggers COX-1-mediated "feedback vasodilators" that limit the extent of evoked vasoconstriction, as well as constrain fictive vasomotion in slices. Astrocyte-Ptgs1 knockdown in vivo increases the power of arteriole oscillations across a broad range of very low frequencies (0.01-0.3 Hz), including ultra-slow vasomotion (∼0.1 Hz). Conversely, clamping astrocyte Ca2+in vivo reduces the power of vasomotion. These data demonstrate bidirectional communication between arterioles and astrocyte endfeet to regulate oscillatory microvasculature activity.
Collapse
Affiliation(s)
- Jordan N Haidey
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Govind Peringod
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kelsea A Gorzo
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Wilten Nicola
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Physics and Astronomy, Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Milène Vandal
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kenichi Ito
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shiying Liu
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cameron Fielding
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Frank Visser
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
29
|
Sherwood MW, Arizono M, Panatier A, Mikoshiba K, Oliet SHR. Astrocytic IP 3Rs: Beyond IP 3R2. Front Cell Neurosci 2021; 15:695817. [PMID: 34393726 PMCID: PMC8363081 DOI: 10.3389/fncel.2021.695817] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are sensitive to ongoing neuronal/network activities and, accordingly, regulate neuronal functions (synaptic transmission, synaptic plasticity, behavior, etc.) by the context-dependent release of several gliotransmitters (e.g., glutamate, glycine, D-serine, ATP). To sense diverse input, astrocytes express a plethora of G-protein coupled receptors, which couple, via Gi/o and Gq, to the intracellular Ca2+ release channel IP3-receptor (IP3R). Indeed, manipulating astrocytic IP3R-Ca2+ signaling is highly consequential at the network and behavioral level: Depleting IP3R subtype 2 (IP3R2) results in reduced GPCR-Ca2+ signaling and impaired synaptic plasticity; enhancing IP3R-Ca2+ signaling affects cognitive functions such as learning and memory, sleep, and mood. However, as a result of discrepancies in the literature, the role of GPCR-IP3R-Ca2+ signaling, especially under physiological conditions, remains inconclusive. One primary reason for this could be that IP3R2 has been used to represent all astrocytic IP3Rs, including IP3R1 and IP3R3. Indeed, IP3R1 and IP3R3 are unique Ca2+ channels in their own right; they have unique biophysical properties, often display distinct distribution, and are differentially regulated. As a result, they mediate different physiological roles to IP3R2. Thus, these additional channels promise to enrich the diversity of spatiotemporal Ca2+ dynamics and provide unique opportunities for integrating neuronal input and modulating astrocyte–neuron communication. The current review weighs evidence supporting the existence of multiple astrocytic-IP3R isoforms, summarizes distinct sub-type specific properties that shape spatiotemporal Ca2+ dynamics. We also discuss existing experimental tools and future refinements to better recapitulate the endogenous activities of each IP3R isoform.
Collapse
Affiliation(s)
- Mark W Sherwood
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Misa Arizono
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Aude Panatier
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Katsuhiko Mikoshiba
- ShanghaiTech University, Shanghai, China.,Faculty of Science, Toho University, Funabashi, Japan.,RIKEN CLST, Kobe, Japan
| | - Stéphane H R Oliet
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| |
Collapse
|
30
|
Vanacker C, Defazio RA, Sykes CM, Moenter SM. A role for glial fibrillary acidic protein (GFAP)-expressing cells in the regulation of gonadotropin-releasing hormone (GnRH) but not arcuate kisspeptin neuron output in male mice. eLife 2021; 10:68205. [PMID: 34292152 PMCID: PMC8337074 DOI: 10.7554/elife.68205] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/21/2021] [Indexed: 01/05/2023] Open
Abstract
GnRH neurons are the final central neural output regulating fertility. Kisspeptin neurons in the hypothalamic arcuate nucleus (KNDy neurons) are considered the main regulator of GnRH output. GnRH and KNDy neurons are surrounded by astrocytes, which can modulate neuronal activity and communicate over distances. Prostaglandin E2 (PGE2), synthesized primarily by astrocytes, increases GnRH neuron activity and downstream pituitary release of luteinizing hormone (LH). We hypothesized that glial fibrillary acidic protein (GFAP)-expressing astrocytes play a role in regulating GnRH and/or KNDy neuron activity and LH release. We used adeno-associated viruses to target designer receptors exclusively activated by designer drugs (DREADDs) to GFAP-expressing cells to activate Gq- or Gi-mediated signaling. Activating Gq signaling in the preoptic area, near GnRH neurons, but not in the arcuate, increases LH release in vivo and GnRH firing in vitro via a mechanism in part dependent upon PGE2. These data suggest that astrocytes can activate GnRH/LH release in a manner independent of KNDy neurons.
Collapse
Affiliation(s)
- Charlotte Vanacker
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - R Anthony Defazio
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Charlene M Sykes
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States.,Internal Medicine, University of Michigan, Ann Arbor, United States.,Obstetrics & Gynecology, University of Michigan, Ann Arbor, United States.,Reproductive Sciences Program, University of Michigan, Ann Arbor, United States
| |
Collapse
|
31
|
Stackhouse TL, Mishra A. Neurovascular Coupling in Development and Disease: Focus on Astrocytes. Front Cell Dev Biol 2021; 9:702832. [PMID: 34327206 PMCID: PMC8313501 DOI: 10.3389/fcell.2021.702832] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Neurovascular coupling is a crucial mechanism that matches the high energy demand of the brain with a supply of energy substrates from the blood. Signaling within the neurovascular unit is responsible for activity-dependent changes in cerebral blood flow. The strength and reliability of neurovascular coupling form the basis of non-invasive human neuroimaging techniques, including blood oxygen level dependent (BOLD) functional magnetic resonance imaging. Interestingly, BOLD signals are negative in infants, indicating a mismatch between metabolism and blood flow upon neural activation; this response is the opposite of that observed in healthy adults where activity evokes a large oversupply of blood flow. Negative neurovascular coupling has also been observed in rodents at early postnatal stages, further implying that this is a process that matures during development. This rationale is consistent with the morphological maturation of the neurovascular unit, which occurs over a similar time frame. While neurons differentiate before birth, astrocytes differentiate postnatally in rodents and the maturation of their complex morphology during the first few weeks of life links them with synapses and the vasculature. The vascular network is also incomplete in neonates and matures in parallel with astrocytes. Here, we review the timeline of the structural maturation of the neurovascular unit with special emphasis on astrocytes and the vascular tree and what it implies for functional maturation of neurovascular coupling. We also discuss similarities between immature astrocytes during development and reactive astrocytes in disease, which are relevant to neurovascular coupling. Finally, we close by pointing out current gaps in knowledge that must be addressed to fully elucidate the mechanisms underlying neurovascular coupling maturation, with the expectation that this may also clarify astrocyte-dependent mechanisms of cerebrovascular impairment in neurodegenerative conditions in which reduced or negative neurovascular coupling is noted, such as stroke and Alzheimer’s disease.
Collapse
Affiliation(s)
- Teresa L Stackhouse
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States.,Knight Cardiovascular Institute, Oregon Health & Sciences University, Portland, OR, United States
| |
Collapse
|
32
|
Disruption of the astrocyte-neuron interaction is responsible for the impairments in learning and memory in 5XFAD mice: an Alzheimer's disease animal model. Mol Brain 2021; 14:111. [PMID: 34246283 PMCID: PMC8272251 DOI: 10.1186/s13041-021-00823-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/05/2021] [Indexed: 12/02/2022] Open
Abstract
The morphological dynamics of astrocytes are altered in the hippocampus during memory induction. Astrocyte–neuron interactions on synapses are called tripartite synapses. These control the synaptic function in the central nervous system. Astrocytes are activated in a reactive state by STAT3 phosphorylation in 5XFAD mice, an Alzheimer’s disease (AD) animal model. However, changes in astrocyte–neuron interactions in reactive or resting-state astrocytes during memory induction remain to be defined. Here, we investigated the time-dependent changes in astrocyte morphology and the number of astrocyte–neuron interactions in the hippocampus over the course of long-term memory formation in 5XFAD mice. Hippocampal-dependent long-term memory was induced using a contextual fear conditioning test in 5XFAD mice. The number of astrocytic processes increased in both wild-type and 5XFAD mice during memory formation. To assess astrocyte–neuron interactions in the hippocampal dentate gyrus, we counted the colocalization of glial fibrillary acidic protein and postsynaptic density protein 95 via immunofluorescence. Both groups revealed an increase in astrocyte–neuron interactions after memory induction. At 24 h after memory formation, the number of tripartite synapses returned to baseline levels in both groups. However, the total number of astrocyte–neuron interactions was significantly decreased in 5XFAD mice. Administration of Stattic, a STAT3 phosphorylation inhibitor, rescued the number of astrocyte–neuron interactions in 5XFAD mice. In conclusion, we suggest that a decreased number of astrocyte–neuron interactions may underlie memory impairment in the early stages of AD.
Collapse
|
33
|
Kumar BS, Khot A, Chakravarthy VS, Pushpavanam S. A Network Architecture for Bidirectional Neurovascular Coupling in Rat Whisker Barrel Cortex. Front Comput Neurosci 2021; 15:638700. [PMID: 34211384 PMCID: PMC8241226 DOI: 10.3389/fncom.2021.638700] [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: 12/22/2020] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Neurovascular coupling is typically considered as a master-slave relationship between the neurons and the cerebral vessels: the neurons demand energy which the vessels supply in the form of glucose and oxygen. In the recent past, both theoretical and experimental studies have suggested that the neurovascular coupling is a bidirectional system, a loop that includes a feedback signal from the vessels influencing neural firing and plasticity. An integrated model of bidirectionally connected neural network and the vascular network is hence required to understand the relationship between the informational and metabolic aspects of neural dynamics. In this study, we present a computational model of the bidirectional neurovascular system in the whisker barrel cortex and study the effect of such coupling on neural activity and plasticity as manifest in the whisker barrel map formation. In this model, a biologically plausible self-organizing network model of rate coded, dynamic neurons is nourished by a network of vessels modeled using the biophysical properties of blood vessels. The neural layer which is designed to simulate the whisker barrel cortex of rat transmits vasodilatory signals to the vessels. The feedback from the vessels is in the form of available oxygen for oxidative metabolism whose end result is the adenosine triphosphate (ATP) necessary to fuel neural firing. The model captures the effect of the feedback from the vascular network on the neuronal map formation in the whisker barrel model under normal and pathological (Hypoxia and Hypoxia-Ischemia) conditions.
Collapse
Affiliation(s)
- Bhadra S. Kumar
- Computational Neuroscience Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Aditi Khot
- Department of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - V. Srinivasa Chakravarthy
- Computational Neuroscience Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Pushpavanam
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
34
|
Lim EY, Ye L, Paukert M. Potential and Realized Impact of Astroglia Ca 2 + Dynamics on Circuit Function and Behavior. Front Cell Neurosci 2021; 15:682888. [PMID: 34163330 PMCID: PMC8215280 DOI: 10.3389/fncel.2021.682888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Astroglia display a wide range of spontaneous and behavioral state-dependent Ca2+ dynamics. During heightened vigilance, noradrenergic signaling leads to quasi-synchronous Ca2+ elevations encompassing soma and processes across the brain-wide astroglia network. Distinct from this vigilance-associated global Ca2+ rise are apparently spontaneous fluctuations within spatially restricted microdomains. Over the years, several strategies have been pursued to shed light on the physiological impact of these signals including deletion of endogenous ion channels or receptors and reduction of intracellular Ca2+ through buffering, extrusion or inhibition of release. Some experiments that revealed the most compelling behavioral alterations employed chemogenetic and optogenetic manipulations to modify astroglia Ca2+ signaling. However, there is considerable contrast between these findings and the comparatively modest effects of inhibiting endogenous sources of Ca2+. In this review, we describe the underlying mechanisms of various forms of astroglia Ca2+ signaling as well as the functional consequences of their inhibition. We then discuss how the effects of exogenous astroglia Ca2+ modification combined with our knowledge of physiological mechanisms of astroglia Ca2+ activation could guide further refinement of behavioral paradigms that will help elucidate the natural Ca2+-dependent function of astroglia.
Collapse
Affiliation(s)
- Eunice Y. Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,*Correspondence: Martin Paukert,
| |
Collapse
|
35
|
Broadhead MJ, Miles GB. A common role for astrocytes in rhythmic behaviours? Prog Neurobiol 2021; 202:102052. [PMID: 33894330 DOI: 10.1016/j.pneurobio.2021.102052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/03/2021] [Accepted: 04/13/2021] [Indexed: 01/16/2023]
Abstract
Astrocytes are a functionally diverse form of glial cell involved in various aspects of nervous system infrastructure, from the metabolic and structural support of neurons to direct neuromodulation of synaptic activity. Investigating how astrocytes behave in functionally related circuits may help us understand whether there is any conserved logic to the role of astrocytes within neuronal networks. Astrocytes are implicated as key neuromodulatory cells within neural circuits that control a number of rhythmic behaviours such as breathing, locomotion and circadian sleep-wake cycles. In this review, we examine the evidence that astrocytes are directly involved in the regulation of the neural circuits underlying six different rhythmic behaviours: locomotion, breathing, chewing, gastrointestinal motility, circadian sleep-wake cycles and oscillatory feeding behaviour. We discuss how astrocytes are integrated into the neuronal networks that regulate these behaviours, and identify the potential gliotransmission signalling mechanisms involved. From reviewing the evidence of astrocytic involvement in a range of rhythmic behaviours, we reveal a heterogenous array of gliotransmission mechanisms, which help to regulate neuronal networks. However, we also observe an intriguing thread of commonality, in the form of purinergic gliotransmission, which is frequently utilised to facilitate feedback inhibition within rhythmic networks to constrain a given behaviour within its operational range.
Collapse
Affiliation(s)
- Matthew J Broadhead
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK.
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| |
Collapse
|
36
|
Nuzzaci D, Cansell C, Liénard F, Nédélec E, Ben Fradj S, Castel J, Foppen E, Denis R, Grouselle D, Laderrière A, Lemoine A, Mathou A, Tolle V, Heurtaux T, Fioramonti X, Audinat E, Pénicaud L, Nahon JL, Rovère C, Benani A. Postprandial Hyperglycemia Stimulates Neuroglial Plasticity in Hypothalamic POMC Neurons after a Balanced Meal. Cell Rep 2021; 30:3067-3078.e5. [PMID: 32130907 DOI: 10.1016/j.celrep.2020.02.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 12/17/2019] [Accepted: 02/06/2020] [Indexed: 12/31/2022] Open
Abstract
Mechanistic studies in rodents evidenced synaptic remodeling in neuronal circuits that control food intake. However, the physiological relevance of this process is not well defined. Here, we show that the firing activity of anorexigenic POMC neurons located in the hypothalamus is increased after a standard meal. Postprandial hyperactivity of POMC neurons relies on synaptic plasticity that engages pre-synaptic mechanisms, which does not involve structural remodeling of synapses but retraction of glial coverage. These functional and morphological neuroglial changes are triggered by postprandial hyperglycemia. Chemogenetically induced glial retraction on POMC neurons is sufficient to increase POMC activity and modify meal patterns. These findings indicate that synaptic plasticity within the melanocortin system happens at the timescale of meals and likely contributes to short-term control of food intake. Interestingly, these effects are lost with a high-fat meal, suggesting that neuroglial plasticity of POMC neurons is involved in the satietogenic properties of foods.
Collapse
Affiliation(s)
- Danaé Nuzzaci
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Céline Cansell
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Fabienne Liénard
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Emmanuelle Nédélec
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Selma Ben Fradj
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Julien Castel
- Unité "Biologie Fonctionnelle & Adaptative," CNRS, Université Paris Diderot, 75005 Paris, France
| | - Ewout Foppen
- Unité "Biologie Fonctionnelle & Adaptative," CNRS, Université Paris Diderot, 75005 Paris, France
| | - Raphael Denis
- Unité "Biologie Fonctionnelle & Adaptative," CNRS, Université Paris Diderot, 75005 Paris, France
| | - Dominique Grouselle
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014 Paris, France
| | - Amélie Laderrière
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Aleth Lemoine
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Alexia Mathou
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Virginie Tolle
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014 Paris, France
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology, Department of Life Sciences and Medicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Xavier Fioramonti
- Laboratoire NutriNeuro, INRA, Université de Bordeaux, 33076 Bordeaux, France
| | - Etienne Audinat
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Luc Pénicaud
- StromaLab, CNRS, EFS, INP-ENVT, INSERM, Université Paul Sabatier, 31100 Toulouse, France
| | - Jean-Louis Nahon
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Carole Rovère
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France.
| |
Collapse
|
37
|
Vaidyanathan TV, Collard M, Yokoyama S, Reitman ME, Poskanzer KE. Cortical astrocytes independently regulate sleep depth and duration via separate GPCR pathways. eLife 2021; 10:63329. [PMID: 33729913 PMCID: PMC7968927 DOI: 10.7554/elife.63329] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Non-rapid eye movement (NREM) sleep, characterized by slow-wave electrophysiological activity, underlies several critical functions, including learning and memory. However, NREM sleep is heterogeneous, varying in duration, depth, and spatially across the cortex. While these NREM sleep features are thought to be largely independently regulated, there is also evidence that they are mechanistically coupled. To investigate how cortical NREM sleep features are controlled, we examined the astrocytic network, comprising a cortex-wide syncytium that influences population-level neuronal activity. We quantified endogenous astrocyte activity in mice over natural sleep and wake, then manipulated specific astrocytic G-protein-coupled receptor (GPCR) signaling pathways in vivo. We find that astrocytic Gi- and Gq-coupled GPCR signaling separately control NREM sleep depth and duration, respectively, and that astrocytic signaling causes differential changes in local and remote cortex. These data support a model in which the cortical astrocyte network serves as a hub for regulating distinct NREM sleep features. Sleep has many roles, from strengthening new memories to regulating mood and appetite. While we might instinctively think of sleep as a uniform state of reduced brain activity, the reality is more complex. First, over the course of the night, we cycle between a number of different sleep stages, which reflect different levels of sleep depth. Second, the amount of sleep depth is not necessarily even across the brain but can vary between regions. These sleep stages consist of either rapid eye movement (REM) sleep or non-REM (NREM) sleep. REM sleep is when most dreaming occurs, whereas NREM sleep is particularly important for learning and memory and can vary in duration and depth. During NREM sleep, large groups of neurons synchronize their firing to create rhythmic waves of activity known as slow waves. The more synchronous the activity, the deeper the sleep. Vaidyanathan et al. now show that brain cells called astrocytes help regulate NREM sleep. Astrocytes are not neurons but belong to a group of specialized cells called glia. They are the largest glia cell type in the brain and display an array of proteins on their surfaces called G-protein-coupled receptors (GPCRs). These enable them to sense sleep-wake signals from other parts of the brain and to generate their own signals. In fact, each astrocyte can communicate with thousands of neurons at once. They are therefore well-poised to coordinate brain activity during NREM sleep. Using innovative tools, Vaidyanathan et al. visualized astrocyte activity in mice as the animals woke up or fell asleep. The results showed that astrocytes change their activity just before each sleep–wake transition. They also revealed that astrocytes control both the depth and duration of NREM sleep via two different types of GPCR signals. Increasing one of these signals (Gi-GPCR) made the mice sleep more deeply but did not change sleep duration. Decreasing the other (Gq-GPCR) made the mice sleep for longer but did not affect sleep depth. Sleep problems affect many people at some point in their lives, and often co-exist with other conditions such as mental health disorders. Understanding how the brain regulates different features of sleep could help us develop better – and perhaps more specific – treatments for sleep disorders. The current study suggests that manipulating GPCRs on astrocytes might increase sleep depth, for example. But before work to test this idea can begin, we must first determine whether findings from sleeping mice also apply to people.
Collapse
Affiliation(s)
- Trisha V Vaidyanathan
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Max Collard
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Sae Yokoyama
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Michael E Reitman
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Kira E Poskanzer
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, San Francisco, United States
| |
Collapse
|
38
|
Moshkforoush A, Balachandar L, Moncion C, Montejo KA, Riera J. Unraveling ChR2-driven stochastic Ca2+ dynamics in astrocytes: A call for new interventional paradigms. PLoS Comput Biol 2021; 17:e1008648. [PMID: 33566841 PMCID: PMC7875401 DOI: 10.1371/journal.pcbi.1008648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023] Open
Abstract
Optogenetic targeting of astrocytes provides a robust experimental model to differentially induce Ca2+ signals in astrocytes in vivo. However, a systematic study quantifying the response of optogenetically modified astrocytes to light is yet to be performed. Here, we propose a novel stochastic model of Ca2+ dynamics in astrocytes that incorporates a light sensitive component-channelrhodopsin 2 (ChR2). Utilizing this model, we investigated the effect of different light stimulation paradigms on cells expressing select variants of ChR2 (wild type, ChETA, and ChRET/TC). Results predict that depending on paradigm specification, astrocytes might undergo drastic changes in their basal Ca2+ level and spiking probability. Furthermore, we performed a global sensitivity analysis to assess the effect of variation in parameters pertinent to the shape of the ChR2 photocurrent on astrocytic Ca2+ dynamics. Results suggest that directing variants towards the first open state of the ChR2 photocycle (o1) enhances spiking activity in astrocytes during optical stimulation. Evaluation of the effect of Ca2+ buffering and coupling coefficient in a network of ChR2-expressing astrocytes demonstrated basal level elevations in the stimulated region and propagation of calcium activity to unstimulated cells. Buffering reduced the diffusion range of Ca2+ within the network, thereby limiting propagation and influencing the activity of astrocytes. Collectively, the framework presented in this study provides valuable information for the selection of light stimulation paradigms that elicit desired astrocytic activity using existing ChR2 constructs, as well as aids in the engineering of future application-oriented optogenetic variants.
Collapse
Affiliation(s)
- Arash Moshkforoush
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Lakshmini Balachandar
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Carolina Moncion
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Karla A. Montejo
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jorge Riera
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
39
|
Institoris A, Murphy-Royal C, Tarantini S, Yabluchanskiy A, Haidey JN, Csiszar A, Ungvari Z, Gordon GR. Whole brain irradiation in mice causes long-term impairment in astrocytic calcium signaling but preserves astrocyte-astrocyte coupling. GeroScience 2021; 43:197-212. [PMID: 33094399 PMCID: PMC8050172 DOI: 10.1007/s11357-020-00289-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/14/2020] [Indexed: 11/29/2022] Open
Abstract
Whole brain irradiation (WBI) therapy is an important treatment for brain metastases and potential microscopic malignancies. WBI promotes progressive cognitive dysfunction in over half of surviving patients, yet, the underlying mechanisms remain obscure. Astrocytes play critical roles in the regulation of neuronal activity, brain metabolism, and cerebral blood flow, and while neurons are considered radioresistant, astrocytes are sensitive to γ-irradiation. Hallmarks of astrocyte function are the ability to generate stimulus-induced intercellular Ca2+ signals and to move metabolic substrates through the connected astrocyte network. We tested the hypothesis that WBI-induced cognitive impairment associates with persistent impairment of astrocytic Ca2+ signaling and/or gap junctional coupling. Mice were subjected to a clinically relevant protocol of fractionated WBI, and 12 to 15 months after irradiation, we confirmed persistent cognitive impairment compared to controls. To test the integrity of astrocyte-to-astrocyte gap junctional coupling postWBI, astrocytes were loaded with Alexa-488-hydrazide by patch-based dye infusion, and the increase of fluorescence signal in neighboring astrocyte cell bodies was assessed with 2-photon microscopy in acute slices of the sensory-motor cortex. We found that WBI did not affect astrocyte-to-astrocyte gap junctional coupling. Astrocytic Ca2+ responses induced by bath administration of phenylephrine (detected with Rhod-2/AM) were also unaltered by WBI. However, an electrical stimulation protocol used in long-term potentiation (theta burst), revealed attenuated astrocyte Ca2+ responses in the astrocyte arbor and soma in WBI. Our data show that WBI causes a long-lasting decrement in synaptic-evoked astrocyte Ca2+ signals 12-15 months postirradiation, which may be an important contributor to cognitive decline seen after WBI.
Collapse
Affiliation(s)
- Adam Institoris
- Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ciaran Murphy-Royal
- Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Stefano Tarantini
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jordan N Haidey
- Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Anna Csiszar
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Grant R Gordon
- Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
40
|
Howarth C, Mishra A, Hall CN. More than just summed neuronal activity: how multiple cell types shape the BOLD response. Philos Trans R Soc Lond B Biol Sci 2021; 376:20190630. [PMID: 33190598 PMCID: PMC7116385 DOI: 10.1098/rstb.2019.0630] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Functional neuroimaging techniques are widely applied to investigations of human cognition and disease. The most commonly used among these is blood oxygen level-dependent (BOLD) functional magnetic resonance imaging. The BOLD signal occurs because neural activity induces an increase in local blood supply to support the increased metabolism that occurs during activity. This supply usually outmatches demand, resulting in an increase in oxygenated blood in an active brain region, and a corresponding decrease in deoxygenated blood, which generates the BOLD signal. Hence, the BOLD response is shaped by an integration of local oxygen use, through metabolism, and supply, in the blood. To understand what information is carried in BOLD signals, we must understand how several cell types in the brain-local excitatory neurons, inhibitory neurons, astrocytes and vascular cells (pericytes, vascular smooth muscle and endothelial cells), and their modulation by ascending projection neurons-contribute to both metabolism and haemodynamic changes. Here, we review the contributions of each cell type to the regulation of cerebral blood flow and metabolism, and discuss situations where a simplified interpretation of the BOLD response as reporting local excitatory activity may misrepresent important biological phenomena, for example with regards to arousal states, ageing and neurological disease. This article is part of the theme issue 'Key relationships between non-invasive functional neuroimaging and the underlying neuronal activity'.
Collapse
Affiliation(s)
- Clare Howarth
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|
41
|
Sharma K, Gordon GRJ, Tran CHT. Heterogeneity of Sensory-Induced Astrocytic Ca 2+ Dynamics During Functional Hyperemia. Front Physiol 2020; 11:611884. [PMID: 33362585 PMCID: PMC7758506 DOI: 10.3389/fphys.2020.611884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022] Open
Abstract
Astrocytic Ca2+ fluctuations associated with functional hyperemia have typically been measured from large cellular compartments such as the soma, the whole arbor and the endfoot. The most prominent Ca2+ event is a large magnitude, delayed signal that follows vasodilation. However, previous work has provided little information about the spatio-temporal properties of such Ca2+ transients or their heterogeneity. Here, using an awake, in vivo two-photon fluorescence-imaging model, we performed detailed profiling of delayed astrocytic Ca2+ signals across astrocytes or within individual astrocyte compartments using small regions of interest next to penetrating arterioles and capillaries along with vasomotor responses to vibrissae stimulation. We demonstrated that while a 5-s air puff that stimulates all whiskers predominantly generated reproducible functional hyperemia in the presence or absence of astrocytic Ca2+ changes, whisker stimulation inconsistently produced astrocytic Ca2+ responses. More importantly, these Ca2+ responses were heterogeneous among subcellular structures of the astrocyte and across different astrocytes that resided within the same field of view. Furthermore, we found that whisker stimulation induced discrete Ca2+ “hot spots” that spread regionally within the endfoot. These data reveal that astrocytic Ca2+ dynamics associated with the microvasculature are more complex than previously thought, and highlight the importance of considering the heterogeneity of astrocytic Ca2+ activity to fully understanding neurovascular coupling.
Collapse
Affiliation(s)
- Kushal Sharma
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Grant R J Gordon
- Department of Physiology and Pharmacology, School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Cam Ha T Tran
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| |
Collapse
|
42
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
43
|
Echagarruga CT, Gheres KW, Norwood JN, Drew PJ. nNOS-expressing interneurons control basal and behaviorally evoked arterial dilation in somatosensory cortex of mice. eLife 2020; 9:e60533. [PMID: 33016877 PMCID: PMC7556878 DOI: 10.7554/elife.60533] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Cortical neural activity is coupled to local arterial diameter and blood flow. However, which neurons control the dynamics of cerebral arteries is not well understood. We dissected the cellular mechanisms controlling the basal diameter and evoked dilation in cortical arteries in awake, head-fixed mice. Locomotion drove robust arterial dilation, increases in gamma band power in the local field potential (LFP), and increases calcium signals in pyramidal and neuronal nitric oxide synthase (nNOS)-expressing neurons. Chemogenetic or pharmocological modulation of overall neural activity up or down caused corresponding increases or decreases in basal arterial diameter. Modulation of pyramidal neuron activity alone had little effect on basal or evoked arterial dilation, despite pronounced changes in the LFP. Modulation of the activity of nNOS-expressing neurons drove changes in the basal and evoked arterial diameter without corresponding changes in population neural activity.
Collapse
Affiliation(s)
| | - Kyle W Gheres
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Jordan N Norwood
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Patrick J Drew
- Bioengineering Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Departments of Engineering Science and Mechanics, Biomedical Engineering, and Neurosurgery, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
44
|
Okubo Y. Astrocytic Ca2+ signaling mediated by the endoplasmic reticulum in health and disease. J Pharmacol Sci 2020; 144:83-88. [DOI: 10.1016/j.jphs.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
|
45
|
Presa JL, Saravia F, Bagi Z, Filosa JA. Vasculo-Neuronal Coupling and Neurovascular Coupling at the Neurovascular Unit: Impact of Hypertension. Front Physiol 2020; 11:584135. [PMID: 33101063 PMCID: PMC7546852 DOI: 10.3389/fphys.2020.584135] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Components of the neurovascular unit (NVU) establish dynamic crosstalk that regulates cerebral blood flow and maintain brain homeostasis. Here, we describe accumulating evidence for cellular elements of the NVU contributing to critical physiological processes such as cerebral autoregulation, neurovascular coupling, and vasculo-neuronal coupling. We discuss how alterations in the cellular mechanisms governing NVU homeostasis can lead to pathological changes in which vascular endothelial and smooth muscle cell, pericyte and astrocyte function may play a key role. Because hypertension is a modifiable risk factor for stroke and accelerated cognitive decline in aging, we focus on hypertension-associated changes on cerebral arteriole function and structure, and the molecular mechanisms through which these may contribute to cognitive decline. We gather recent emerging evidence concerning cognitive loss in hypertension and the link with vascular dementia and Alzheimer’s disease. Collectively, we summarize how vascular dysfunction, chronic hypoperfusion, oxidative stress, and inflammatory processes can uncouple communication at the NVU impairing cerebral perfusion and contributing to neurodegeneration.
Collapse
Affiliation(s)
- Jessica L Presa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
46
|
Verkhratsky A, Semyanov A, Zorec R. Physiology of Astroglial Excitability. FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa016. [PMID: 35330636 PMCID: PMC8788756 DOI: 10.1093/function/zqaa016] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023]
Abstract
Classic physiology divides all neural cells into excitable neurons and nonexcitable neuroglia. Neuroglial cells, chiefly responsible for homeostasis and defense of the nervous tissue, coordinate their complex homeostatic responses with neuronal activity. This coordination reflects a specific form of glial excitability mediated by complex changes in intracellular concentration of ions and second messengers organized in both space and time. Astrocytes are equipped with multiple molecular cascades, which are central for regulating homeostasis of neurotransmitters, ionostasis, synaptic connectivity, and metabolic support of the central nervous system. Astrocytes are further provisioned with multiple receptors for neurotransmitters and neurohormones, which upon activation trigger intracellular signals mediated by Ca2+, Na+, and cyclic AMP. Calcium signals have distinct organization and underlying mechanisms in different astrocytic compartments thus allowing complex spatiotemporal signaling. Signals mediated by fluctuations in cytosolic Na+ are instrumental for coordination of Na+ dependent astrocytic transporters with tissue state and homeostatic demands. Astroglial ionic excitability may also involve K+, H+, and Cl-. The cyclic AMP signalling system is, in comparison to ions, much slower in targeting astroglial effector mechanisms. This evidence review summarizes the concept of astroglial intracellular excitability.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK,Achucarro Center for Neuroscience, Ikerbasque, 48011 Bilbao, Spain,Address correspondence to A.V. (e-mail: )
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia,Faculty of Biology, Moscow State University, Moscow, Russia,Sechenov First Moscow State Medical University, Moscow, Russia
| | - Robert Zorec
- Celica Biomedical, Ljubljana 1000, Slovenia,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| |
Collapse
|
47
|
Semyanov A, Henneberger C, Agarwal A. Making sense of astrocytic calcium signals — from acquisition to interpretation. Nat Rev Neurosci 2020; 21:551-564. [DOI: 10.1038/s41583-020-0361-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
48
|
Xie AX, Madayag A, Minton SK, McCarthy KD, Malykhina AP. Sensory satellite glial Gq-GPCR activation alleviates inflammatory pain via peripheral adenosine 1 receptor activation. Sci Rep 2020; 10:14181. [PMID: 32843670 PMCID: PMC7447794 DOI: 10.1038/s41598-020-71073-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Glial fibrillary acidic protein expressing (GFAP+) glia modulate nociceptive neuronal activity in both the peripheral nervous system (PNS) and the central nervous system (CNS). Resident GFAP+ glia in dorsal root ganglia (DRG) known as satellite glial cells (SGCs) potentiate neuronal activity by releasing pro-inflammatory cytokines and neuroactive compounds. In this study, we tested the hypothesis that SGC Gq-coupled receptor (Gq-GPCR) signaling modulates pain sensitivity in vivo using Gfap-hM3Dq mice. Complete Freund's adjuvant (CFA) was used to induce inflammatory pain, and mechanical sensitivity and thermal sensitivity were used to assess the neuromodulatory effect of glial Gq-GPCR activation in awake mice. Pharmacogenetic activation of Gq-GPCR signaling in sensory SGCs decreased heat-induced nociceptive responses and reversed inflammation-induced mechanical allodynia via peripheral adenosine A1 receptor activation. These data reveal a previously unexplored role of sensory SGCs in decreasing afferent excitability. The identified molecular mechanism underlying the analgesic role of SGCs offers new approaches for reversing peripheral nociceptive sensitization.
Collapse
MESH Headings
- Animals
- Benzilates/pharmacology
- Clozapine/analogs & derivatives
- Clozapine/pharmacology
- Freund's Adjuvant/toxicity
- GTP-Binding Protein alpha Subunits, Gq-G11/physiology
- Genes, Synthetic
- Hot Temperature
- Hyperalgesia/physiopathology
- Hyperalgesia/prevention & control
- Inflammation/chemically induced
- Inflammation/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscarinic Agonists/pharmacology
- Neuroglia/enzymology
- Neuroglia/physiology
- Nociception/physiology
- Nortropanes/pharmacology
- Promoter Regions, Genetic
- Purinergic P1 Receptor Agonists/pharmacology
- Purinergic P1 Receptor Antagonists/pharmacology
- Receptor, Adenosine A1/drug effects
- Receptor, Adenosine A1/physiology
- Receptor, Muscarinic M3/drug effects
- Receptor, Muscarinic M3/genetics
- Receptor, Muscarinic M3/physiology
- Receptors, G-Protein-Coupled
- Recombinant Fusion Proteins/drug effects
- Recombinant Fusion Proteins/metabolism
- Theophylline/analogs & derivatives
- Theophylline/pharmacology
- Touch
- Xanthines/pharmacology
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, USA.
- Division of Urology, Department of Surgery, University of Colorado Denver (UCD), Anschutz Medical Campus (AMC), 12700E 19th Ave., Room 6440D, Mail stop C317, Aurora, CO, 80045, USA.
- Department of Surgery, UCD-AMC, Aurora, CO, USA.
| | - Aric Madayag
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, USA
- NeuroCycle Therapeutics, Inc., 3829 N Cramer St., Shorewood, WI, 53211, USA
| | - Suzanne K Minton
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, USA
- Certara, 5511 Capital Center Drive, Ste. 204, Raleigh, NC, 27606, USA
| | - Ken D McCarthy
- Professor Emeritus in the Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, 120 Mason Farm Road, 4010 Genetic Medicine Bldg, Campus Box 7365, Chapel Hill, NC, 27599-7365, USA
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver (UCD), Anschutz Medical Campus (AMC), 12700E 19th Ave., Room 6440D, Mail stop C317, Aurora, CO, 80045, USA
- Department of Physiology and Biophysics, University of Colorado School of Medicine, 12700 East 19th Ave., Rm 6001, Mail Stop C317, Aurora, CO, 80045, USA
| |
Collapse
|
49
|
Guillot de Suduiraut I, Grosse J, Ramos-Fernández E, Sandi C, Hollis F. Astrocytic release of ATP through type 2 inositol 1,4,5-trisphosphate receptor calcium signaling and social dominance behavior in mice. Eur J Neurosci 2020; 53:2973-2985. [PMID: 32609904 DOI: 10.1111/ejn.14892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/22/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023]
Abstract
Brain mitochondrial function is critical for numerous neuronal processes. We recently identified a link between brain energy and social dominance, where higher levels of mitochondrial function resulted in increased social competitive ability. The underlying mechanism of this link, however, remains unclear. Here, we investigated the contribution of astrocytic release of adenosine triphosphate (ATP) through the type 2 inositol 1,4,5-triphosphate receptor to social dominance behavior. Mice lacking the type 2 inositol 1,4,5-triphosphate receptor were characterized for their social dominance behavior, as well as their performance on a nonsocial task, the Morris Water Maze. In parallel, we also examined mitochondrial function in the medial prefrontal cortex, nucleus accumbens, and hippocampus to investigate how deficiencies in astrocytic ATP could modulate overall mitochondrial function. While knockout mice showed similar competitive ability compared with their wild-type littermates, dominant knockout mice exhibited a significant delay in exerting their dominance during the initial encounter. Otherwise, there were no differences in anxiety and exploratory traits, spatial learning and memory, or brain mitochondrial function in either light or dark circadian phases. Our findings point to a marginal role of astrocytic ATP through IP3 R2 in social competition, suggesting that, under basal conditions, the neuronal compartment is predominant for social dominance exertion.
Collapse
Affiliation(s)
| | - Jocelyn Grosse
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eva Ramos-Fernández
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fiona Hollis
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia
| |
Collapse
|
50
|
Sten S, Elinder F, Cedersund G, Engström M. A quantitative analysis of cell-specific contributions and the role of anesthetics to the neurovascular coupling. Neuroimage 2020; 215:116827. [PMID: 32289456 DOI: 10.1016/j.neuroimage.2020.116827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/26/2020] [Indexed: 11/18/2022] Open
Abstract
The neurovascular coupling (NVC) connects neuronal activity to hemodynamic responses in the brain. This connection is the basis for the interpretation of functional magnetic resonance imaging data. Despite the central role of this coupling, we lack detailed knowledge about cell-specific contributions and our knowledge about NVC is mainly based on animal experiments performed during anesthesia. Anesthetics are known to affect neuronal excitability, but how this affects the vessel diameters is not known. Due to the high complexity of NVC data, mathematical modeling is needed for a meaningful analysis. However, neither the relevant neuronal subtypes nor the effects of anesthetics are covered by current models. Here, we present a mathematical model including GABAergic interneurons and pyramidal neurons, as well as the effect of an anesthetic agent. The model is consistent with data from optogenetic experiments from both awake and anesthetized animals, and it correctly predicts data from experiments with different pharmacological modulators. The analysis suggests that no downstream anesthetic effects are necessary if one of the GABAergic interneuron signaling pathways include a Michaelis-Menten expression. This is the first example of a quantitative model that includes both the cell-specific contributions and the effect of an anesthetic agent on the NVC.
Collapse
Affiliation(s)
- Sebastian Sten
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Fredrik Elinder
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Maria Engström
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.
| |
Collapse
|