51
|
Chen AB, Duque M, Engert F. Seeing stars: Astroglia modulate visual circuits during behavioral-state transitions. Neuron 2023; 111:3903-3905. [PMID: 38128478 DOI: 10.1016/j.neuron.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023]
Abstract
In this issue of Neuron, Uribe-Arias et al.1 show that, in larval zebrafish, astrocyte-like cells exhibit calcium responses to norepinephrine during behavioral-state transitions and alter neuronal response properties. Thus, astroglia can sculpt neuronal dynamics in behaviorally meaningful ways.
Collapse
Affiliation(s)
- Alex B Chen
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Marc Duque
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
52
|
Rosenberg MF, Godoy MI, Wade SD, Paredes MF, Zhang Y, Molofsky AV. β-Adrenergic Signaling Promotes Morphological Maturation of Astrocytes in Female Mice. J Neurosci 2023; 43:8621-8636. [PMID: 37845031 PMCID: PMC10727121 DOI: 10.1523/jneurosci.0357-23.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 10/18/2023] Open
Abstract
Astrocytes play essential roles in the developing nervous system, including supporting synapse function. These astrocyte support functions emerge coincident with brain maturation and may be tailored in a region-specific manner. For example, gray matter astrocytes have elaborate synapse-associated processes and are morphologically and molecularly distinct from white matter astrocytes. This raises the question of whether there are unique environmental cues that promote gray matter astrocyte identity and synaptogenic function. We previously identified adrenergic receptors as preferentially enriched in developing gray versus white matter astrocytes, suggesting that noradrenergic signaling could be a cue that promotes the functional maturation of gray matter astrocytes. We first characterized noradrenergic projections during postnatal brain development in mouse and human, finding that process density was higher in the gray matter and increased concurrently with astrocyte maturation. RNA sequencing revealed that astrocytes in both species expressed α- and β-adrenergic receptors. We found that stimulation of β-adrenergic receptors increased primary branching of rodent astrocytes in vitro Conversely, astrocyte-conditional knockout of the β1-adrenergic receptor reduced the size of gray matter astrocytes and led to dysregulated sensorimotor integration in female mice. These studies suggest that adrenergic signaling to developing astrocytes impacts their morphology and has implications for adult behavior, particularly in female animals. More broadly, they demonstrate a mechanism through which environmental cues impact astrocyte development. Given the key roles of norepinephrine in brain states, such as arousal, stress, and learning, these findings could prompt further inquiry into how developmental stressors impact astrocyte development and adult brain function.SIGNIFICANCE STATEMENT This study demonstrates a role for noradrenergic signaling in the development of gray matter astrocytes. We provide new evidence that the β1-adrenergic receptor is robustly expressed by both mouse and human astrocytes, and that conditional KO of the β1-adrenergic receptor from female mouse astrocytes impairs gray matter astrocyte maturation. Moreover, female conditional KO mice exhibit behavioral deficits in two paradigms that test sensorimotor function. Given the emerging interest in moving beyond RNA sequencing to probe specific pathways that underlie astrocyte heterogeneity, this study provides a foundation for future investigation into the effect of noradrenergic signaling on astrocyte functions in conditions where noradrenergic signaling is altered, such as stress, arousal, and learning.
Collapse
Affiliation(s)
- Marci F Rosenberg
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Medical Scientist Training Program and Biomedical Sciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
| | - Marlesa I Godoy
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Sarah D Wade
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Neurosciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
| | - Mercedes F Paredes
- Department of Neurology, Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, California 94143
- Chan Zuckerberg Biohub-San Francisco, San Francisco, California 94158
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Anna V Molofsky
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Neurosciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
53
|
Fiore F, Alhalaseh K, Dereddi RR, Bodaleo Torres F, Çoban I, Harb A, Agarwal A. Norepinephrine regulates calcium signals and fate of oligodendrocyte precursor cells in the mouse cerebral cortex. Nat Commun 2023; 14:8122. [PMID: 38065932 PMCID: PMC10709653 DOI: 10.1038/s41467-023-43920-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, contributing to myelination and myelin repair. OPCs contact axons and respond to neuronal activity, but how the information relayed by the neuronal activity translates into OPC Ca2+ signals, which in turn influence their fate, remains unknown. We generated transgenic mice for concomitant monitoring of OPCs Ca2+ signals and cell fate using 2-photon microscopy in the somatosensory cortex of awake-behaving mice. Ca2+ signals in OPCs mainly occur within processes and confine to Ca2+ microdomains. A subpopulation of OPCs enhances Ca2+ transients while mice engaged in exploratory locomotion. We found that OPCs responsive to locomotion preferentially differentiate into oligodendrocytes, and locomotion-non-responsive OPCs divide. Norepinephrine mediates locomotion-evoked Ca2+ increases in OPCs by activating α1 adrenergic receptors, and chemogenetic activation of OPCs or noradrenergic neurons promotes OPC differentiation. Hence, we uncovered that for fate decisions OPCs integrate Ca2+ signals, and norepinephrine is a potent regulator of OPC fate.
Collapse
Affiliation(s)
- Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Khaleel Alhalaseh
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Felipe Bodaleo Torres
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ilknur Çoban
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ali Harb
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
54
|
Bame X, Hill RA. Mitochondrial network reorganization and transient expansion during oligodendrocyte generation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570104. [PMID: 38106204 PMCID: PMC10723275 DOI: 10.1101/2023.12.05.570104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes of the central nervous system. This process persists throughout life and is essential for recovery from neurodegeneration. To better understand the cellular checkpoints that occur during oligodendrogenesis, we determined the mitochondrial distribution and morphometrics across the oligodendrocyte lineage in mouse and human cerebral cortex. During oligodendrocyte generation, mitochondrial content expanded concurrently with a change in subcellular partitioning towards the distal processes. These changes were followed by an abrupt loss of mitochondria in the oligodendrocyte processes and myelin, coinciding with sheath compaction. This reorganization and extensive expansion and depletion took 3 days. Oligodendrocyte mitochondria were stationary over days while OPC mitochondrial motility was modulated by animal arousal state within minutes. Aged OPCs also displayed decreased mitochondrial size, content, and motility. Thus, mitochondrial dynamics are linked to oligodendrocyte generation, dynamically modified by their local microenvironment, and altered in the aging brain.
Collapse
Affiliation(s)
- Xhoela Bame
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
55
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.568932. [PMID: 38106119 PMCID: PMC10723263 DOI: 10.1101/2023.12.01.568932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Astrocytes-the most abundant non-neuronal cell type in the mammalian brain-are crucial circuit components that respond to and modulate neuronal activity via calcium (Ca 2+ ) signaling 1-8 . Astrocyte Ca 2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales: from fast, subcellular activity 3,4 to slow, synchronized activity that travels across connected astrocyte networks 9-11 . Furthermore, astrocyte network activity has been shown to influence a wide range of processes 5,8,12 . While astrocyte network activity has important implications for neuronal circuit function, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon Ca 2+ imaging of astrocytes while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca 2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca 2+ activity-propagative events-differentiates astrocyte network responses to these two major neurotransmitters, and gates responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over the course of minutes, contributing to accumulating evidence across multiple model organisms that significant astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales 13-15 . We anticipate that this study will be a starting point for future studies investigating the link between specific astrocyte Ca 2+ activity and specific astrocyte functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
|
56
|
Ngoupaye GT, Mokgokong M, Madlala T, Mabandla MV. Alteration of the α5 GABA receptor and 5HTT lead to cognitive deficits associated with major depressive-like behaviors in a 14-day combined stress rat model. Int J Neurosci 2023; 133:959-976. [PMID: 34937496 DOI: 10.1080/00207454.2021.2019033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 07/13/2021] [Accepted: 12/08/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Current models used to study the pathophysiology of major depressive disorder (MDD) are laborious and time consuming. This study examined the effect of a 14-day combined stress model (CS; corticosterone injection and restraint stress) in male Sprague-Dawley rats and also compare the effect of CS versus 28-day corticosterone treatment on depressive-like behaviour and cognitive deficits. MATERIEL AND METHODS Depressive-like behaviours and cognitive deficits were assessed in the forced swim test (FST), sucrose preference (SPT), Morris water maze (MWM) and novel object recognition (NORT) tests. Real-time PCR and ELISA were respectively used to detect expression of the serotonin transporter (5-HTT), serotonin 1 A receptor (5-HT1A), α5 GABAA receptor, and the concentrations of corticosterone (plasma), GABA and acetylcholinesterase (AChE) in the hippocampus and Prefrontal cortex (PFC).Results CS group showed increased immobility time in the FST, time to reach the MWM platform, higher corticosterone level, and increased expressions of hippocampal and PFC 5-HT1A and α5 GABAA receptors, and AChE compared to their control groups. In contrast, reductions in SPT ratio, discrimination index in NORT, time in target quadrant, and hippocampal 5-HTT expression was noted relative to their control group. Compared to the 28-day corticosterone only group, PFC 5-HT1A, Hippocampal 5-HTT were reduced, while PFC 5-HTT, Hippocampal α5 GABAA receptors, and AChE concentrations were higher in the CS group. CONCLUSION Our CS model induced depressive-like behaviour with early cognitive deficits in rats affecting both hippocampus and PFC. The CS model may be useful in investigating new and comprehensive treatment strategies for MDD.
Collapse
Affiliation(s)
- Gwladys Temkou Ngoupaye
- Discipline of Human Physiology, School of Laboratory Medicine & Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Animal Biology, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Makwena Mokgokong
- Discipline of Human Physiology, School of Laboratory Medicine & Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thobeka Madlala
- Discipline of Human Physiology, School of Laboratory Medicine & Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Musa Vuyisile Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine & Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
57
|
Postnov D, Semyachkina-Glushkovskaya O, Litvinenko E, Kurths J, Penzel T. Mechanisms of Activation of Brain's Drainage during Sleep: The Nightlife of Astrocytes. Cells 2023; 12:2667. [PMID: 37998402 PMCID: PMC10670149 DOI: 10.3390/cells12222667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
The study of functions, mechanisms of generation, and pathways of movement of cerebral fluids has a long history, but the last decade has been especially productive. The proposed glymphatic hypothesis, which suggests a mechanism of the brain waste removal system (BWRS), caused an active discussion on both the criticism of some of the perspectives and our intensive study of new experimental facts. It was especially found that the intensity of the metabolite clearance changes significantly during the transition between sleep and wakefulness. Interestingly, at the cellular level, a number of aspects of this problem have been focused on, such as astrocytes-glial cells, which, over the past two decades, have been recognized as equal partners of neurons and perform many important functions. In particular, an important role was assigned to astrocytes within the framework of the glymphatic hypothesis. In this review, we return to the "astrocytocentric" view of the BWRS function and the explanation of its activation during sleep from the viewpoint of new findings over the last decade. Our main conclusion is that the BWRS's action may be analyzed both at the systemic (whole-brain) and at the local (cellular) level. The local level means here that the neuro-glial-vascular unit can also be regarded as the smallest functional unit of sleep, and therefore, the smallest functional unit of the BWRS.
Collapse
Affiliation(s)
- Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Oxana Semyachkina-Glushkovskaya
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
| | - Elena Litvinenko
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Jürgen Kurths
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Charité — Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
58
|
Pace SA, Myers B. Hindbrain Adrenergic/Noradrenergic Control of Integrated Endocrine and Autonomic Stress Responses. Endocrinology 2023; 165:bqad178. [PMID: 38015813 DOI: 10.1210/endocr/bqad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Hindbrain adrenergic/noradrenergic nuclei facilitate endocrine and autonomic responses to physical and psychological challenges. Neurons that synthesize adrenaline and noradrenaline target hypothalamic structures to modulate endocrine responses while descending spinal projections regulate sympathetic function. Furthermore, these neurons respond to diverse stress-related metabolic, autonomic, and psychosocial challenges. Accordingly, adrenergic and noradrenergic nuclei are integrative hubs that promote physiological adaptation to maintain homeostasis. However, the precise mechanisms through which adrenaline- and noradrenaline-synthesizing neurons sense interoceptive and exteroceptive cues to coordinate physiological responses have yet to be fully elucidated. Additionally, the regulatory role of these cells in the context of chronic stress has received limited attention. This mini-review consolidates reports from preclinical rodent studies on the organization and function of brainstem adrenaline and noradrenaline cells to provide a framework for how these nuclei coordinate endocrine and autonomic physiology. This includes identification of hindbrain adrenaline- and noradrenaline-producing cell groups and their role in stress responding through neurosecretory and autonomic engagement. Although temporally and mechanistically distinct, the endocrine and autonomic stress axes are complementary and interconnected. Therefore, the interplay between brainstem adrenergic/noradrenergic nuclei and peripheral physiological systems is necessary for integrated stress responses and organismal survival.
Collapse
Affiliation(s)
- Sebastian A Pace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
59
|
Rupareliya VP, Singh AA, Butt AM, A H, Kumar H. The "molecular soldiers" of the CNS: Astrocytes, a comprehensive review on their roles and molecular signatures. Eur J Pharmacol 2023; 959:176048. [PMID: 37758010 DOI: 10.1016/j.ejphar.2023.176048] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
For a long time, neurons held the position of central players in the nervous system. Since there are far more astrocytes than neurons in the brain, it makes us wonder if these cells just take up space and support the neurons or if they are actively participating in central nervous system (CNS) homeostasis. Now, astrocytes' contribution to CNS physiology is appreciated as they are known to regulate ion and neurotransmitter levels, synapse formation and elimination, blood-brain barrier integrity, immune function, cerebral blood flow, and many more. In many neurological and psychiatric disorders, astrocyte functions are altered. Advancements in microscopic and transcriptomic tools revealed populations of astrocytes with varied morphology, electrophysiological properties, and transcriptomic profiles. Neuron-circuit-specific functions and neuron-specific interactions of astroglial subpopulations are found, which suggests that diversity is essential in carrying out diverse region-specific CNS functions. Investigations on heterogeneous astrocyte populations are revealing new astrocyte functions and their role in pathological conditions, opening a new therapeutic avenue for targeting neurological conditions. The true extent of astrocytic heterogeneity and its functional implications are yet to be fully explored. This review summarizes essential astrocytic functions and their relevance in pathological conditions and discusses astrocytic diversity in relation to morphology, function, and gene expression throughout the CNS.
Collapse
Affiliation(s)
- Vimal P Rupareliya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Aditya A Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Ayub Mohammed Butt
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hariharan A
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
60
|
Barnett D, Bohmbach K, Grelot V, Charlet A, Dallérac G, Ju YH, Nagai J, Orr AG. Astrocytes as Drivers and Disruptors of Behavior: New Advances in Basic Mechanisms and Therapeutic Targeting. J Neurosci 2023; 43:7463-7471. [PMID: 37940585 PMCID: PMC10634555 DOI: 10.1523/jneurosci.1376-23.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
Astrocytes are emerging as key regulators of cognitive function and behavior. This review highlights some of the latest advances in the understanding of astrocyte roles in different behavioral domains across lifespan and in disease. We address specific molecular and circuit mechanisms by which astrocytes modulate behavior, discuss their functional diversity and versatility, and highlight emerging astrocyte-targeted treatment strategies that might alleviate behavioral and cognitive dysfunction in pathologic conditions. Converging evidence across different model systems and manipulations is revealing that astrocytes regulate behavioral processes in a precise and context-dependent manner. Improved understanding of these astrocytic functions may generate new therapeutic strategies for various conditions with cognitive and behavioral impairments.
Collapse
Affiliation(s)
- Daniel Barnett
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, New York 10021
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York 10021
| | - Kirsten Bohmbach
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Valentin Grelot
- Institute of Cellular and Integrative Neuroscience, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, 67000, France
| | - Alexandre Charlet
- Institute of Cellular and Integrative Neuroscience, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, 67000, France
| | - Glenn Dallérac
- Centre National de la Recherche Scientifique and Paris-Saclay University, Paris-Saclay Institute for Neurosciences, Paris, 91400, France
| | - Yeon Ha Ju
- Department of Psychiatry and Neuroscience, University of Texas-Austin Dell Medical School, Austin, Texas 78712
| | - Jun Nagai
- RIKEN Center for Brain Science, Laboratory for Glia-Neuron Circuit Dynamics, Saitama, 351-0198, Japan
| | - Anna G Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, New York 10021
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
61
|
Murphy-Royal C, Ching S, Papouin T. A conceptual framework for astrocyte function. Nat Neurosci 2023; 26:1848-1856. [PMID: 37857773 PMCID: PMC10990637 DOI: 10.1038/s41593-023-01448-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/01/2023] [Indexed: 10/21/2023]
Abstract
The participation of astrocytes in brain computation was hypothesized in 1992, coinciding with the discovery that these cells display a form of intracellular Ca2+ signaling sensitive to neuroactive molecules. This finding fostered conceptual leaps crystalized around the idea that astrocytes, once thought to be passive, participate actively in brain signaling and outputs. A multitude of disparate roles of astrocytes has since emerged, but their meaningful integration has been muddied by the lack of consensus and models of how we conceive the functional position of these cells in brain circuitry. In this Perspective, we propose an intuitive, data-driven and transferable conceptual framework we coin 'contextual guidance'. It describes astrocytes as 'contextual gates' that shape neural circuitry in an adaptive, state-dependent fashion. This paradigm provides fresh perspectives on principles of astrocyte signaling and its relevance to brain function, which could spur new experimental avenues, including in computational space.
Collapse
Affiliation(s)
- Ciaran Murphy-Royal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) & Département de Neurosciences, Université de Montréal, Montréal, Quebec, Canada
| | - ShiNung Ching
- Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
62
|
Stopper G, Caudal LC, Rieder P, Gobbo D, Stopper L, Felix L, Everaerts K, Bai X, Rose CR, Scheller A, Kirchhoff F. Novel algorithms for improved detection and analysis of fluorescent signal fluctuations. Pflugers Arch 2023; 475:1283-1300. [PMID: 37700120 PMCID: PMC10567899 DOI: 10.1007/s00424-023-02855-3] [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: 05/22/2023] [Revised: 08/02/2023] [Accepted: 08/26/2023] [Indexed: 09/14/2023]
Abstract
Fluorescent dyes and genetically encoded fluorescence indicators (GEFI) are common tools for visualizing concentration changes of specific ions and messenger molecules during intra- as well as intercellular communication. Using advanced imaging technologies, fluorescence indicators are a prerequisite for the analysis of physiological molecular signaling. Automated detection and analysis of fluorescence signals require to overcome several challenges, including correct estimation of fluorescence fluctuations at basal concentrations of messenger molecules, detection, and extraction of events themselves as well as proper segmentation of neighboring events. Moreover, event detection algorithms need to be sensitive enough to accurately capture localized and low amplitude events exhibiting a limited spatial extent. Here, we present two algorithms (PBasE and CoRoDe) for accurate baseline estimation and automated detection and segmentation of fluorescence fluctuations.
Collapse
Affiliation(s)
- Gebhard Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Laura C Caudal
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Phillip Rieder
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Laura Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Lisa Felix
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Katharina Everaerts
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Xianshu Bai
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421, Homburg, Germany.
| |
Collapse
|
63
|
Nanclares C, Noriega-Prieto JA, Labrada-Moncada FE, Cvetanovic M, Araque A, Kofuji P. Altered calcium signaling in Bergmann glia contributes to spinocerebellar ataxia type-1 in a mouse model of SCA1. Neurobiol Dis 2023; 187:106318. [PMID: 37802154 PMCID: PMC10624966 DOI: 10.1016/j.nbd.2023.106318] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the ATAXIN1 (ATXN1) gene and characterized by progressive cerebellar ataxia, dysarthria, and eventual deterioration of bulbar functions. SCA1 shows severe degeneration of cerebellar Purkinje cells (PCs) and activation of Bergmann glia (BG), a type of cerebellar astroglia closely associated with PCs. Combining electrophysiological recordings, calcium imaging techniques, and chemogenetic approaches, we have investigated the electrical intrinsic and synaptic properties of PCs and the physiological properties of BG in SCA1 mouse model expressing mutant ATXN1 only in PCs. PCs of SCA1 mice displayed lower spontaneous firing rate and larger slow afterhyperpolarization currents (sIAHP) than wildtype mice, whereas the properties of the synaptic inputs were unaffected. BG of SCA1 mice showed higher calcium hyperactivity and gliotransmission, manifested by higher frequency of NMDAR-mediated slow inward currents (SICs) in PC. Preventing the BG calcium hyperexcitability of SCA1 mice by loading BG with the calcium chelator BAPTA restored sIAHP and spontaneous firing rate of PCs to similar levels of wildtype mice. Moreover, mimicking the BG hyperactivity by activating BG expressing Gq-DREADDs in wildtype mice reproduced the SCA1 pathological phenotype of PCs, i.e., enhancement of sIAHP and decrease of spontaneous firing rate. These results indicate that the intrinsic electrical properties of PCs, but not their synaptic properties, were altered in SCA1 mice and that these alterations were associated with the hyperexcitability of BG. Moreover, preventing BG hyperexcitability in SCA1 mice and promoting BG hyperexcitability in wildtype mice prevented and mimicked, respectively, the pathological electrophysiological phenotype of PCs. Therefore, BG plays a relevant role in the dysfunction of the electrical intrinsic properties of PCs in SCA1 mice, suggesting that they may serve as potential targets for therapeutic approaches to treat the spinocerebellar ataxia type 1.
Collapse
Affiliation(s)
- Carmen Nanclares
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
64
|
Lu TY, Hanumaihgari P, Hsu ET, Agarwal A, Kawaguchi R, Calabresi PA, Bergles DE. Norepinephrine modulates calcium dynamics in cortical oligodendrocyte precursor cells promoting proliferation during arousal in mice. Nat Neurosci 2023; 26:1739-1750. [PMID: 37697112 PMCID: PMC10630072 DOI: 10.1038/s41593-023-01426-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated from oligodendrocyte precursor cells (OPCs) that express neurotransmitter receptors. However, the mechanisms that affect OPC activity in vivo and the physiological roles of neurotransmitter signaling in OPCs are unclear. In this study, we generated a transgenic mouse line that expresses membrane-anchored GCaMP6s in OPCs and used longitudinal two-photon microscopy to monitor OPC calcium (Ca2+) dynamics in the cerebral cortex. OPCs exhibit focal and transient Ca2+ increases within their processes that are enhanced during locomotion-induced increases in arousal. The Ca2+ transients occur independently of excitatory neuron activity, rapidly decline when OPCs differentiate and are inhibited by anesthesia, sedative agents or noradrenergic receptor antagonists. Conditional knockout of α1A adrenergic receptors in OPCs suppresses spontaneous and locomotion-induced Ca2+ increases and reduces OPC proliferation. Our results demonstrate that OPCs are directly modulated by norepinephrine in vivo to enhance Ca2+ dynamics and promote population homeostasis.
Collapse
Affiliation(s)
- Tsai-Yi Lu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Priyanka Hanumaihgari
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Eric T Hsu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Amit Agarwal
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Riki Kawaguchi
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter A Calabresi
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Division of Neuroimmunology and Neurological Infections, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
65
|
Brazhe A, Verisokin A, Verveyko D, Postnov D. Astrocytes: new evidence, new models, new roles. Biophys Rev 2023; 15:1303-1333. [PMID: 37975000 PMCID: PMC10643736 DOI: 10.1007/s12551-023-01145-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023] Open
Abstract
Astrocytes have been in the limelight of active research for about 3 decades now. Over this period, ideas about their function and role in the nervous system have evolved from simple assistance in energy supply and homeostasis maintenance to a complex informational and metabolic hub that integrates data on local neuronal activity, sensory and arousal context, and orchestrates many crucial processes in the brain. Rapid progress in experimental techniques and data analysis produces a growing body of data, which can be used as a foundation for formulation of new hypotheses, building new refined mathematical models, and ultimately should lead to a new level of understanding of the contribution of astrocytes to the cognitive tasks performed by the brain. Here, we highlight recent progress in astrocyte research, which we believe expands our understanding of how low-level signaling at a cellular level builds up to processes at the level of the whole brain and animal behavior. We start our review with revisiting data on the role of noradrenaline-mediated astrocytic signaling in locomotion, arousal, sensory integration, memory, and sleep. We then briefly review astrocyte contribution to the regulation of cerebral blood flow regulation, which is followed by a discussion of biophysical mechanisms underlying astrocyte effects on different brain processes. The experimental section is closed by an overview of recent experimental techniques available for modulation and visualization of astrocyte dynamics. We then evaluate how the new data can be potentially incorporated into the new mathematical models or where and how it already has been done. Finally, we discuss an interesting prospect that astrocytes may be key players in important processes such as the switching between sleep and wakefulness and the removal of toxic metabolites from the brain milieu.
Collapse
Affiliation(s)
- Alexey Brazhe
- Department of Biophysics, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory, 1/24, Moscow, 119234 Russia
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklay Str., 16/10, Moscow, 117997 Russia
| | - Andrey Verisokin
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Darya Verveyko
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya st., 83, Saratov, 410012 Russia
| |
Collapse
|
66
|
Tanguay E, Bouchard SJ, Lévesque M, De Koninck P, Breton-Provencher V. Shining light on the noradrenergic system. NEUROPHOTONICS 2023; 10:044406. [PMID: 37766924 PMCID: PMC10519836 DOI: 10.1117/1.nph.10.4.044406] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Despite decades of research on the noradrenergic system, our understanding of its impact on brain function and behavior remains incomplete. Traditional recording techniques are challenging to implement for investigating in vivo noradrenergic activity, due to the relatively small size and the position in the brain of the locus coeruleus (LC), the primary location for noradrenergic neurons. However, recent advances in optical and fluorescent methods have enabled researchers to study the LC more effectively. Use of genetically encoded calcium indicators to image the activity of noradrenergic neurons and biosensors that monitor noradrenaline release with fluorescence can be an indispensable tool for studying noradrenergic activity. In this review, we examine how these methods are being applied to record the noradrenergic system in the rodent brain during behavior.
Collapse
Affiliation(s)
| | | | - Martin Lévesque
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Psychiatry and Neuroscience, Faculty of Medicine, Quebec, Quebec, Canada
| | - Paul De Koninck
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Biochemistry, Microbiology, and Bioinformatics, Faculty of Science and Engineering, Quebec, Quebec, Canada
| | - Vincent Breton-Provencher
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Psychiatry and Neuroscience, Faculty of Medicine, Quebec, Quebec, Canada
| |
Collapse
|
67
|
Chaudoin TR, Bonasera SJ, Dunaevsky A, Padmashri R. Exploring behavioral phenotypes in a mouse model of fetal alcohol spectrum disorders. Dev Neurobiol 2023; 83:184-204. [PMID: 37433012 PMCID: PMC10546278 DOI: 10.1002/dneu.22922] [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: 12/12/2022] [Revised: 04/18/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Fetal alcohol spectrum disorders are one of the leading causes of developmental abnormalities worldwide. Maternal consumption of alcohol during pregnancy leads to a diverse range of cognitive and neurobehavioral deficits. Although moderate-to-heavy levels of prenatal alcohol exposure (PAE) have been associated with adverse offspring outcomes, there is limited data on the consequences of chronic low-level PAE. Here, we use a model of maternal voluntary alcohol consumption throughout gestation in a mouse model to investigate the effects of PAE on behavioral phenotypes during late adolescence and early adulthood in male and female offspring. Body composition was measured by dual-energy X-ray absorptiometry. Baseline behaviors, including feeding, drinking, and movement, were examined by performing home cage monitoring studies. The impact of PAE on motor function, motor skill learning, hyperactivity, acoustic reactivity, and sensorimotor gating was investigated by performing a battery of behavioral tests. PAE was found to be associated with altered body composition. No differences in overall movement, food, or water consumption were observed between control and PAE mice. Although PAE offspring of both sexes exhibited deficits in motor skill learning, no differences were observed in basic motor skills such as grip strength and motor coordination. PAE females exhibited a hyperactive phenotype in a novel environment. PAE mice exhibited increased reactivity to acoustic stimuli, and PAE females showed disrupted short-term habituation. Sensorimotor gating was not altered in PAE mice. Collectively, our data show that chronic low-level exposure to alcohol in utero results in behavioral impairments.
Collapse
Affiliation(s)
- Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
68
|
Stogsdill JA, Harwell CC, Goldman SA. Astrocytes as master modulators of neural networks: Synaptic functions and disease-associated dysfunction of astrocytes. Ann N Y Acad Sci 2023; 1525:41-60. [PMID: 37219367 DOI: 10.1111/nyas.15004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Astrocytes are the most abundant glial cell type in the central nervous system and are essential to the development, plasticity, and maintenance of neural circuits. Astrocytes are heterogeneous, with their diversity rooted in developmental programs modulated by the local brain environment. Astrocytes play integral roles in regulating and coordinating neural activity extending far beyond their metabolic support of neurons and other brain cell phenotypes. Both gray and white matter astrocytes occupy critical functional niches capable of modulating brain physiology on time scales slower than synaptic activity but faster than those adaptive responses requiring a structural change or adaptive myelination. Given their many associations and functional roles, it is not surprising that astrocytic dysfunction has been causally implicated in a broad set of neurodegenerative and neuropsychiatric disorders. In this review, we focus on recent discoveries concerning the contributions of astrocytes to the function of neural networks, with a dual focus on the contribution of astrocytes to synaptic development and maturation, and on their role in supporting myelin integrity, and hence conduction and its regulation. We then address the emerging roles of astrocytic dysfunction in disease pathogenesis and on potential strategies for targeting these cells for therapeutic purposes.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Steven A Goldman
- Sana Biotechnology Inc., Cambridge, Massachusetts, USA
- Center for Translational Neuromedicine, University of Rochester, Rochester, New York, USA
- University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
69
|
Götz V, Qiao S, Das D, Wartenberg P, Wyatt A, Wahl V, Gamayun I, Alasmi S, Fecher-Trost C, Meyer MR, Rad R, Kaltenbacher T, Kattler K, Lipp P, Becherer U, Mollard P, Candlish M, Boehm U. Ovulation is triggered by a cyclical modulation of gonadotropes into a hyperexcitable state. Cell Rep 2023; 42:112543. [PMID: 37224016 DOI: 10.1016/j.celrep.2023.112543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Gonadotropes in the anterior pituitary gland are essential for fertility and provide a functional link between the brain and the gonads. To trigger ovulation, gonadotrope cells release massive amounts of luteinizing hormone (LH). The mechanism underlying this remains unclear. Here, we utilize a mouse model expressing a genetically encoded Ca2+ indicator exclusively in gonadotropes to dissect this mechanism in intact pituitaries. We demonstrate that female gonadotropes exclusively exhibit a state of hyperexcitability during the LH surge, resulting in spontaneous [Ca2+]i transients in these cells, which persist in the absence of any in vivo hormonal signals. L-type Ca2+ channels and transient receptor potential channel A1 (TRPA1) together with intracellular reactive oxygen species (ROS) levels ensure this state of hyperexcitability. Consistent with this, virus-assisted triple knockout of Trpa1 and L-type Ca2+ subunits in gonadotropes leads to vaginal closure in cycling females. Our data provide insight into molecular mechanisms required for ovulation and reproductive success in mammals.
Collapse
Affiliation(s)
- Viktoria Götz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Sen Qiao
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Debajyoti Das
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Philipp Wartenberg
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Amanda Wyatt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Vanessa Wahl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Igor Gamayun
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Samer Alasmi
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Claudia Fecher-Trost
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Markus R Meyer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Thorsten Kaltenbacher
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Kathrin Kattler
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken 66123, Germany
| | - Peter Lipp
- Molecular Cell Biology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Ute Becherer
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University School of Medicine, Homburg 66421, Germany
| | - Patrice Mollard
- IGF, CNRS, INSERM, University of Montpellier, Montpellier 34090, France
| | - Michael Candlish
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Ulrich Boehm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany.
| |
Collapse
|
70
|
Sardar D, Cheng YT, Woo J, Choi DJ, Lee ZF, Kwon W, Chen HC, Lozzi B, Cervantes A, Rajendran K, Huang TW, Jain A, Arenkiel B, Maze I, Deneen B. Induction of astrocytic Slc22a3 regulates sensory processing through histone serotonylation. Science 2023; 380:eade0027. [PMID: 37319217 PMCID: PMC10874521 DOI: 10.1126/science.ade0027] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 04/28/2023] [Indexed: 06/17/2023]
Abstract
Neuronal activity drives alterations in gene expression within neurons, yet how it directs transcriptional and epigenomic changes in neighboring astrocytes in functioning circuits is unknown. We found that neuronal activity induces widespread transcriptional up-regulation and down-regulation in astrocytes, highlighted by the identification of Slc22a3 as an activity-inducible astrocyte gene that encodes neuromodulator transporter Slc22a3 and regulates sensory processing in the mouse olfactory bulb. Loss of astrocytic Slc22a3 reduced serotonin levels in astrocytes, leading to alterations in histone serotonylation. Inhibition of histone serotonylation in astrocytes reduced the expression of γ-aminobutyric acid (GABA) biosynthetic genes and GABA release, culminating in olfactory deficits. Our study reveals that neuronal activity orchestrates transcriptional and epigenomic responses in astrocytes while illustrating new mechanisms for how astrocytes process neuromodulatory input to gate neurotransmitter release for sensory processing.
Collapse
Affiliation(s)
- Debosmita Sardar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Yi-Ting Cheng
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Program in Developmental Biology, Baylor College of Medicine, Houston TX
| | - Junsung Woo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Dong-Joo Choi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Zhung-Fu Lee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
- Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston TX
| | - Wookbong Kwon
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Hsiao-Chi Chen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston TX
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston TX
| | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Kavitha Rajendran
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
| | - Teng-Wei Huang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston TX
| | - Benjamin Arenkiel
- Program in Developmental Biology, Baylor College of Medicine, Houston TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX
- Neurological Research Institute, Texas Children’s Hospital, Houston TX
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York NY
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston TX
- Program in Developmental Biology, Baylor College of Medicine, Houston TX
- Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston TX
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| |
Collapse
|
71
|
Kersbergen CJ, Babola TA, Kanold PO, Bergles DE. Preservation of developmental spontaneous activity enables early auditory system maturation in deaf mice. PLoS Biol 2023; 21:e3002160. [PMID: 37368868 PMCID: PMC10298803 DOI: 10.1371/journal.pbio.3002160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/11/2023] [Indexed: 06/29/2023] Open
Abstract
Intrinsically generated neural activity propagates through the developing auditory system to promote maturation and refinement of sound processing circuits prior to hearing onset. This early patterned activity is induced by non-sensory supporting cells in the organ of Corti, which are highly interconnected through gap junctions containing connexin 26 (Gjb2). Although loss of function mutations in Gjb2 impair cochlear development and are the most common cause of congenital deafness, it is not known if these variants disrupt spontaneous activity and the developmental trajectory of sound processing circuits in the brain. Here, we show in a new mouse model of Gjb2-mediated congenital deafness that cochlear supporting cells adjacent to inner hair cells (IHCs) unexpectedly retain intercellular coupling and the capacity to generate spontaneous activity, exhibiting only modest deficits prior to hearing onset. Supporting cells lacking Gjb2 elicited coordinated activation of IHCs, leading to coincident bursts of activity in central auditory neurons that will later process similar frequencies of sound. Despite alterations in the structure of the sensory epithelium, hair cells within the cochlea of Gjb2-deficient mice were intact and central auditory neurons could be activated within appropriate tonotopic domains by loud sounds at hearing onset, indicating that early maturation and refinement of auditory circuits was preserved. Only after cessation of spontaneous activity following hearing onset did progressive hair cell degeneration and enhanced auditory neuron excitability manifest. This preservation of cochlear spontaneous neural activity in the absence of connexin 26 may increase the effectiveness of early therapeutic interventions to restore hearing.
Collapse
Affiliation(s)
- Calvin J. Kersbergen
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Travis A. Babola
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Patrick O. Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Dwight E. Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, United States of America
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
72
|
Salinas-Birt A, Zhu X, Lim EY, Cruz Santory AJ, Ye L, Paukert M. Constraints of vigilance-dependent noradrenergic signaling to mouse cerebellar Bergmann glia. Glia 2023; 71:1451-1465. [PMID: 36790089 PMCID: PMC10082684 DOI: 10.1002/glia.24350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023]
Abstract
Behavioral state plays an important role in determining astroglia Ca2+ signaling. In particular, locomotion-mediated elevated vigilance has been found to trigger norepinephrine-dependent whole cell Ca2+ elevations in astroglia throughout the brain. For cerebellar Bergmann glia it has recently been found that locomotion-induced transient Ca2+ elevations depend on their α1A -adrenergic receptors. With increasing availability and implementation of locomotion as behavioral parameter it becomes important to understand the constraints of noradrenergic signaling to astroglia. Here we evaluated the effect of speed, duration and interval of locomotion on Ca2+ signals in Bergmann glia as well as cerebellar noradrenergic axon terminals. We found almost no dependence on locomotion speed, but following the initial Ca2+ transient prolonged locomotion events revealed a steady-state Ca2+ elevation. Comparison of time course and recovery of transient Bergmann glia and noradrenergic terminal Ca2+ dynamics suggested that noradrenergic terminal Ca2+ activity determines Bergmann glia Ca2+ activation and does not require noradrenergic receptor desensitization to account for attenuation during prolonged locomotion. Further, analyzing the correlation among Ca2+ dynamics within regions within the field of observation we found that coordinated activity among noradrenergic terminals accounts for fluctuations of steady-state Bergmann glia Ca2+ activity. Together, our findings will help to better understand astroglia Ca2+ dynamics during less controlled awake behavior and may guide the identification of behavioral contexts preferably dependent on astroglia Ca2+ signaling.
Collapse
Affiliation(s)
- Angelica Salinas-Birt
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Xiangyu Zhu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Eunice Y. Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Aryana J. Cruz Santory
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
73
|
Rasmussen RN, Asiminas A, Carlsen EMM, Kjaerby C, Smith NA. Astrocytes: integrators of arousal state and sensory context. Trends Neurosci 2023; 46:418-425. [PMID: 37003933 PMCID: PMC10192111 DOI: 10.1016/j.tins.2023.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/24/2023] [Accepted: 03/05/2023] [Indexed: 04/01/2023]
Abstract
The integration of external information with the internal state of the body is central to the survival of virtually every multicellular organism. However, a complete picture of the mechanisms that govern this process is lacking. In this opinion article, we synthesize evidence demonstrating that astrocytes sense the momentary arousal state - through neuromodulator release - as well as the sensory inputs - through local synaptic activity - and respond to them with changes in calcium (Ca2+) signaling. We hypothesize that astrocytes integrate sensory signals with the internal state and that this process is necessary to secure optimal behavior. Finally, we argue that dysfunctional astrocytic Ca2+ signaling could be an underlying factor in disorders characterized by disrupted sensory processing.
Collapse
Affiliation(s)
- Rune Nguyen Rasmussen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Antonis Asiminas
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Celia Kjaerby
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nathan Anthony Smith
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
74
|
Wei H, Wu X, Withrow J, Cuevas-Diaz Duran R, Singh S, Chaboub LS, Rakshit J, Mejia J, Rolfe A, Herrera JJ, Horner PJ, Wu JQ. Glial progenitor heterogeneity and key regulators revealed by single-cell RNA sequencing provide insight to regeneration in spinal cord injury. Cell Rep 2023; 42:112486. [PMID: 37149868 PMCID: PMC10511029 DOI: 10.1016/j.celrep.2023.112486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/12/2023] [Accepted: 04/22/2023] [Indexed: 05/09/2023] Open
Abstract
Recent studies have revealed the heterogeneous nature of astrocytes; however, how diverse constituents of astrocyte-lineage cells are regulated in adult spinal cord after injury and contribute to regeneration remains elusive. We perform single-cell RNA sequencing of GFAP-expressing cells from sub-chronic spinal cord injury models and identify and compare with the subpopulations in acute-stage data. We find subpopulations with distinct functional enrichment and their identities defined by subpopulation-specific transcription factors and regulons. Immunohistochemistry, RNAscope experiments, and quantification by stereology verify the molecular signature, location, and morphology of potential resident neural progenitors or neural stem cells in the adult spinal cord before and after injury and uncover the populations of the intermediate cells enriched in neuronal genes that could potentially transition into other subpopulations. This study has expanded the knowledge of the heterogeneity and cell state transition of glial progenitors in adult spinal cord before and after injury.
Collapse
Affiliation(s)
- Haichao Wei
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA
| | - Xizi Wu
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA
| | - Joseph Withrow
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Raquel Cuevas-Diaz Duran
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León 64710, Mexico
| | - Simranjit Singh
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA
| | - Lesley S Chaboub
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jyotirmoy Rakshit
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA
| | - Julio Mejia
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Andrew Rolfe
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA
| | - Juan J Herrera
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, UTHealth, Houston, TX 77030, USA
| | - Philip J Horner
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Jia Qian Wu
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX 77030, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
75
|
Gau YTA, Hsu E, Cha J, Pak RW, Looger LL, Kang JU, Bergles DE. Multicore fiber optic imaging reveals that astrocyte calcium activity in the cerebral cortex is modulated by internal motivational state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541390. [PMID: 37292710 PMCID: PMC10245653 DOI: 10.1101/2023.05.18.541390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astrocytes are a direct target of neuromodulators and can influence neuronal activity on broad spatial and temporal scales through their close proximity to synapses. However, our knowledge about how astrocytes are functionally recruited during different animal behaviors and their diverse effects on the CNS remains limited. To enable measurement of astrocyte activity patterns in vivo during normative behaviors, we developed a high-resolution, long working distance, multi-core fiber optic imaging platform that allows visualization of cortical astrocyte calcium transients through a cranial window in freely moving mice. Using this platform, we defined the spatiotemporal dynamics of astrocytes during diverse behaviors, ranging from circadian fluctuations to novelty exploration, showing that astrocyte activity patterns are more variable and less synchronous than apparent in head-immobilized imaging conditions. Although the activity of astrocytes in visual cortex was highly synchronized during quiescence to arousal transitions, individual astrocytes often exhibited distinct thresholds and activity patterns during explorative behaviors, in accordance with their molecular diversity, allowing temporal sequencing across the astrocyte network. Imaging astrocyte activity during self-initiated behaviors revealed that noradrenergic and cholinergic systems act synergistically to recruit astrocytes during state transitions associated with arousal and attention, which was profoundly modulated by internal state. The distinct activity patterns exhibited by astrocytes in the cerebral cortex may provide a means to vary their neuromodulatory influence in response to different behaviors and internal states.
Collapse
Affiliation(s)
- Yung-Tian A. Gau
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Eric Hsu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Jaepyeong Cha
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Rebecca W. Pak
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Loren L. Looger
- Howard Hughes Medical Institute, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States
| | - Jin U. Kang
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Dwight E. Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
76
|
Barbay T, Pecchi E, Ducrocq M, Rouach N, Brocard F, Bos R. Astrocytic Kir4.1 channels regulate locomotion by orchestrating neuronal rhythmicity in the spinal network. Glia 2023; 71:1259-1277. [PMID: 36645018 DOI: 10.1002/glia.24337] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/17/2023]
Abstract
Neuronal rhythmogenesis in the spinal cord is correlated with variations in extracellular K+ levels ([K+ ]e ). Astrocytes play important role in [K+ ]e homeostasis and compute neuronal information. Yet it is unclear how neuronal oscillations are regulated by astrocytic K+ homeostasis. Here we identify the astrocytic inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) as a key molecular player for neuronal rhythmicity in the spinal central pattern generator (CPG). By combining two-photon calcium imaging with electrophysiology, immunohistochemistry and genetic tools, we report that astrocytes display Ca2+ transients before and during oscillations of neighboring neurons. Inhibition of astrocytic Ca2+ transients with BAPTA decreases the barium-sensitive Kir4.1 current responsible of K+ clearance. Finally, we show in mice that Kir4.1 knockdown in astrocytes progressively prevents neuronal oscillations and alters the locomotor pattern resulting in lower motor performances in challenging tasks. These data identify astroglial Kir4.1 channels as key regulators of neuronal rhythmogenesis in the CPG driving locomotion.
Collapse
Affiliation(s)
- Tony Barbay
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Emilie Pecchi
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Myriam Ducrocq
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Frédéric Brocard
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Rémi Bos
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| |
Collapse
|
77
|
Zorec R, Vardjan N. Adrenergic regulation of astroglial aerobic glycolysis and lipid metabolism: Towards a noradrenergic hypothesis of neurodegeneration. Neurobiol Dis 2023; 182:106132. [PMID: 37094775 DOI: 10.1016/j.nbd.2023.106132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ageing is a key factor in the development of cognitive decline and dementia, an increasing and challenging problem of the modern world. The most commonly diagnosed cognitive decline is related to Alzheimer's disease (AD), the pathophysiology of which is poorly understood. Several hypotheses have been proposed. The cholinergic hypothesis is the oldest, however, recently the noradrenergic system has been considered to have a role as well. The aim of this review is to provide evidence that supports the view that an impaired noradrenergic system is causally linked to AD. Although dementia is associated with neurodegeneration and loss of neurons, this likely develops due to a primary failure of homeostatic cells, astrocytes, abundant and heterogeneous neuroglial cells in the central nervous system (CNS). The many functions that astrocytes provide to maintain the viability of neural networks include the control of ionic balance, neurotransmitter turnover, synaptic connectivity and energy balance. This latter function is regulated by noradrenaline, released from the axon varicosities of neurons arising from the locus coeruleus (LC), the primary site of noradrenaline release in the CNS. The demise of the LC is linked to AD, whereby a hypometabolic CNS state is observed clinically. This is likely due to impaired release of noradrenaline in the AD brain during states of arousal, attention and awareness. These functions controlled by the LC are needed for learning and memory formation and require activation of the energy metabolism. In this review, we address first the process of neurodegeneration and cognitive decline, highlighting the function of astrocytes. Cholinergic and/or noradrenergic deficits lead to impaired astroglial function. Then, we focus on adrenergic control of astroglial aerobic glycolysis and lipid droplet metabolism, which play a protective role but also promote neurodegeneration under some circumstances, supporting the noradrenergic hypothesis of cognitive decline. We conclude that targeting astroglial metabolism, glycolysis and/or mitochondrial processes may lead to important new developments in the future when searching for medicines to prevent or even halt cognitive decline.
Collapse
Affiliation(s)
- Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
78
|
Wang F, Wang W, Gu S, Qi D, Smith NA, Peng W, Dong W, Yuan J, Zhao B, Mao Y, Cao P, Lu QR, Shapiro LA, Yi SS, Wu E, Huang JH. Distinct astrocytic modulatory roles in sensory transmission during sleep, wakefulness, and arousal states in freely moving mice. Nat Commun 2023; 14:2186. [PMID: 37069258 PMCID: PMC10110578 DOI: 10.1038/s41467-023-37974-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/04/2023] [Indexed: 04/19/2023] Open
Abstract
Despite extensive research on astrocytic Ca2+ in synaptic transmission, its contribution to the modulation of sensory transmission during different brain states remains largely unknown. Here, by using two-photon microscopy and whole-cell recordings, we show two distinct astrocytic Ca2+ signals in the murine barrel cortex: a small, long-lasting Ca2+ increase during sleep and a large, widespread but short-lasting Ca2+ spike when aroused. The large Ca2+ wave in aroused mice was inositol trisphosphate (IP3)-dependent, evoked by the locus coeruleus-norepinephrine system, and enhanced sensory input, contributing to reliable sensory transmission. However, the small Ca2+ transient was IP3-independent and contributed to decreased extracellular K+, hyperpolarization of the neurons, and suppression of sensory transmission. These events respond to different pharmacological inputs and contribute to distinct sleep and arousal functions by modulating the efficacy of sensory transmission. Together, our data demonstrate an important function for astrocytes in sleep and arousal states via astrocytic Ca2+ waves.
Collapse
Affiliation(s)
- Fushun Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, 610060, China.
- Department of Neurosurgery, University of Rochester, Rochester, NY, 14643, USA.
| | - Wei Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, 610060, China
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Simeng Gu
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, 610060, China
- Department of Medical Psychology, Jiangsu University Medical School, Zhenjiang, 212013, China
| | - Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA
| | - Nathan A Smith
- Department of Neurosurgery, University of Rochester, Rochester, NY, 14643, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Weiguo Peng
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, 610060, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Jiajin Yuan
- Department of Neurosurgery, University of Rochester, Rochester, NY, 14643, USA
| | - Binbin Zhao
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Peng Cao
- School of Psychology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qing Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Lee A Shapiro
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, Bryan, TX, 77807, USA.
| | - S Stephen Yi
- Livestrong Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA.
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, 78712, USA.
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA.
- Livestrong Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA.
- Texas A & M University Health Science Center, College Station, TX, 77843, USA.
| | - Jason H Huang
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA.
- Texas A & M University Health Science Center, College Station, TX, 77843, USA.
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76508, USA.
| |
Collapse
|
79
|
Reitman ME, Tse V, Mi X, Willoughby DD, Peinado A, Aivazidis A, Myagmar BE, Simpson PC, Bayraktar OA, Yu G, Poskanzer KE. Norepinephrine links astrocytic activity to regulation of cortical state. Nat Neurosci 2023; 26:579-593. [PMID: 36997759 PMCID: PMC10089924 DOI: 10.1038/s41593-023-01284-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 02/14/2023] [Indexed: 04/01/2023]
Abstract
Cortical state, defined by population-level neuronal activity patterns, determines sensory perception. While arousal-associated neuromodulators-including norepinephrine (NE)-reduce cortical synchrony, how the cortex resynchronizes remains unknown. Furthermore, general mechanisms regulating cortical synchrony in the wake state are poorly understood. Using in vivo imaging and electrophysiology in mouse visual cortex, we describe a critical role for cortical astrocytes in circuit resynchronization. We characterize astrocytes' calcium responses to changes in behavioral arousal and NE, and show that astrocytes signal when arousal-driven neuronal activity is reduced and bi-hemispheric cortical synchrony is increased. Using in vivo pharmacology, we uncover a paradoxical, synchronizing response to Adra1a receptor stimulation. We reconcile these results by demonstrating that astrocyte-specific deletion of Adra1a enhances arousal-driven neuronal activity, while impairing arousal-related cortical synchrony. Our findings demonstrate that astrocytic NE signaling acts as a distinct neuromodulatory pathway, regulating cortical state and linking arousal-associated desynchrony to cortical circuit resynchronization.
Collapse
Affiliation(s)
- Michael E Reitman
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Xuelong Mi
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Drew D Willoughby
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Alba Peinado
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Bat-Erdene Myagmar
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Kira E Poskanzer
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
80
|
Manninen T, Aćimović J, Linne ML. Analysis of Network Models with Neuron-Astrocyte Interactions. Neuroinformatics 2023; 21:375-406. [PMID: 36959372 PMCID: PMC10085960 DOI: 10.1007/s12021-023-09622-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 03/25/2023]
Abstract
Neural networks, composed of many neurons and governed by complex interactions between them, are a widely accepted formalism for modeling and exploring global dynamics and emergent properties in brain systems. In the past decades, experimental evidence of computationally relevant neuron-astrocyte interactions, as well as the astrocytic modulation of global neural dynamics, have accumulated. These findings motivated advances in computational glioscience and inspired several models integrating mechanisms of neuron-astrocyte interactions into the standard neural network formalism. These models were developed to study, for example, synchronization, information transfer, synaptic plasticity, and hyperexcitability, as well as classification tasks and hardware implementations. We here focus on network models of at least two neurons interacting bidirectionally with at least two astrocytes that include explicitly modeled astrocytic calcium dynamics. In this study, we analyze the evolution of these models and the biophysical, biochemical, cellular, and network mechanisms used to construct them. Based on our analysis, we propose how to systematically describe and categorize interaction schemes between cells in neuron-astrocyte networks. We additionally study the models in view of the existing experimental data and present future perspectives. Our analysis is an important first step towards understanding astrocytic contribution to brain functions. However, more advances are needed to collect comprehensive data about astrocyte morphology and physiology in vivo and to better integrate them in data-driven computational models. Broadening the discussion about theoretical approaches and expanding the computational tools is necessary to better understand astrocytes' roles in brain functions.
Collapse
Affiliation(s)
- Tiina Manninen
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland.
| | - Jugoslava Aćimović
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland.
| |
Collapse
|
81
|
Yu Q, Gamayun I, Wartenberg P, Zhang Q, Qiao S, Kusumakshi S, Candlish S, Götz V, Wen S, Das D, Wyatt A, Wahl V, Ectors F, Kattler K, Yildiz D, Prevot V, Schwaninger M, Ternier G, Giacobini P, Ciofi P, Müller TD, Boehm U. Bitter taste cells in the ventricular walls of the murine brain regulate glucose homeostasis. Nat Commun 2023; 14:1588. [PMID: 36949050 PMCID: PMC10033832 DOI: 10.1038/s41467-023-37099-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
The median eminence (ME) is a circumventricular organ at the base of the brain that controls body homeostasis. Tanycytes are its specialized glial cells that constitute the ventricular walls and regulate different physiological states, however individual signaling pathways in these cells are incompletely understood. Here, we identify a functional tanycyte subpopulation that expresses key taste transduction genes including bitter taste receptors, the G protein gustducin and the gustatory ion channel TRPM5 (M5). M5 tanycytes have access to blood-borne cues via processes extended towards diaphragmed endothelial fenestrations in the ME and mediate bidirectional communication between the cerebrospinal fluid and blood. This subpopulation responds to metabolic signals including leptin and other hormonal cues and is transcriptionally reprogrammed upon fasting. Acute M5 tanycyte activation induces insulin secretion and acute diphtheria toxin-mediated M5 tanycyte depletion results in impaired glucose tolerance in diet-induced obese mice. We provide a cellular and molecular framework that defines how bitter taste cells in the ME integrate chemosensation with metabolism.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Igor Gamayun
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Qian Zhang
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Soumya Kusumakshi
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Sarah Candlish
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Viktoria Götz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Shuping Wen
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Debajyoti Das
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Kathrin Kattler
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Daniela Yildiz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Philippe Ciofi
- Neurocentre Magendie - INSERM Unit 1215, University of Bordeaux, Bordeaux, France
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ulrich Boehm
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany.
| |
Collapse
|
82
|
Lia A, Sansevero G, Chiavegato A, Sbrissa M, Pendin D, Mariotti L, Pozzan T, Berardi N, Carmignoto G, Fasolato C, Zonta M. Rescue of astrocyte activity by the calcium sensor STIM1 restores long-term synaptic plasticity in female mice modelling Alzheimer's disease. Nat Commun 2023; 14:1590. [PMID: 36949142 PMCID: PMC10033875 DOI: 10.1038/s41467-023-37240-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 03/09/2023] [Indexed: 03/24/2023] Open
Abstract
Calcium dynamics in astrocytes represent a fundamental signal that through gliotransmitter release regulates synaptic plasticity and behaviour. Here we present a longitudinal study in the PS2APP mouse model of Alzheimer's disease (AD) linking astrocyte Ca2+ hypoactivity to memory loss. At the onset of plaque deposition, somatosensory cortical astrocytes of AD female mice exhibit a drastic reduction of Ca2+ signaling, closely associated with decreased endoplasmic reticulum Ca2+ concentration and reduced expression of the Ca2+ sensor STIM1. In parallel, astrocyte-dependent long-term synaptic plasticity declines in the somatosensory circuitry, anticipating specific tactile memory loss. Notably, we show that both astrocyte Ca2+ signaling and long-term synaptic plasticity are fully recovered by selective STIM1 overexpression in astrocytes. Our data unveil astrocyte Ca2+ hypoactivity in neocortical astrocytes as a functional hallmark of early AD stages and indicate astrocytic STIM1 as a target to rescue memory deficits.
Collapse
Affiliation(s)
- Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Gabriele Sansevero
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Miriana Sbrissa
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Diana Pendin
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Letizia Mariotti
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, Padua, Italy
| | - Nicoletta Berardi
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
83
|
Salmon CK, Syed TA, Kacerovsky JB, Alivodej N, Schober AL, Sloan TFW, Pratte MT, Rosen MP, Green M, Chirgwin-Dasgupta A, Mehta S, Jilani A, Wang Y, Vali H, Mandato CA, Siddiqi K, Murai KK. Organizing principles of astrocytic nanoarchitecture in the mouse cerebral cortex. Curr Biol 2023; 33:957-972.e5. [PMID: 36805126 DOI: 10.1016/j.cub.2023.01.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/01/2022] [Accepted: 01/20/2023] [Indexed: 02/18/2023]
Abstract
Astrocytes are increasingly understood to be important regulators of central nervous system (CNS) function in health and disease; yet, we have little quantitative understanding of their complex architecture. While broad categories of astrocytic structures are known, the discrete building blocks that compose them, along with their geometry and organizing principles, are poorly understood. Quantitative investigation of astrocytic complexity is impeded by the absence of high-resolution datasets and robust computational approaches to analyze these intricate cells. To address this, we produced four ultra-high-resolution datasets of mouse cerebral cortex using serial electron microscopy and developed astrocyte-tailored computer vision methods for accurate structural analysis. We unearthed specific anatomical building blocks, structural motifs, connectivity hubs, and hierarchical organizations of astrocytes. Furthermore, we found that astrocytes interact with discrete clusters of synapses and that astrocytic mitochondria are distributed to lie closer to larger clusters of synapses. Our findings provide a geometrically principled, quantitative understanding of astrocytic nanoarchitecture and point to an unexpected level of complexity in how astrocytes interact with CNS microanatomy.
Collapse
Affiliation(s)
- Christopher K Salmon
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Tabish A Syed
- School of Computer Science and Centre for Intelligent Machines, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada; MILA - Québec AI Institute, 6666 Rue Saint-Urbain, Montreal, QC H2S 3H1, Canada
| | - J Benjamin Kacerovsky
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Nensi Alivodej
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Alexandra L Schober
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | | | - Michael T Pratte
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Michael P Rosen
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Miranda Green
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Adario Chirgwin-Dasgupta
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Shaurya Mehta
- School of Computer Science and Centre for Intelligent Machines, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada
| | - Affan Jilani
- School of Computer Science and Centre for Intelligent Machines, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada
| | - Yanan Wang
- School of Computer Science and Centre for Intelligent Machines, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada
| | - Hojatollah Vali
- Department of Anatomy & Cell Biology, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| | - Craig A Mandato
- Department of Anatomy & Cell Biology, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| | - Kaleem Siddiqi
- School of Computer Science and Centre for Intelligent Machines, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada; MILA - Québec AI Institute, 6666 Rue Saint-Urbain, Montreal, QC H2S 3H1, Canada.
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.
| |
Collapse
|
84
|
Astrocytes in the pathophysiology of neuroinfection. Essays Biochem 2023; 67:131-145. [PMID: 36562155 DOI: 10.1042/ebc20220082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Key homeostasis providing cells in the central nervous system (CNS) are astrocytes, which belong to the class of cells known as atroglia, a highly heterogeneous type of neuroglia and a prominent element of the brain defence. Diseases evolve due to altered homeostatic state, associated with pathology-induced astroglia remodelling represented by reactive astrocytes, astroglial atrophy and astrodegeneration. These features are hallmarks of most infectious insults, mediated by bacteria, protozoa and viruses; they are also prominent in the systemic infection. The COVID-19 pandemic revived the focus into neurotropic viruses such as SARS-CoV2 (Coronaviridae) but also the Flaviviridae viruses including tick-borne encephalitis (TBEV) and Zika virus (ZIKV) causing the epidemic in South America prior to COVID-19. Astrocytes provide a key response to neurotropic infections in the CNS. Astrocytes form a parenchymal part of the blood-brain barrier, the site of virus entry into the CNS. Astrocytes exhibit aerobic glycolysis, a form of metabolism characteristic of highly morphologically plastic cells, like cancer cells, hence a suitable milieu for multiplication of infectious agent, including viral particles. However, why the protection afforded by astrocytes fails in some circumstances is an open question to be studied in the future.
Collapse
|
85
|
Goenaga J, Araque A, Kofuji P, Herrera Moro Chao D. Calcium signaling in astrocytes and gliotransmitter release. Front Synaptic Neurosci 2023; 15:1138577. [PMID: 36937570 PMCID: PMC10017551 DOI: 10.3389/fnsyn.2023.1138577] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Glia are as numerous in the brain as neurons and widely known to serve supportive roles such as structural scaffolding, extracellular ionic and neurotransmitter homeostasis, and metabolic support. However, over the past two decades, several lines of evidence indicate that astrocytes, which are a type of glia, play active roles in neural information processing. Astrocytes, although not electrically active, can exhibit a form of excitability by dynamic changes in intracellular calcium levels. They sense synaptic activity and release neuroactive substances, named gliotransmitters, that modulate neuronal activity and synaptic transmission in several brain areas, thus impacting animal behavior. This "dialogue" between astrocytes and neurons is embodied in the concept of the tripartite synapse that includes astrocytes as integral elements of synaptic function. Here, we review the recent work and discuss how astrocytes via calcium-mediated excitability modulate synaptic information processing at various spatial and time scales.
Collapse
Affiliation(s)
| | | | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
86
|
Analysis of a cell niche with proliferative potential at the roof of the aqueduct of Sylvius. Neurosci Res 2023; 188:28-38. [PMID: 36375656 DOI: 10.1016/j.neures.2022.11.004] [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: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
The aqueduct of Sylvius connects the third with the fourth ventricle and is surrounded by the Periaqueductal Grey. Here, we report a novel niche of cells in the dorsal section of the aqueduct, hereby named dorsal aqueduct niche or DAN, by applying a battery of selective markers and transgenic mouse lines. The somata of DAN cells are located toward the lumen of the ventricle forming multiple layers in close association with the cerebrospinal fluid (CSF). A single process emerges from the soma and run with the blood vessels. Cells of the DAN express radial glia/stem cell markers such as GFAP, vimentin and nestin, and the glutamate transporter GLAST or the oligodendrocyte precursor/pericyte marker NG2, thereby suggesting their potential for the generation of new cells. Morphologically, DAN cells resemble tanycytes of the third ventricle, which transfer biochemical signals from the CSF to the central nervous system and display proliferative capacity. The aqueduct ependymal lining can proliferate as observed by the integration of BrdU and expression of Ki67. Thus, the dorsal section of the aqueduct of Sylvius possesses cells that may act a niche of new glial cells in the adult mouse brain.
Collapse
|
87
|
Sardar D, Cheng YT, Woo J, Choi DJ, Lee ZF, Kwon W, Chen HC, Lozzi B, Cervantes A, Rajendran K, Huang TW, Jain A, Arenkiel B, Maze I, Deneen B. Activity-dependent induction of astrocytic Slc22a3 regulates sensory processing through histone serotonylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529904. [PMID: 36909526 PMCID: PMC10002681 DOI: 10.1101/2023.02.24.529904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Neuronal activity drives global alterations in gene expression within neurons, yet how it directs transcriptional and epigenomic changes in neighboring astrocytes in functioning circuits is unknown. Here we show that neuronal activity induces widespread transcriptional upregulation and downregulation in astrocytes, highlighted by the identification of a neuromodulator transporter Slc22a3 as an activity-inducible astrocyte gene regulating sensory processing in the olfactory bulb. Loss of astrocytic Slc22a3 reduces serotonin levels in astrocytes, leading to alterations in histone serotonylation. Inhibition of histone serotonylation in astrocytes reduces expression of GABA biosynthetic genes and GABA release, culminating in olfactory deficits. Our study reveals that neuronal activity orchestrates transcriptional and epigenomic responses in astrocytes, while illustrating new mechanisms for how astrocytes process neuromodulatory input to gate neurotransmitter release for sensory processing.
Collapse
|
88
|
Pan L, Trimarco A, Zhang AJ, Fujimori K, Urade Y, Sun LO, Taveggia C, Zhang Y. Oligodendrocyte-lineage cell exocytosis and L-type prostaglandin D synthase promote oligodendrocyte development and myelination. eLife 2023; 12:e77441. [PMID: 36779701 PMCID: PMC9946447 DOI: 10.7554/elife.77441] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/12/2023] [Indexed: 02/14/2023] Open
Abstract
In the developing central nervous system, oligodendrocyte precursor cells (OPCs) differentiate into oligodendrocytes, which form myelin around axons. Oligodendrocytes and myelin are essential for the function of the central nervous system, as evidenced by the severe neurological symptoms that arise in demyelinating diseases such as multiple sclerosis and leukodystrophy. Although many cell-intrinsic mechanisms that regulate oligodendrocyte development and myelination have been reported, it remains unclear whether interactions among oligodendrocyte-lineage cells (OPCs and oligodendrocytes) affect oligodendrocyte development and myelination. Here, we show that blocking vesicle-associated membrane protein (VAMP) 1/2/3-dependent exocytosis from oligodendrocyte-lineage cells impairs oligodendrocyte development, myelination, and motor behavior in mice. Adding oligodendrocyte-lineage cell-secreted molecules to secretion-deficient OPC cultures partially restores the morphological maturation of oligodendrocytes. Moreover, we identified L-type prostaglandin D synthase as an oligodendrocyte-lineage cell-secreted protein that promotes oligodendrocyte development and myelination in vivo. These findings reveal a novel autocrine/paracrine loop model for the regulation of oligodendrocyte and myelin development.
Collapse
Affiliation(s)
- Lin Pan
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Amelia Trimarco
- Division of Neuroscience, IRCCS, San Raffaele HospitalMilanItaly
| | - Alice J Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Ko Fujimori
- Department of Pathobiochemistry, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Yoshihiro Urade
- Hirono Satellite, Isotope Science Center, The University of TokyoFukushimaJapan
| | - Lu O Sun
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Carla Taveggia
- Division of Neuroscience, IRCCS, San Raffaele HospitalMilanItaly
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Brain Research Institute, University of California, Los AngelesLos AngelesUnited States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los AngelesLos AngelesUnited States
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
89
|
Turner KL, Gheres KW, Drew PJ. Relating Pupil Diameter and Blinking to Cortical Activity and Hemodynamics across Arousal States. J Neurosci 2023; 43:949-964. [PMID: 36517240 PMCID: PMC9908322 DOI: 10.1523/jneurosci.1244-22.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Arousal state affects neural activity and vascular dynamics in the cortex, with sleep associated with large changes in the local field potential and increases in cortical blood flow. We investigated the relationship between pupil diameter and blink rate with neural activity and blood volume in the somatosensory cortex in male and female unanesthetized, head-fixed mice. We monitored these variables while the mice were awake, during periods of rapid eye movement (REM), and non-rapid eye movement (NREM) sleep. Pupil diameter was smaller during sleep than in the awake state. Changes in pupil diameter were coherent with both gamma-band power and blood volume in the somatosensory cortex, but the strength and sign of this relationship varied with arousal state. We observed a strong negative correlation between pupil diameter and both gamma-band power and blood volume during periods of awake rest and NREM sleep, although the correlations between pupil diameter and these signals became positive during periods of alertness, active whisking, and REM. Blinking was associated with increases in arousal and decreases in blood volume when the mouse was asleep. Bilateral coherence in gamma-band power and in blood volume dropped following awake blinking, indicating a reset of neural and vascular activity. Using only eye metrics (pupil diameter and eye motion), we could determine the arousal state of the mouse ('Awake,' 'NREM,' 'REM') with >90% accuracy with a 5 s resolution. There is a strong relationship between pupil diameter and hemodynamics signals in mice, reflecting the pronounced effects of arousal on cerebrovascular dynamics.SIGNIFICANCE STATEMENT Determining arousal state is a critical component of any neuroscience experiment. Pupil diameter and blinking are influenced by arousal state, as are hemodynamics signals in the cortex. We investigated the relationship between cortical hemodynamics and pupil diameter and found that pupil diameter was strongly related to the blood volume in the cortex. Mice were more likely to be awake after blinking than before, and blinking resets neural activity. Pupil diameter and eye motion can be used as a reliable, noninvasive indicator of arousal state. As mice transition from wake to sleep and back again over a timescale of seconds, monitoring pupil diameter and eye motion permits the noninvasive detection of sleep events during behavioral or resting-state experiments.
Collapse
Affiliation(s)
- Kevin L Turner
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Kyle W Gheres
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Departments of Engineering Science and Mechanics
| | - Patrick J Drew
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Departments of Engineering Science and Mechanics
- Biology and Neurosurgery, Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
90
|
Peng W, Liu X, Ma G, Wu Z, Wang Z, Fei X, Qin M, Wang L, Li Y, Zhang S, Xu M. Adenosine-independent regulation of the sleep-wake cycle by astrocyte activity. Cell Discov 2023; 9:16. [PMID: 36746933 PMCID: PMC9902472 DOI: 10.1038/s41421-022-00498-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/20/2022] [Indexed: 02/08/2023] Open
Abstract
Astrocytes play a crucial role in regulating sleep-wake behavior, and adenosine signaling is generally thought to be involved. Here we show multiple lines of evidence supporting that modulation of the sleep-wake behavior by astrocyte Ca2+ activity could occur without adenosine signaling. In the basal forebrain and the brainstem, two brain regions that are known to be essential for sleep-wake regulation, chemogenetically-induced astrocyte Ca2+ elevation significantly modulated the sleep-wake cycle. Although astrocyte Ca2+ level positively correlated with the amount of extracellular adenosine, as revealed by a genetically encoded adenosine sensor, we found no detectable change in adenosine level after suppressing astrocyte Ca2+ elevation, and transgenic mice lacking one of the major extracellular ATP-adenosine conversion enzymes showed similar extracellular adenosine level and astrocyte Ca2+-induced sleep modulation. Furthermore, astrocyte Ca2+ is dependent primarily on local neuronal activity, causing brain region-specific regulation of the sleep-wake cycle. Thus, neural activity-dependent astrocyte activity could regulate the sleep-wake behavior independent of adenosine signaling.
Collapse
Affiliation(s)
- Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaotong Liu
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Guofen Ma
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaofa Wu
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziyue Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Fei
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Meiling Qin
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lizhao Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulong Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Siyu Zhang
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shangha, China.
| |
Collapse
|
91
|
Fang LP, Liu Q, Meyer E, Welle A, Huang W, Scheller A, Kirchhoff F, Bai X. A subset of OPCs do not express Olig2 during development which can be increased in the adult by brain injuries and complex motor learning. Glia 2023; 71:415-430. [PMID: 36308278 DOI: 10.1002/glia.24284] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are uniformly distributed in the mammalian brain; however, their function is rather heterogeneous in respect to their origin, location, receptor/channel expression and age. The basic helix-loop-helix transcription factor Olig2 is expressed in all OPCs as a pivotal determinant of their differentiation. Here, we identified a subset (2%-26%) of OPCs lacking Olig2 in various brain regions including cortex, corpus callosum, CA1 and dentate gyrus. These Olig2 negative (Olig2neg ) OPCs were enriched in the juvenile brain and decreased subsequently with age, being rarely detectable in the adult brain. However, the loss of this population was not due to apoptosis or microglia-dependent phagocytosis. Unlike Olig2pos OPCs, these subset cells were rarely labeled for the mitotic marker Ki67. And, accordingly, BrdU was incorporated only by a three-day long-term labeling but not by a 2-hour short pulse, suggesting these cells do not proliferate any more but were derived from proliferating OPCs. The Olig2neg OPCs exhibited a less complex morphology than Olig2pos ones. Olig2neg OPCs preferentially remain in a precursor stage rather than differentiating into highly branched oligodendrocytes. Changing the adjacent brain environment, for example, by acute injuries or by complex motor learning tasks, stimulated the transition of Olig2pos OPCs to Olig2neg cells in the adult. Taken together, our results demonstrate that OPCs transiently suppress Olig2 upon changes of the brain activity.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Qing Liu
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany.,Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Anna Welle
- Department of Genetics and EpiGenetics, University of Saarland, Saarbrücken, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany.,Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| |
Collapse
|
92
|
Natsubori A, Hirai S, Kwon S, Ono D, Deng F, Wan J, Miyazawa M, Kojima T, Okado H, Karashima A, Li Y, Tanaka KF, Honda M. Serotonergic neurons control cortical neuronal intracellular energy dynamics by modulating astrocyte-neuron lactate shuttle. iScience 2023; 26:105830. [PMID: 36713262 PMCID: PMC9881222 DOI: 10.1016/j.isci.2022.105830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
The central serotonergic system has multiple roles in animal physiology and behavior, including sleep-wake control. However, its function in controlling brain energy metabolism according to the state of animals remains undetermined. Through in vivo monitoring of energy metabolites and signaling, we demonstrated that optogenetic activation of raphe serotonergic neurons increased cortical neuronal intracellular concentration of ATP, an indispensable cellular energy molecule, which was suppressed by inhibiting neuronal uptake of lactate derived from astrocytes. Raphe serotonergic neuronal activation induced cortical astrocytic Ca2+ and cAMP surges and increased extracellular lactate concentrations, suggesting the facilitation of lactate release from astrocytes. Furthermore, chemogenetic inhibition of raphe serotonergic neurons partly attenuated the increase in cortical neuronal intracellular ATP levels as arousal increased in mice. Serotonergic neuronal activation promoted an increase in cortical neuronal intracellular ATP levels, partly mediated by the facilitation of the astrocyte-neuron lactate shuttle, contributing to state-dependent optimization of neuronal intracellular energy levels.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Corresponding author
| | - Shinobu Hirai
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Daisuke Ono
- Department of Neuroscience Ⅱ, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Fei Deng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Momoka Miyazawa
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Faculty of Science Division Ⅱ, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruo Okado
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
93
|
Lu L, Gao Z, Wei Z, Yi M. Working memory depends on the excitatory-inhibitory balance in neuron-astrocyte network. CHAOS (WOODBURY, N.Y.) 2023; 33:013127. [PMID: 36725632 DOI: 10.1063/5.0126890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/19/2022] [Indexed: 06/18/2023]
Abstract
Previous studies have shown that astrocytes are involved in information processing and working memory (WM) in the central nervous system. Here, the neuron-astrocyte network model with biological properties is built to study the effects of excitatory-inhibitory balance and neural network structures on WM tasks. It is found that the performance metrics of WM tasks under the scale-free network are higher than other network structures, and the WM task can be successfully completed when the proportion of excitatory neurons in the network exceeds 30%. There exists an optimal region for the proportion of excitatory neurons and synaptic weight that the memory performance metrics of the WM tasks are higher. The multi-item WM task shows that the spatial calcium patterns for different items overlap significantly in the astrocyte network, which is consistent with the formation of cognitive memory in the brain. Moreover, complex image tasks show that cued recall can significantly reduce systematic noise and maintain the stability of the WM tasks. The results may contribute to understand the mechanisms of WM formation and provide some inspirations into the dynamic storage and recall of memory.
Collapse
Affiliation(s)
- Lulu Lu
- School of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Zhuoheng Gao
- School of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Zhouchao Wei
- School of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Ming Yi
- School of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| |
Collapse
|
94
|
Tiberi A, Carucci NM, Testa G, Rizzi C, Pacifico P, Borgonovo G, Arisi I, D’Onofrio M, Brandi R, Gan WB, Capsoni S, Cattaneo A. Reduced levels of NGF shift astrocytes toward a neurotoxic phenotype. Front Cell Dev Biol 2023; 11:1165125. [PMID: 37143894 PMCID: PMC10151754 DOI: 10.3389/fcell.2023.1165125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
Nerve growth factor (NGF) is critical for neuronal physiology during development and adulthood. Despite the well-recognized effect of NGF on neurons, less is known about whether NGF can actually affect other cell types in the central nervous system (CNS). In this work, we show that astrocytes are susceptible to changes in ambient levels of NGF. First, we observe that interfering with NGF signaling in vivo via the constitutive expression of an antiNGF antibody induces astrocytic atrophy. A similar asthenic phenotype is encountered in an uncleavable proNGF transgenic mouse model (TgproNGF#72), effectively increasing the brain proNGF levels. To examine whether this effect on astrocytes is cell-autonomous, we cultured wild-type primary astrocytes in the presence of antiNGF antibodies, uncovering that a short incubation period is sufficient to potently and rapidly trigger calcium oscillations. Acute induction of calcium oscillations by antiNGF antibodies is followed by progressive morphological changes similar to those observed in antiNGF AD11 mice. Conversely, incubation with mature NGF has no effect on either calcium activity nor on astrocytic morphology. At longer timescales, transcriptomic analysis revealed that NGF-deprived astrocytes acquire a proinflammatory profile. In particular, antiNGF-treated astrocytes show upregulation of neurotoxic transcripts and downregulation of neuroprotective mRNAs. Consistent with that data, culturing wild-type neurons in the presence of NGF-deprived astrocytes leads to neuronal cell death. Finally, we report that in both awake and anesthetized mice, astrocytes in layer I of the motor cortex respond with an increase in calcium activity to acute NGF inhibition using either NGF-neutralizing antibodies or a TrkA-Fc NGF scavenger. Moreover, in vivo calcium imaging in the cortex of the 5xFAD neurodegeneration mouse model shows an increased level of spontaneous calcium activity in astrocytes, which is significantly reduced after acute administration of NGF. In conclusion, we unveil a novel neurotoxic mechanism driven by astrocytes, triggered by their sensing and reacting to changes in the levels of ambient NGF.
Collapse
Affiliation(s)
- Alexia Tiberi
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
| | | | | | | | | | | | - Ivan Arisi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Mara D’Onofrio
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rossella Brandi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Wen-Biao Gan
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Simona Capsoni
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Institute of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Antonino Cattaneo
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
- *Correspondence: Antonino Cattaneo,
| |
Collapse
|
95
|
Eraso‐Pichot A, Pouvreau S, Olivera‐Pinto A, Gomez‐Sotres P, Skupio U, Marsicano G. Endocannabinoid signaling in astrocytes. Glia 2023; 71:44-59. [PMID: 35822691 PMCID: PMC9796923 DOI: 10.1002/glia.24246] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023]
Abstract
The study of the astrocytic contribution to brain functions has been growing in popularity in the neuroscience field. In the last years, and especially since the demonstration of the involvement of astrocytes in synaptic functions, the astrocyte field has revealed multiple functions of these cells that seemed inconceivable not long ago. In parallel, cannabinoid investigation has also identified different ways by which cannabinoids are able to interact with these cells, modify their functions, alter their communication with neurons and impact behavior. In this review, we will describe the expression of different endocannabinoid system members in astrocytes. Moreover, we will relate the latest findings regarding cannabinoid modulation of some of the most relevant astroglial functions, namely calcium (Ca2+ ) dynamics, gliotransmission, metabolism, and inflammation.
Collapse
Affiliation(s)
- Abel Eraso‐Pichot
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Sandrine Pouvreau
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Alexandre Olivera‐Pinto
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Paula Gomez‐Sotres
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Urszula Skupio
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Giovanni Marsicano
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| |
Collapse
|
96
|
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
|
97
|
Aghighi Z, Ghorbani Z, Moghaddam MH, Fathi M, Abdollahifar MA, Soleimani M, Karimzadeh F, Rasoolijazi H, Aliaghaei A. Melittin ameliorates motor function and prevents autophagy-induced cell death and astrogliosis in rat models of cerebellar ataxia induced by 3-acetylpyridine. Neuropeptides 2022; 96:102295. [PMID: 36280441 DOI: 10.1016/j.npep.2022.102295] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cerebellar ataxia (CA) is a form of ataxia that adversely affects the cerebellum. This study aims to investigate the therapeutic effects of melittin (MEL) on a 3-acetylpyridine-induced (3-AP) cerebellar ataxia (CA) rat model. METHODS Initially, CA rat models were generated by 3-AP administration followed by the subcutaneous injection of MEL. The open-field test was used for the evaluation of locomotion and anxiety. Immunohistochemistry was also conducted for the autophagy markers of LC3 and Beclin1. In the next step, the morphology of the astrocyte, the cell responsible for maintaining homeostasis in the CNS, was evaluated by the Sholl analysis. RESULTS The findings suggested that the administration of MEL in a 3-AP model of ataxia improved locomotion and anxiety (P < 0.001), decreased the expression of LC3 (P < 0.01) and Beclin1 (P < 0.05), increased astrocyte complexity (P < 0.05) and reduced astrocyte cell soma size (P < 0.001). CONCLUSIONS Overall, the findings imply that the MEL attenuates the 3-AP-induced autophagy, causes cell death and improves motor function. As such, it could be used as a therapeutic procedure for CA due to its neuroprotective effects.
Collapse
Affiliation(s)
- Zahra Aghighi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zeynab Ghorbani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fariba Karimzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Homa Rasoolijazi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
98
|
Kersbergen CJ, Babola TA, Rock J, Bergles DE. Developmental spontaneous activity promotes formation of sensory domains, frequency tuning and proper gain in central auditory circuits. Cell Rep 2022; 41:111649. [PMID: 36384119 PMCID: PMC9730452 DOI: 10.1016/j.celrep.2022.111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/24/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
Abstract
Neurons that process sensory information exhibit bursts of electrical activity during development, providing early training to circuits that will later encode similar features of the external world. In the mammalian auditory system, this intrinsically generated activity emerges from the cochlea prior to hearing onset, but its role in maturation of auditory circuitry remains poorly understood. We show that selective suppression of cochlear supporting cell spontaneous activity disrupts patterned burst firing of central auditory neurons without affecting cell survival or acoustic thresholds. However, neurons in the inferior colliculus of these mice exhibit enhanced acoustic sensitivity and broader frequency tuning, resulting in wider isofrequency laminae. Despite this enhanced neural responsiveness, total tone-responsive regions in the auditory cortex are substantially smaller. Thus, disruption of pre-hearing cochlear activity causes profound changes in neural encoding of sound, with important implications for restoration of hearing in individuals who experience reduced activity during this critical developmental period.
Collapse
Affiliation(s)
- Calvin J Kersbergen
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Travis A Babola
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | | | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA; Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
99
|
Ma L, Day-Cooney J, Benavides OJ, Muniak MA, Qin M, Ding JB, Mao T, Zhong H. Locomotion activates PKA through dopamine and adenosine in striatal neurons. Nature 2022; 611:762-768. [PMID: 36352228 PMCID: PMC10752255 DOI: 10.1038/s41586-022-05407-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/03/2022] [Indexed: 11/10/2022]
Abstract
The canonical model of striatal function predicts that animal locomotion is associated with the opposing regulation of protein kinase A (PKA) in direct and indirect pathway striatal spiny projection neurons (SPNs) by dopamine1-7. However, the precise dynamics of PKA in dorsolateral SPNs during locomotion remain to be determined. It is also unclear whether other neuromodulators are involved. Here we show that PKA activity in both types of SPNs is essential for normal locomotion. Using two-photon fluorescence lifetime imaging8-10 of a PKA sensor10 through gradient index lenses, we measured PKA activity within individual SPNs of the mouse dorsolateral striatum during locomotion. Consistent with the canonical view, dopamine activated PKA activity in direct pathway SPNs during locomotion through the dopamine D1 receptor. However, indirect pathway SPNs exhibited a greater increase in PKA activity, which was largely abolished through the blockade of adenosine A2A receptors. In agreement with these results, fibre photometry measurements of an adenosine sensor11 revealed an acute increase in extracellular adenosine during locomotion. Functionally, antagonism of dopamine or adenosine receptors resulted in distinct changes in SPN PKA activity, neuronal activity and locomotion. Together, our results suggest that acute adenosine accumulation interplays with dopamine release to orchestrate PKA activity in SPNs and proper striatal function during animal locomotion.
Collapse
Affiliation(s)
- Lei Ma
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Omar Jáidar Benavides
- Department of Neurosurgery and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael A Muniak
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jun B Ding
- Department of Neurosurgery and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
100
|
Meyer E, Rieder P, Gobbo D, Candido G, Scheller A, de Oliveira RMW, Kirchhoff F. Cannabidiol Exerts a Neuroprotective and Glia-Balancing Effect in the Subacute Phase of Stroke. Int J Mol Sci 2022; 23:12886. [PMID: 36361675 PMCID: PMC9659180 DOI: 10.3390/ijms232112886] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022] Open
Abstract
Pharmacological agents limiting secondary tissue loss and improving functional outcomes after stroke are still limited. Cannabidiol (CBD), the major non-psychoactive component of Cannabis sativa, has been proposed as a neuroprotective agent against experimental cerebral ischemia. The effects of CBD mostly relate to the modulation of neuroinflammation, including glial activation. To investigate the effects of CBD on glial cells after focal ischemia in vivo, we performed time-lapse imaging of microglia and astroglial Ca2+ signaling in the somatosensory cortex in the subacute phase of stroke by in vivo two-photon laser-scanning microscopy using transgenic mice with microglial EGFP expression and astrocyte-specific expression of the genetically encoded Ca2+ sensor GCaMP3. CBD (10 mg/kg, intraperitoneally) prevented ischemia-induced neurological impairment, reducing the neurological deficit score from 2.0 ± 1.2 to 0.8 ± 0.8, and protected against neurodegeneration, as shown by the reduction (more than 70%) in Fluoro-Jade C staining (18.8 ± 7.5 to 5.3 ± 0.3). CBD reduced ischemia-induced microglial activation assessed by changes in soma area and total branch length, and exerted a balancing effect on astroglial Ca2+ signals. Our findings indicate that the neuroprotective effects of CBD may occur in the subacute phase of ischemia, and reinforce its strong anti-inflammatory property. Nevertheless, its mechanism of action on glial cells still requires further studies.
Collapse
Affiliation(s)
- Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Phillip Rieder
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Gabriella Candido
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Rúbia Maria Weffort de Oliveira
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| |
Collapse
|