1
|
Ciani C, Falcone C. Interlaminar and varicose-projection astrocytes: toward a new understanding of the primate brain. Front Cell Neurosci 2024; 18:1477753. [PMID: 39655243 PMCID: PMC11626530 DOI: 10.3389/fncel.2024.1477753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
In the last years, science started to move toward a more glio-neurocentric view, in which astrocytes are hypothesized to be directly involved in cognitive functions. Indeed, astrocytes show a variety of shapes with species-specific characteristics, suggesting a specialization of roles during evolution. Interlaminar (ILA) and varicose-projection (VP-As) astrocytes show an anatomical organization that is different compared to the classical horizontal net typically formed by protoplasmic and fibrous astrocytes. ILAs show a modular architecture with the soma in the first cortical layer and processes toward the deep layers with species-specific length. VP-As reside in the deep layers of the cortex, are characterized by varicosities on the longest processes, and are individual-specific. These characteristics suggest roles that are more complex than what was theorized until now. Here, we recapitulate what we know so far from literature from the first time ILAs were described to the most recent discoveries, spanning from morphology description, hypothesis on the development to their features in diseases. For a complete glance on this topic, we included a final paragraph on which techniques and models were used to study ILAs and VP-As, and what new avenues may be opened thanks to more novel methods.
Collapse
Affiliation(s)
| | - Carmen Falcone
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
2
|
Huff A, Oliveira LM, Karlen-Amarante M, Ebiala F, Ramirez JM, Kalume F. Ndufs4 inactivation in glutamatergic neurons reveals swallow-breathing discoordination in a mouse model of Leigh Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612506. [PMID: 39314482 PMCID: PMC11419062 DOI: 10.1101/2024.09.11.612506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Swallowing, both nutritive and non-nutritive, is highly dysfunctional in children with Leigh Syndrome (LS) and contributes to the need for both gastrostomy and tracheostomy tube placement. Without these interventions aspiration of food, liquid, and mucus occur resulting in repeated bouts of respiratory infection. No study has investigated whether mouse models of LS, a neurometabolic disorder, exhibit dysfunctions in neuromuscular activity of swallow and breathing integration. We used a genetic mouse model of LS in which the NDUFS4 gene is knocked out (KO) specifically in Vglut2 or Gad2 neurons. We found increased variability of the swallow motor pattern, disruption in breathing regeneration post swallow, and water-induced apneas only in Vglut2 KO mice. These physiological changes likely contribute to weight loss and premature death seen in this mouse model. Following chronic hypoxia (CH) exposure, swallow motor pattern, breathing regeneration, weight, and life expectancy were not changed in the Vglut2-Ndufs4-KO CH mice compared to control, indicating a rescue of phenotypes. These findings show that like patients with LS, Ndufs4 mouse models of LS exhibit swallow impairments as well as swallow-breathing dyscoordination alongside the other phenotypic traits described in previous studies. Understanding this aspect of LS will open roads for the development of future more efficacious therapeutic intervention for this illness.
Collapse
Affiliation(s)
- Alyssa Huff
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Luiz Marcelo Oliveira
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Favour Ebiala
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA, 98108
| | - Jan Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA, 98108
| | - Franck Kalume
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA, 98108
| |
Collapse
|
3
|
Silva AI, Socodato R, Pinto C, Terceiro AF, Canedo T, Relvas JB, Saraiva M, Summavielle T. IL-10 and Cdc42 modulate astrocyte-mediated microglia activation in methamphetamine-induced neuroinflammation. Glia 2024; 72:1501-1517. [PMID: 38780232 DOI: 10.1002/glia.24542] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Methamphetamine (Meth) use is known to induce complex neuroinflammatory responses, particularly involving astrocytes and microglia. Building upon our previous research, which demonstrated that Meth stimulates astrocytes to release tumor necrosis factor (TNF) and glutamate, leading to microglial activation, this study investigates the role of the anti-inflammatory cytokine interleukin-10 (IL-10) in this process. Our findings reveal that the presence of recombinant IL-10 (rIL-10) counteracts Meth-induced excessive glutamate release in astrocyte cultures, which significantly reduces microglial activation. This reduction is associated with the modulation of astrocytic intracellular calcium (Ca2+) dynamics, particularly by restricting the release of Ca2+ from the endoplasmic reticulum to the cytoplasm. Furthermore, we identify the small Rho GTPase Cdc42 as a crucial intermediary in the astrocyte-to-microglia communication pathway under Meth exposure. By employing a transgenic mouse model that overexpresses IL-10 (pMT-10), we also demonstrate in vivo that IL-10 prevents Meth-induced neuroinflammation. These findings not only enhance our understanding of Meth-related neuroinflammatory mechanisms, but also suggest IL-10 and Cdc42 as putative therapeutic targets for treating Meth-induced neuroinflammation.
Collapse
Affiliation(s)
- Ana Isabel Silva
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Renato Socodato
- Glia Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Carolina Pinto
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ana Filipa Terceiro
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Teresa Canedo
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Glia Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Margarida Saraiva
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Immune Regulation Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Teresa Summavielle
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal
| |
Collapse
|
4
|
Khalilpour J, Soltani Zangbar H, Alipour MR, Shahabi P. The hypoxic respiratory response of the pre-Bötzinger complex. Heliyon 2024; 10:e34491. [PMID: 39114066 PMCID: PMC11305331 DOI: 10.1016/j.heliyon.2024.e34491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Since the discovery of the pre-Bötzinger Complex (preBötC) as a crucial region for generating the main respiratory rhythm, our understanding of its cellular and molecular aspects has rapidly increased within the last few decades. It is now apparent that preBötC is a highly flexible neuronal network that reconfigures state-dependently to produce the most appropriate respiratory output in response to various metabolic challenges, such as hypoxia. However, the responses of the preBötC to hypoxic conditions can be varied based on the intensity, pattern, and duration of the hypoxic challenge. This review discusses the preBötC response to hypoxic challenges at the cellular and network level. Particularly, the involvement of preBötC in the classical biphasic response of the respiratory network to acute hypoxia is illuminated. Furthermore, the article discusses the functional and structural changes of preBötC neurons following intermittent and sustained hypoxic challenges. Accumulating evidence shows that the preBötC neural circuits undergo substantial changes following hypoxia and contribute to several types of the respiratory system's hypoxic ventilatory responses.
Collapse
Affiliation(s)
- Jamal Khalilpour
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
C Benincasa J, Madias MI, Kandell RM, Delgado-Garcia LM, Engler AJ, Kwon EJ, Porcionatto MA. Mechanobiological Modulation of In Vitro Astrocyte Reactivity Using Variable Gel Stiffness. ACS Biomater Sci Eng 2024; 10:4279-4296. [PMID: 38870483 PMCID: PMC11234334 DOI: 10.1021/acsbiomaterials.4c00229] [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] [Indexed: 06/15/2024]
Abstract
After traumatic brain injury, the brain extracellular matrix undergoes structural rearrangement due to changes in matrix composition, activation of proteases, and deposition of chondroitin sulfate proteoglycans by reactive astrocytes to produce the glial scar. These changes lead to a softening of the tissue, where the stiffness of the contusion "core" and peripheral "pericontusional" regions becomes softer than that of healthy tissue. Pioneering mechanotransduction studies have shown that soft substrates upregulate intermediate filament proteins in reactive astrocytes; however, many other aspects of astrocyte biology remain unclear. Here, we developed a platform for the culture of cortical astrocytes using polyacrylamide (PA) gels of varying stiffness (measured in Pascal; Pa) to mimic injury-related regions in order to investigate the effects of tissue stiffness on astrocyte reactivity and morphology. Our results show that substrate stiffness influences astrocyte phenotype; soft 300 Pa substrates led to increased GFAP immunoreactivity, proliferation, and complexity of processes. Intermediate 800 Pa substrates increased Aggrecan+, Brevican+, and Neurocan+ astrocytes. The stiffest 1 kPa substrates led to astrocytes with basal morphologies, similar to a physiological state. These results advance our understanding of astrocyte mechanotransduction processes and provide evidence of how substrates with engineered stiffness can mimic the injury microenvironment.
Collapse
Affiliation(s)
- Julia C Benincasa
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039032, Brazil
| | - Marianne I Madias
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Rebecca M Kandell
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Lina M Delgado-Garcia
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039032, Brazil
| | - Adam J Engler
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Marimelia A Porcionatto
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039032, Brazil
| |
Collapse
|
6
|
Ferreira AFF, Ulrich H, Feng ZP, Sun HS, Britto LR. Neurodegeneration and glial morphological changes are both prevented by TRPM2 inhibition during the progression of a Parkinson's disease mouse model. Exp Neurol 2024; 377:114780. [PMID: 38649091 DOI: 10.1016/j.expneurol.2024.114780] [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: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by dopaminergic neuron death and neuroinflammation. Emerging evidence points to the involvement of the transient receptor potential melastatin 2 (TRPM2) channel in neuron death and glial activation in several neurodegenerative diseases. However, the involvement of TRPM2 in PD and specifically its relation to the neuroinflammation aspect of the disease remains poorly understood. Here, we hypothesized that AG490, a TRPM2 inhibitor, can be used as a treatment in a mouse model of PD. Mice underwent stereotaxic surgery for 6-hydroxydopamine (6-OHDA) administration in the right striatum. Motor behavioral tests (apomorphine, cylinder, and rotarod) were performed on day 3 post-injection to confirm the PD model induction. AG490 was then daily injected i.p. between days 3 to 6 after surgery. On day 6, motor behavior was assessed again. Substantia nigra (SNc) and striatum (CPu) were collected for immunohistochemistry, immunoblotting, and RT-qPCR analysis on day 7. Our results revealed that AG490 post-treatment reduced motor behavior impairment and nigrostriatal neurodegeneration. In addition, the compound prevented TRPM2 upregulation and changes of the Akt/GSK-3β/caspase-3 signaling pathway. The TRPM2 inhibition also avoids the glial morphology changes observed in the PD group. Remarkably, the morphometrical analysis revealed that the ameboid-shaped microglia, found in 6-OHDA-injected animals, were no longer present in the AG490-treated group. These results indicate that AG490 treatment can reduce dopaminergic neuronal death and suppress neuroinflammation in a PD mouse model. Inhibition of TRPM2 by AG490 could then represent a potential therapeutical strategy to be evaluated for PD treatment.
Collapse
Affiliation(s)
- Ana Flavia F Ferreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Han M, Yildiz E, Bozuyuk U, Aydin A, Yu Y, Bhargava A, Karaz S, Sitti M. Janus microparticles-based targeted and spatially-controlled piezoelectric neural stimulation via low-intensity focused ultrasound. Nat Commun 2024; 15:2013. [PMID: 38443369 PMCID: PMC10915158 DOI: 10.1038/s41467-024-46245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Electrical stimulation is a fundamental tool in studying neural circuits, treating neurological diseases, and advancing regenerative medicine. Injectable, free-standing piezoelectric particle systems have emerged as non-genetic and wireless alternatives for electrode-based tethered stimulation systems. However, achieving cell-specific and high-frequency piezoelectric neural stimulation remains challenging due to high-intensity thresholds, non-specific diffusion, and internalization of particles. Here, we develop cell-sized 20 μm-diameter silica-based piezoelectric magnetic Janus microparticles (PEMPs), enabling clinically-relevant high-frequency neural stimulation of primary neurons under low-intensity focused ultrasound. Owing to its functionally anisotropic design, half of the PEMP acts as a piezoelectric electrode via conjugated barium titanate nanoparticles to induce electrical stimulation, while the nickel-gold nanofilm-coated magnetic half provides spatial and orientational control on neural stimulation via external uniform rotating magnetic fields. Furthermore, surface functionalization with targeting antibodies enables cell-specific binding/targeting and stimulation of dopaminergic neurons. Taking advantage of such functionalities, the PEMP design offers unique features towards wireless neural stimulation for minimally invasive treatment of neurological diseases.
Collapse
Affiliation(s)
- Mertcan Han
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, 8092, Zurich, Switzerland
| | - Erdost Yildiz
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Ugur Bozuyuk
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Asli Aydin
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yan Yu
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Aarushi Bhargava
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Selcan Karaz
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, 8092, Zurich, Switzerland
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany.
- Institute for Biomedical Engineering, ETH Zurich, 8092, Zurich, Switzerland.
- School of Medicine and College of Engineering, Koç University, 34450, Istanbul, Türkiye.
| |
Collapse
|
8
|
Bishop M, SheikhBahei S. Brainstem astrocytes regulate breathing and may affect arousal state in rats. Physiol Behav 2024; 275:114457. [PMID: 38184289 PMCID: PMC10853942 DOI: 10.1016/j.physbeh.2024.114457] [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: 09/26/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
Variations in arousal levels can impact respiratory patterns. The mechanisms by which breathing behaviors can influence arousal state is not fully understood. In this study, we investigated the role of astrocytes in the preBötzinger complex (preBötC) in modulating arousal states via breathing in adult conscious rats. Using viral vector tools, we selectively interfered with astrocytic signaling in the preBötC. Rats with inhibited astrocytic signaling exhibited slower breathing rates and behaviors indicative of a calmer state, whereas enhanced purinergic signaling in preBötC astrocytes led to faster breathing and heightened arousal. Our findings reveal a key role for an astrocyte-mediated mechanism in the preBötC that influences both respiratory behaviors and higher-order brain functions like arousal, suggesting a bidirectional link between breathing behaviors and mental states.
Collapse
Affiliation(s)
- Mitchell Bishop
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Shahriar SheikhBahei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA.
| |
Collapse
|
9
|
Adeck A, Millwater M, Bragg C, Zhang R, SheikhBahaei S. Morphological deficits of glial cells in a transgenic mouse model for developmental stuttering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574051. [PMID: 38260402 PMCID: PMC10802298 DOI: 10.1101/2024.01.04.574051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Vocal production involves intricate neural coordination across various brain regions. Stuttering, a common speech disorder, has genetic underpinnings, including mutations in lysosomal-targeting pathway genes. Using a Gnptab-mutant mouse model linked to stuttering, we examined neuron and glial cell morphology in vocal production circuits. Our findings revealed altered astrocyte and microglia processes in these circuits in Gnptab-mutant mice, while control regions remained unaffected. Our results shed light on the potential role of glial cells in stuttering pathophysiology and highlight their relevance in modulating vocal production behaviors.
Collapse
|
10
|
SheikhBahaei S, Marina N, Rajani V, Kasparov S, Funk GD, Smith JC, Gourine AV. Contributions of carotid bodies, retrotrapezoid nucleus neurons and preBötzinger complex astrocytes to the CO 2 -sensitive drive for breathing. J Physiol 2024; 602:223-240. [PMID: 37742121 PMCID: PMC10841148 DOI: 10.1113/jp283534] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/06/2023] [Indexed: 09/25/2023] Open
Abstract
Current models of respiratory CO2 chemosensitivity are centred around the function of a specific population of neurons residing in the medullary retrotrapezoid nucleus (RTN). However, there is significant evidence suggesting that chemosensitive neurons exist in other brainstem areas, including the rhythm-generating region of the medulla oblongata - the preBötzinger complex (preBötC). There is also evidence that astrocytes, non-neuronal brain cells, contribute to central CO2 chemosensitivity. In this study, we reevaluated the relative contributions of the RTN neurons, the preBötC astrocytes, and the carotid body chemoreceptors in mediating the respiratory responses to CO2 in experimental animals (adult laboratory rats). To block astroglial signalling via exocytotic release of transmitters, preBötC astrocytes were targeted to express the tetanus toxin light chain (TeLC). Bilateral expression of TeLC in preBötC astrocytes was associated with ∼20% and ∼30% reduction of the respiratory response to CO2 in conscious and anaesthetized animals, respectively. Carotid body denervation reduced the CO2 respiratory response by ∼25%. Bilateral inhibition of RTN neurons transduced to express Gi-coupled designer receptors exclusively activated by designer drug (DREADDGi ) by application of clozapine-N-oxide reduced the CO2 response by ∼20% and ∼40% in conscious and anaesthetized rats, respectively. Combined blockade of astroglial signalling in the preBötC, inhibition of RTN neurons and carotid body denervation reduced the CO2 -induced respiratory response by ∼70%. These data further support the hypothesis that the CO2 -sensitive drive to breathe requires inputs from the peripheral chemoreceptors and several central chemoreceptor sites. At the preBötC level, astrocytes modulate the activity of the respiratory network in response to CO2 , either by relaying chemosensory information (i.e. they act as CO2 sensors) or by enhancing the preBötC network excitability to chemosensory inputs. KEY POINTS: This study reevaluated the roles played by the carotid bodies, neurons of the retrotrapezoid nucleus (RTN) and astrocytes of the preBötC in mediating the CO2 -sensitive drive to breathe. The data obtained show that disruption of preBötC astroglial signalling, blockade of inputs from the peripheral chemoreceptors or inhibition of RTN neurons similarly reduce the respiratory response to hypercapnia. These data provide further support for the hypothesis that the CO2 -sensitive drive to breathe is mediated by the inputs from the peripheral chemoreceptors and several central chemoreceptor sites.
Collapse
Affiliation(s)
- Shahriar SheikhBahaei
- Centre for Cardiovascular and Metabolic Neuroscience, Research Department of Neuroscience Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
- present address: Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic Neuroscience, Research Department of Neuroscience Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Vishaal Rajani
- Department of Physiology, Neuroscience & Mental Health Institute, Women and Children’s Health Research Institute, University of Alberta, T6G 2E1, Canada
- present address: Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada
| | - Sergey Kasparov
- Department of Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Gregory D. Funk
- Department of Physiology, Neuroscience & Mental Health Institute, Women and Children’s Health Research Institute, University of Alberta, T6G 2E1, Canada
| | - Jeffrey C. Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Alexander V. Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Research Department of Neuroscience Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
11
|
Battaglia M, Rossignol O, Lorenzo LE, Deguire J, Godin AG, D’Amato FR, De Koninck Y. Enhanced harm detection following maternal separation: Transgenerational transmission and reversibility by inhaled amiloride. SCIENCE ADVANCES 2023; 9:eadi8750. [PMID: 37792939 PMCID: PMC10550232 DOI: 10.1126/sciadv.adi8750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Early-life adversities are associated with altered defensive responses. Here, we demonstrate that the repeated cross-fostering (RCF) paradigm of early maternal separation is associated with enhancements of distinct homeostatic reactions: hyperventilation in response to hypercapnia and nociceptive sensitivity, among the first generation of RCF-exposed animals, as well as among two successive generations of their normally reared offspring, through matrilineal transmission. Parallel enhancements of acid-sensing ion channel 1 (ASIC1), ASIC2, and ASIC3 messenger RNA transcripts were detected transgenerationally in central neurons, in the medulla oblongata, and in periaqueductal gray matter of RCF-lineage animals. A single, nebulized dose of the ASIC-antagonist amiloride renormalized respiratory and nociceptive responsiveness across the entire RCF lineage. These findings reveal how, following an early-life adversity, a biological memory reducible to a molecular sensor unfolds, shaping adaptation mechanisms over three generations. Our findings are entwined with multiple correlates of human anxiety and pain conditions and suggest nebulized amiloride as a therapeutic avenue.
Collapse
Affiliation(s)
- Marco Battaglia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Child Youth and Emerging Adult Programme, Centre for Addiction and Mental Health, Toronto, ON, Canada
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| | - Orlane Rossignol
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Louis-Etienne Lorenzo
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Jasmin Deguire
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Antoine G. Godin
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| | - Francesca R. D’Amato
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| |
Collapse
|
12
|
Bishop M, SheikhBahei S. Brainstem astrocytes regulate breathing and may affect arousal state in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559604. [PMID: 37841850 PMCID: PMC10576258 DOI: 10.1101/2023.09.28.559604] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Variations in arousal levels can impact respiratory patterns. However, whether changes in breathing behaviors can influence arousal state is not fully understood. In this study, we investigated the role of astrocytes in the preBötzinger complex (preBötC) in modulating arousal states via breathing in adult conscious rats. Using viral vector tools, we selectively interfered with astrocytic signaling in the preBötC. Rats with inhibited astrocytic signaling exhibited slower breathing rates and behaviors indicative of a calmer state, whereas enhanced purinergic signaling in preBötC astrocytes led to faster breathing and heightened arousal. Our findings reveal a key role for astrocyte-mediated mechanism in the preBötC that influences both respiratory behaviors and higher-order brain functions like arousal, suggesting a bidirectional link between breathing behaviors and mental states.
Collapse
Affiliation(s)
- Mitchell Bishop
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Shahriar SheikhBahei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| |
Collapse
|
13
|
Turk AZ, Millwater M, SheikhBahaei S. Whole-brain analysis of CO 2 chemosensitive regions and identification of the retrotrapezoid and medullary raphé nuclei in the common marmoset ( Callithrix jacchus). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.558361. [PMID: 37986845 PMCID: PMC10659419 DOI: 10.1101/2023.09.26.558361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Respiratory chemosensitivity is an important mechanism by which the brain senses changes in blood partial pressure of CO2 (PCO2). It is proposed that special neurons (and astrocytes) in various brainstem regions play key roles as CO2 central respiratory chemosensors in rodents. Although common marmosets (Callithrix jacchus), New-World non-human primates, show similar respiratory responses to elevated inspired CO2 as rodents, the chemosensitive regions in marmoset brain have not been defined yet. Here, we used c-fos immunostainings to identify brain-wide CO2-activated brain regions in common marmosets. In addition, we mapped the location of the retrotrapezoid nucleus (RTN) and raphé nuclei in the marmoset brainstem based on colocalization of CO2-induced c-fos immunoreactivity with Phox2b, and TPH immunostaining, respectively. Our data also indicated that, similar to rodents, marmoset RTN astrocytes express Phox2b and have complex processes that create a meshwork structure at the ventral surface of medulla. Our data highlight some cellular and structural regional similarities in brainstem of the common marmosets and rodents.
Collapse
Affiliation(s)
- Ariana Z. Turk
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Marissa Millwater
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| |
Collapse
|
14
|
Nguyen TT, Camp CR, Doan JK, Traynelis SF, Sloan SA, Hall RA. GPR37L1 controls maturation and organization of cortical astrocytes during development. Glia 2023; 71:1921-1946. [PMID: 37029775 PMCID: PMC10315172 DOI: 10.1002/glia.24375] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Astrocyte maturation is crucial to proper brain development and function. This maturation process includes the ramification of astrocytic morphology and the establishment of astrocytic domains. While this process has been well-studied, the mechanisms by which astrocyte maturation is initiated are not well understood. GPR37L1 is an astrocyte-specific G protein-coupled receptor (GPCR) that is predominantly expressed in mature astrocytes and has been linked to the modulation of seizure susceptibility in both humans and mice. To investigate the role of GPR37L1 in astrocyte biology, RNA-seq analyses were performed on astrocytes immunopanned from P7 Gpr37L1-/- knockout (L1KO) mouse cortex and compared to those from wild-type (WT) mouse cortex. These RNA-seq studies revealed that pathways involved in central nervous system development were altered and that L1KO cortical astrocytes express lower amounts of mature astrocytic genes compared to WT astrocytes. Immunohistochemical studies of astrocytes from L1KO mouse brain revealed that these astrocytes exhibit overall shorter total process length, and are also less complex and spaced further apart from each other in the mouse cortex. This work sheds light on how GPR37L1 regulates cellular processes involved in the control of astrocyte biology and maturation.
Collapse
Affiliation(s)
| | - Chad R. Camp
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Juleva K. Doan
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Stephen F. Traynelis
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Steven A. Sloan
- Emory University School of Medicine, Department of Human Genetics
| | - Randy A. Hall
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| |
Collapse
|
15
|
Li K, Gonye EC, Stornetta RL, Bayliss CB, Yi G, Stornetta DS, Baca SM, Abbott SB, Guyenet PG, Bayliss DA. The astrocytic Na + -HCO 3 - cotransporter, NBCe1, is dispensable for respiratory chemosensitivity. J Physiol 2023; 601:3667-3686. [PMID: 37384821 PMCID: PMC10528273 DOI: 10.1113/jp284960] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
The interoceptive homeostatic mechanism that controls breathing, blood gases and acid-base balance in response to changes in CO2 /H+ is exquisitely sensitive, with convergent roles proposed for chemosensory brainstem neurons in the retrotrapezoid nucleus (RTN) and their supporting glial cells. For astrocytes, a central role for NBCe1, a Na+ -HCO3 - cotransporter encoded by Slc4a4, has been envisaged in multiple mechanistic models (i.e. underlying enhanced CO2 -induced local extracellular acidification or purinergic signalling). We tested these NBCe1-centric models by using conditional knockout mice in which Slc4a4 was deleted from astrocytes. In GFAP-Cre;Slc4a4fl/fl mice we found diminished expression of Slc4a4 in RTN astrocytes by comparison to control littermates, and a concomitant reduction in NBCe1-mediated current. Despite disrupted NBCe1 function in RTN-adjacent astrocytes from these conditional knockout mice, CO2 -induced activation of RTN neurons or astrocytes in vitro and in vivo, and CO2 -stimulated breathing, were indistinguishable from NBCe1-intact littermates; hypoxia-stimulated breathing and sighs were likewise unaffected. We obtained a more widespread deletion of NBCe1 in brainstem astrocytes by using tamoxifen-treated Aldh1l1-Cre/ERT2;Slc4a4fl/fl mice. Again, there was no difference in effects of CO2 or hypoxia on breathing or on neuron/astrocyte activation in NBCe1-deleted mice. These data indicate that astrocytic NBCe1 is not required for the respiratory responses to these chemoreceptor stimuli in mice, and that any physiologically relevant astrocytic contributions must involve NBCe1-independent mechanisms. KEY POINTS: The electrogenic NBCe1 transporter is proposed to mediate local astrocytic CO2 /H+ sensing that enables excitatory modulation of nearby retrotrapezoid nucleus (RTN) neurons to support chemosensory control of breathing. We used two different Cre mouse lines for cell-specific and/or temporally regulated deletion of the NBCe1 gene (Slc4a4) in astrocytes to test this hypothesis. In both mouse lines, Slc4a4 was depleted from RTN-associated astrocytes but CO2 -induced Fos expression (i.e. cell activation) in RTN neurons and local astrocytes was intact. Likewise, respiratory chemoreflexes evoked by changes in CO2 or O2 were unaffected by loss of astrocytic Slc4a4. These data do not support the previously proposed role for NBCe1 in respiratory chemosensitivity mediated by astrocytes.
Collapse
Affiliation(s)
- Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Elizabeth C. Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Ruth L. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | | | - Grace Yi
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Daniel S. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Serapio M. Baca
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Stephen B.G. Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Patrice G. Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Douglas A. Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| |
Collapse
|
16
|
Siemsen BM, Denton AR, Parrila-Carrero J, Hooker KN, Carpenter EA, Prescot ME, Brock AG, Westphal AM, Leath MN, McFaddin JA, Jhou TC, McGinty JF, Scofield MD. Heroin Self-Administration and Extinction Increase Prelimbic Cortical Astrocyte-Synapse Proximity and Alter Dendritic Spine Morphometrics That Are Reversed by N-Acetylcysteine. Cells 2023; 12:1812. [PMID: 37508477 PMCID: PMC10378353 DOI: 10.3390/cells12141812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/09/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Clinical and preclinical studies indicate that adaptations in corticostriatal neurotransmission significantly contribute to heroin relapse vulnerability. In animal models, heroin self-administration and extinction produce cellular adaptations in both neurons and astrocytes within the nucleus accumbens (NA) core that are required for cue-induced heroin seeking. Specifically, decreased glutamate clearance and reduced association of perisynaptic astrocytic processes with NAcore synapses allow glutamate release from prelimbic (PrL) cortical terminals to engage synaptic and structural plasticity in NAcore medium spiny neurons. Normalizing astrocyte glutamate homeostasis with drugs like the antioxidant N-acetylcysteine (NAC) prevents cue-induced heroin seeking. Surprisingly, little is known about heroin-induced alterations in astrocytes or pyramidal neurons projecting to the NAcore in the PrL cortex (PrL-NAcore). Here, we observe functional adaptations in the PrL cortical astrocyte following heroin self-administration (SA) and extinction as measured by the electrophysiologically evoked plasmalemmal glutamate transporter 1 (GLT-1)-dependent current. We likewise observed the increased complexity of the glial fibrillary acidic protein (GFAP) cytoskeletal arbor and increased association of the astrocytic plasma membrane with synaptic markers following heroin SA and extinction training in the PrL cortex. Repeated treatment with NAC during extinction reversed both the enhanced astrocytic complexity and synaptic association. In PrL-NAcore neurons, heroin SA and extinction decreased the apical tuft dendritic spine density and enlarged dendritic spine head diameter in male Sprague-Dawley rats. Repeated NAC treatment during extinction prevented decreases in spine density but not dendritic spine head expansion. Moreover, heroin SA and extinction increased the co-registry of the GluA1 subunit of AMPA receptors in both the dendrite shaft and spine heads of PrL-NAcore neurons. Interestingly, the accumulation of GluA1 immunoreactivity in spine heads was further potentiated by NAC treatment during extinction. Finally, we show that the NAC treatment and elimination of thrombospondin 2 (TSP-2) block cue-induced heroin relapse. Taken together, our data reveal circuit-level adaptations in cortical dendritic spine morphology potentially linked to heroin-induced alterations in astrocyte complexity and association at the synapses. Additionally, these data demonstrate that NAC reverses PrL cortical heroin SA-and-extinction-induced adaptations in both astrocytes and corticostriatal neurons.
Collapse
Affiliation(s)
- Benjamin M. Siemsen
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Adam R. Denton
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Kaylee N. Hooker
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eilish A. Carpenter
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Meagan E. Prescot
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ashley G. Brock
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Annaka M. Westphal
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mary-Nan Leath
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John A. McFaddin
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thomas C. Jhou
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jacqueline F. McGinty
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D. Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
17
|
Zhou LT, Liu D, Kang HC, Lu L, Huang HZ, Ai WQ, Zhou Y, Deng MF, Li H, Liu ZQ, Zhang WF, Hu YZ, Han ZT, Zhang HH, Jia JJ, Sarkar AK, Sharaydeh S, Wang J, Man HY, Schilling M, Bertram L, Lu Y, Guo Z, Zhu LQ. Tau pathology epigenetically remodels the neuron-glial cross-talk in Alzheimer's disease. SCIENCE ADVANCES 2023; 9:eabq7105. [PMID: 37083538 PMCID: PMC10121173 DOI: 10.1126/sciadv.abq7105] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
The neuron-glia cross-talk is critical to brain homeostasis and is particularly affected by neurodegenerative diseases. How neurons manipulate the neuron-astrocyte interaction under pathological conditions, such as hyperphosphorylated tau, a pathological hallmark in Alzheimer's disease (AD), remains elusive. In this study, we identified excessively elevated neuronal expression of adenosine receptor 1 (Adora1 or A1R) in 3×Tg mice, MAPT P301L (rTg4510) mice, patients with AD, and patient-derived neurons. The up-regulation of A1R was found to be tau pathology dependent and posttranscriptionally regulated by Mef2c via miR-133a-3p. Rebuilding the miR-133a-3p/A1R signal effectively rescued synaptic and memory impairments in AD mice. Furthermore, neuronal A1R promoted the release of lipocalin 2 (Lcn2) and resulted in astrocyte activation. Last, silencing neuronal Lcn2 in AD mice ameliorated astrocyte activation and restored synaptic plasticity and learning/memory. Our findings reveal that the tau pathology remodels neuron-glial cross-talk and promotes neurodegenerative progression. Approaches targeting A1R and modulating this signaling pathway might be a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Lan-Ting Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, Hubei 441053, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui-Cong Kang
- Department of Neurology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - He-Zhou Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Qing Ai
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Man-Fei Deng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhi-Qiang Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei-Feng Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ya-Zhuo Hu
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Zhi-Tao Han
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Hong-Hong Zhang
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Jian-Jun Jia
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Saldin Sharaydeh
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Marcel Schilling
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck 23562, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck 23562, Germany
| | - Youming Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
18
|
Bondi H, Chiazza F, Masante I, Bortolotto V, Canonico PL, Grilli M. Heterogenous response to aging of astrocytes in murine Substantia Nigra pars compacta and pars reticulata. Neurobiol Aging 2023; 123:23-34. [PMID: 36630756 DOI: 10.1016/j.neurobiolaging.2022.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Currently, little is known about the impact of aging on astrocytes in substantia nigra pars compacta (SNpc), where dopaminergic neurons degenerate both in physiological aging and in Parkinson's disease, an age-related neurodegenerative disorder. We performed a morphometric analysis of GFAP+ astrocytes in SNpc and, for comparison, in the pars reticulata (SNpr) of young (4-6 months), middle-aged (14-17 months) and old (20-24 months) C57BL/6J male mice. We demonstrated an age-dependent increase of structural complexity only in astrocytes localized in SNpc, and not in SNpr. Astrocytic structural remodelling was not accompanied by changes in GFAP expression, while GFAP increased in SNpr of old compared to young mice. In parallel, transcript levels of selected astrocyte-enriched genes were evaluated. With aging, decreased GLT1 expression occurred only in SNpc, while xCT transcript increased both in SNpc and SNpr, suggesting a potential loss of homeostatic control of extracellular glutamate only in the subregion where age-dependent neurodegeneration occurs. Altogether, our results support an heterogenous morphological and biomolecular response to aging of GFAP+ astrocytes in SNpc and SNpr.
Collapse
Affiliation(s)
- Heather Bondi
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Fausto Chiazza
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Irene Masante
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valeria Bortolotto
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.
| |
Collapse
|
19
|
Wiese BM, Alvarez Reyes A, Vanderah TW, Largent-Milnes TM. The endocannabinoid system and breathing. Front Neurosci 2023; 17:1126004. [PMID: 37144090 PMCID: PMC10153446 DOI: 10.3389/fnins.2023.1126004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
Recent changes in cannabis accessibility have provided adjunct therapies for patients across numerous disease states and highlights the urgency in understanding how cannabinoids and the endocannabinoid (EC) system interact with other physiological structures. The EC system plays a critical and modulatory role in respiratory homeostasis and pulmonary functionality. Respiratory control begins in the brainstem without peripheral input, and coordinates the preBötzinger complex, a component of the ventral respiratory group that interacts with the dorsal respiratory group to synchronize burstlet activity and drive inspiration. An additional rhythm generator: the retrotrapezoid nucleus/parafacial respiratory group drives active expiration during conditions of exercise or high CO2. Combined with the feedback information from the periphery: through chemo- and baroreceptors including the carotid bodies, the cranial nerves, stretch of the diaphragm and intercostal muscles, lung tissue, and immune cells, and the cranial nerves, our respiratory system can fine tune motor outputs that ensure we have the oxygen necessary to survive and can expel the CO2 waste we produce, and every aspect of this process can be influenced by the EC system. The expansion in cannabis access and potential therapeutic benefits, it is essential that investigations continue to uncover the underpinnings and mechanistic workings of the EC system. It is imperative to understand the impact cannabis, and exogenous cannabinoids have on these physiological systems, and how some of these compounds can mitigate respiratory depression when combined with opioids or other medicinal therapies. This review highlights the respiratory system from the perspective of central versus peripheral respiratory functionality and how these behaviors can be influenced by the EC system. This review will summarize the literature available on organic and synthetic cannabinoids in breathing and how that has shaped our understanding of the role of the EC system in respiratory homeostasis. Finally, we look at some potential future therapeutic applications the EC system has to offer for the treatment of respiratory diseases and a possible role in expanding the safety profile of opioid therapies while preventing future opioid overdose fatalities that result from respiratory arrest or persistent apnea.
Collapse
Affiliation(s)
- Beth M. Wiese
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Angelica Alvarez Reyes
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Tally M. Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- *Correspondence: Tally M. Largent-Milnes,
| |
Collapse
|
20
|
Fabres RB, Nunes RR, de Medeiros de Mattos M, Andrade MKG, Martini APR, Tassinari ID, Sanches EF, de Fraga LS, Netto CA. Therapeutic hypothermia for the treatment of neonatal hypoxia-ischemia: sex-dependent modulation of reactive astrogliosis. Metab Brain Dis 2022; 37:2315-2329. [PMID: 35778625 DOI: 10.1007/s11011-022-01030-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022]
Abstract
Therapeutic hypothermia (TH) is the standard treatment for neonatal hypoxia-ischemia (HI) with a time window limited up to 6 h post injury. However, influence of sexual dimorphism in the therapeutic window for TH has not yet been elucidated in animal models of HI. Therefore, the aim of this study was to investigate the most effective time window to start TH in male and female rats submitted to neonatal HI. Wistar rats (P7) were divided into the following groups: NAÏVE and SHAM (control groups), HI (submitted to HI) and TH (submitted to HI and TH; 32ºC for 5 h). TH was started at 2 h (TH-2 h group), 4 h (TH-4 h group), or 6 h (TH-6 h group) after HI. At P14, animals were subjected to behavioural tests, volume of lesion and reactive astrogliosis assessments. Male and female rats from the TH-2 h group showed reduction in the latency of behavioral tests, and decrease in volume of lesion and intensity of GFAP immunofluorescence. TH-2 h females also showed reduction of degenerative cells and morphological changes in astrocytes. Interestingly, females from the TH-6 h group showed an increase in volume of lesion and in number of degenerative hippocampal cells, associated with worse behavioral performance. Together, these results indicate that TH neuroprotection is time- and sex-dependent. Moreover, TH started later (6 h) can worsen volume of brain lesion in females. These data indicate the need to develop specific therapeutic protocols for each sex and reinforce the importance of early onset of the hypothermic treatment.
Collapse
Affiliation(s)
- Rafael Bandeira Fabres
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil.
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil.
- ICBS/UFRGS - Campus Centro, Rua Sarmento Leite, 500 - 2º Andar, 90050170, Porto Alegre, RS, Brazil.
| | - Ricardo Ribeiro Nunes
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Marcel de Medeiros de Mattos
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
| | - Mirella Kielek Galvan Andrade
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Ana Paula Rodrigues Martini
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Eduardo Farias Sanches
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| |
Collapse
|
21
|
Vuorimaa M, Kareinen I, Toivanen P, Karlsson S, Ruohonen S. Deep Learning-Based Segmentation of Morphologically Distinct Rat Hippocampal Reactive Astrocytes After Trimethyltin Exposure. Toxicol Pathol 2022; 50:754-762. [PMID: 36125102 DOI: 10.1177/01926233221124497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As regulators of homeostasis, astrocytes undergo morphological changes after injury to limit the insult in central nervous system (CNS). Trimethyltin (TMT) is a known neurotoxicant that induces reactive astrogliosis in rat CNS. To evaluate the degree of reactive astrogliosis, the assessment relies on manual counting or semiquantitative scoring. We hypothesized that deep learning algorithm could be used to identify the grade of reactive astrogliosis in immunoperoxidase-stained sections in a quantitative manner. The astrocyte algorithm was created using a commercial supervised deep learning platform and the used training set consisted of 940 astrocytes manually annotated from hippocampus and cortex. Glial fibrillary acidic protein-labeled brain sections of rat TMT model were analyzed for astrocytes with the trained algorithm. Algorithm was able to count the number of individual cells, cell areas, and circumferences. The astrocyte algorithm identified astrocytes with varying sizes from immunostained sections with high confidence. Algorithm analysis data revealed a novel morphometric marker based on cell area and circumference. This marker correlated with the time-dependent progression of the neurotoxic profile of TMT. This study highlights the potential of using novel deep learning-based image analysis tools in neurotoxicity and pharmacology studies.
Collapse
|
22
|
Gourine AV, Dale N. Brain H + /CO 2 sensing and control by glial cells. Glia 2022; 70:1520-1535. [PMID: 35102601 DOI: 10.1002/glia.24152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 01/04/2023]
Abstract
Maintenance of constant brain pH is critically important to support the activity of individual neurons, effective communication within the neuronal circuits, and, thus, efficient processing of information by the brain. This review article focuses on how glial cells detect and respond to changes in brain tissue pH and concentration of CO2 , and then trigger systemic and local adaptive mechanisms that ensure a stable milieu for the operation of brain circuits. We give a detailed account of the cellular and molecular mechanisms underlying sensitivity of glial cells to H+ and CO2 and discuss the role of glial chemosensitivity and signaling in operation of three key mechanisms that work in concert to keep the brain pH constant. We discuss evidence suggesting that astrocytes and marginal glial cells of the brainstem are critically important for central respiratory CO2 chemoreception-a fundamental physiological mechanism that regulates breathing in accord with changes in blood and brain pH and partial pressure of CO2 in order to maintain systemic pH homeostasis. We review evidence suggesting that astrocytes are also responsible for the maintenance of local brain tissue extracellular pH in conditions of variable acid loads associated with changes in the neuronal activity and metabolism, and discuss potential role of these glial cells in mediating the effects of CO2 on cerebral vasculature.
Collapse
Affiliation(s)
- Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
23
|
L. F. Nascimento A, O. S. Medeiros P, F. A. T. Pedrão L, Queiroz VC, Oliveira LM, Novaes LS, Caetano AL, Munhoz CD, Takakura AC, Falquetto B. Oxidative stress inhibition via apocynin prevents contributes to medullary respiratory neurodegeneration and respiratory pattern dysfunction in 6-OHDA animal model of Parkinson's disease. Neuroscience 2022; 502:91-106. [DOI: 10.1016/j.neuroscience.2022.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/15/2022]
|
24
|
Oliveira LM, Fernandes-Junior SA, Cabral LMC, Miranda NCS, Czeisler CM, Otero JJ, Moreira TS, Takakura AC. Regulation of blood vessels by ATP in the ventral medullary surface in a rat model of Parkinson's disease. Brain Res Bull 2022; 187:138-154. [PMID: 35777704 DOI: 10.1016/j.brainresbull.2022.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) patients often experience impairment of autonomic and respiratory functions. These include conditions such as orthostatic hypotension and sleep apnea, which are highly correlated with dysfunctional central chemoreception. Blood flow is a fundamental determinant of tissue CO2/H+, yet the extent to which blood flow regulation within chemoreceptor regions contributes to respiratory behavior during neurological disease remains unknown. Here, we tested the hypothesis that 6-hydroxydopamine injection to inducing a known model of PD results in dysfunctional vascular homeostasis, biochemical dysregulation, and glial morphology of the ventral medullary surface (VMS). We show that hypercapnia (FiCO2 = 10%) induced elevated VMS pial vessel constriction in PD animals through a P2-receptor dependent mechanism. Similarly, we found a greater CO2-induced vascular constriction after ARL67156 (an ectonucleotidase inhibitor) in control and PD-induced animals. In addition, we also report that weighted gene correlational network analysis of the proteomic data showed a protein expression module differentially represented between both groups. This module showed that gene ontology enrichment for components of the ATP machinery were reduced in our PD-model compared to control animals. Altogether, our data indicate that dysfunction in purinergic signaling, potentially through altered ATP bioavailability in the VMS region, may compromise the RTN neuroglial vascular unit in a PD animal model.
Collapse
Affiliation(s)
- Luiz M Oliveira
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Silvio A Fernandes-Junior
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil; The Ohio State University College of Medicine, Department of Pathology, USA
| | - Laís M C Cabral
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Nicole C S Miranda
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | | | - José J Otero
- The Ohio State University College of Medicine, Department of Pathology, USA
| | - Thiago S Moreira
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Ana C Takakura
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
25
|
Turk AZ, Bishop M, Adeck A, SheikhBahaei S. Astrocytic modulation of central pattern generating motor circuits. Glia 2022; 70:1506-1519. [PMID: 35212422 DOI: 10.1002/glia.24162] [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: 08/13/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/26/2022]
Abstract
Central pattern generators (CPGs) generate the rhythmic and coordinated neural features necessary for the proper conduction of complex behaviors. In particular, CPGs are crucial for complex motor behaviors such as locomotion, mastication, respiration, and vocal production. While the importance of these networks in modulating behavior is evident, the mechanisms driving these CPGs are still not fully understood. On the other hand, accumulating evidence suggests that astrocytes have a significant role in regulating the function of some of these CPGs. Here, we review the location, function, and role of astrocytes in locomotion, respiration, and mastication CPGs and propose that, similarly, astrocytes may also play a significant role in the vocalization CPG.
Collapse
Affiliation(s)
- Ariana Z Turk
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Mitchell Bishop
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Afuh Adeck
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
26
|
Smith JC. Respiratory rhythm and pattern generation: Brainstem cellular and circuit mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:1-35. [PMID: 35965022 DOI: 10.1016/b978-0-323-91534-2.00004-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Breathing movements in mammals are driven by rhythmic neural activity automatically generated within spatially and functionally organized brainstem neural circuits comprising the respiratory central pattern generator (CPG). This chapter reviews up-to-date experimental information and theoretical studies of the cellular and circuit mechanisms of respiratory rhythm and pattern generation operating within critical components of this CPG in the lower brainstem. Over the past several decades, there have been substantial advances in delineating the spatial architecture of essential medullary regions and their regional cellular and circuit properties required to understand rhythm and pattern generation mechanisms. A fundamental concept is that the circuits in these regions have rhythm-generating capabilities at multiple cellular and circuit organization levels. The regional cellular properties, circuit organization, and control mechanisms allow flexible expression of neural activity patterns for a repertoire of respiratory behaviors under various physiologic conditions that are dictated by requirements for homeostatic regulation and behavioral integration. Many mechanistic insights have been provided by computational modeling studies driven by experimental results and have advanced understanding in the field. These conceptual and theoretical developments are discussed.
Collapse
Affiliation(s)
- Jeffrey C Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
27
|
Chappus-McCendie H, Poulin MA, Chouinard-Watkins R, Vandal M, Calon F, Lauzon MA, Plourde M. A diet rich in docosahexaenoic acid enhances reactive astrogliosis and ramified microglia morphology in apolipoprotein E epsilon 4-targeted replacement mice. AGING BRAIN 2022; 2:100046. [PMID: 36908881 PMCID: PMC9997137 DOI: 10.1016/j.nbas.2022.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 10/16/2022] Open
Abstract
Docosahexaenoic acid (DHA) consumption reduces spatial memory impairment in mice carrying the human apolipoprotein E ε4 (APOE4) allele. The current study evaluated whether astrocyte and microglia morphology contribute to the mechanism of this result. APOE3 and APOE4 mice were fed either a DHA-enriched diet or a control diet from 4 to 12 months of age. Coronal brain sections were immunostained for GFAP, Iba1, and NeuN. Astrocytes from APOE4 mice exhibited signs of reactive astrogliosis compared to APOE3 mice. Consumption of DHA exacerbated reactive astrocyte morphology in APOE4 carriers. Microglia from APOE4-control mice exhibited characteristics of amoeboid morphology and other characteristics of ramified morphology (more processes, greater process complexity, and greater distance between neighboring microglia). DHA enhanced ramified microglia morphology in APOE4 mice. In addition, APOE4 mice fed the DHA diet had lower hippocampal concentrations of interleukin-7, lipopolysaccharide-induced CXC chemokine and monocyte chemoattractant protein 1, and higher concentration of interferon-gamma compared to APOE4-control mice. Our results indicate that a diet rich in DHA enhances reactive astrogliosis and ramified microglia morphology in APOE4 mice.
Collapse
Affiliation(s)
- Hillary Chappus-McCendie
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marc-Antoine Poulin
- Département de génie chimique et de génie biotechnologique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Raphaël Chouinard-Watkins
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada
| | - Milène Vandal
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Faculté de pharmacie et centre de recherche du CHU de Québec-Université Laval, QC, Canada
| | - Frédéric Calon
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Faculté de pharmacie et centre de recherche du CHU de Québec-Université Laval, QC, Canada
| | - Marc-Antoine Lauzon
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de génie chimique et de génie biotechnologique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
28
|
Adebiyi OE, Omobowale TO, Abatan MO. Neurocognitive domains and neuropathological changes in experimental infection with Trypanosoma brucei brucei in Wistar rats. Heliyon 2021; 7:e08260. [PMID: 34765779 PMCID: PMC8571699 DOI: 10.1016/j.heliyon.2021.e08260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/29/2021] [Accepted: 10/22/2021] [Indexed: 01/05/2023] Open
Abstract
Trypanosoma brucei brucei causes animal trypanosomiasis in several vertebrates and human African trypanosomiasis. Previous studies have only explored the incidence, clinical symptoms, haematology and biochemical changes associated with the disease. The behavioral manipulation hypothesis posits that parasites alter the behavior of host to increase the reproductive abilities of such parasites. Hence, the present study was carried out to investigate changes in behavior and cognition following experimental infection of T. brucei brucei in rat model. This study involved two groups of animals (uninfected control and T. brucei infected) with 8 rats per group. After confirmation of parasitaemia in the infected rats both groups were assessed to investigate if infection led to behavioral alterations and neuropathological changes using the open field, social interaction and forelimb suspension tests. Immunohistochemistry was performed on brain tissues using glial fibrillary acidic protein and anticalbindin-D28k, antibodies. We demonstrated that T. brucei infection triggered a significant decrease in exploratory activity, anxiety-like behavior, altered recognition of social novelty and reduced hanging latency in the hanging wire test. Immunohistochemistry revealed significant astrocytosis, loss of dendritic spines and reduction of Purkinje cell layer of the cerebellum. These results demonstrate that T. brucei infection induce signs of anxiety, impaired motor co-ordination with degeneration and loss of Purkinje cells.
Collapse
Affiliation(s)
| | | | - Mathew Oluwole Abatan
- Department of Veterinary Pharmacology and Toxicology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
29
|
Oliveira LM, Takakura AC, Moreira TS. Forebrain and Hindbrain Projecting-neurons Target the Post-inspiratory Complex Cholinergic Neurons. Neuroscience 2021; 476:102-115. [PMID: 34582982 DOI: 10.1016/j.neuroscience.2021.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023]
Abstract
The postinspiratory complex (PiCo) is a region located in the ventromedial medulla involved with the post-inspiratory activity. PiCo neurons are excitatory (VGlut2+) and express the enzyme choline acetyl transferase (ChAT+). Evidence also suggests that PiCo is coupled to two additional groups of neurons involved in breathing process, i.e. the pre-Bötzinger complex (preBötC, inspiration) and the retrotrapezoid nucleus (RTN, active expiration), composing all together, the hypothesized triple respiratory oscillator. Here, our main objective is to demonstrate the afferent connections to PiCo region. We mapped projecting-neurons to PiCo by injecting Fluorogold (FG) retrograde tracer into the PiCo of adult Long-Evans Chat-cre male rats. We reported extensive projections from periaqueductal grey matter and Kölliker-Fuse regions and mild projections from the nucleus of the solitary tract, ventrolateral medulla and hypothalamus. We also injected a cre-dependent vector expressing channelrhodopsin 2 (AAV5-ChR2) fused with enhanced mCherry into the PiCo of ChAT-cre rats to optogenetic activate those neurons and investigate the role of PiCo for inspiratory/postinspiratory activity. Both in urethane-anesthetized and unrestrained conscious rats the response of ChR2-transduced neurons to light induced an increase in postinspiratory activity. Our data confirmed that PiCo seems to be dedicated to postinspiratory activity and represent a site of integration for autonomic and motor components of respiratory and non-respiratory pathways.
Collapse
Affiliation(s)
- Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, 05508-000 Sao Paulo, SP, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, 05508-000 Sao Paulo, SP, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, 05508-000 Sao Paulo, SP, Brazil.
| |
Collapse
|
30
|
Patodia S, Somani A, Thom M. Review: Neuropathology findings in autonomic brain regions in SUDEP and future research directions. Auton Neurosci 2021; 235:102862. [PMID: 34411885 PMCID: PMC8455454 DOI: 10.1016/j.autneu.2021.102862] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022]
Abstract
Autonomic dysfunction is implicated from clinical, neuroimaging and experimental studies in sudden and unexpected death in epilepsy (SUDEP). Neuropathological analysis in SUDEP series enable exploration of acquired, seizure-related cellular adaptations in autonomic and brainstem autonomic centres of relevance to dysfunction in the peri-ictal period. Alterations in SUDEP compared to control groups have been identified in the ventrolateral medulla, amygdala, hippocampus and central autonomic regions. These involve neuropeptidergic, serotonergic and adenosine systems, as well as specific regional astroglial and microglial populations, as potential neuronal modulators, orchestrating autonomic dysfunction. Future research studies need to extend to clinically and genetically characterized epilepsies, to explore if common or distinct pathways of autonomic dysfunction mediate SUDEP. The ultimate objective of SUDEP research is the identification of disease biomarkers for at risk patients, to improve post-mortem recognition and disease categorisation, but ultimately, for exposing potential treatment targets of pharmacologically modifiable and reversible cellular alterations.
Collapse
Affiliation(s)
- Smriti Patodia
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alyma Somani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.
| |
Collapse
|
31
|
Gourine AV, Spyer KM. Geoff Burnstock, purinergic signalling, and chemosensory control of breathing. Auton Neurosci 2021; 235:102839. [PMID: 34198056 DOI: 10.1016/j.autneu.2021.102839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/11/2021] [Accepted: 06/20/2021] [Indexed: 12/14/2022]
Abstract
This article is the authors' contribution to the tribute issue in honour of Geoffrey Burnstock, the founder of this journal and the field of purinergic signalling. We give a brief account of the results of experimental studies which at the beginning received valuable input from Geoff, who both directly and indirectly influenced our research undertaken over the last two decades. Research into the mechanisms controlling breathing identified ATP as the common mediator of the central and peripheral chemosensory transduction. Studies of the sources and mechanisms of chemosensory ATP release in the CNS suggested that this signalling pathway is universally engaged in conditions of increased metabolic demand by brain glial cells - astrocytes. Astrocytes appear to function as versatile CNS metabolic sensors that detect changes in brain tissue pH, CO2, oxygen, and cerebral perfusion pressure. Experimental studies on various aspects of astrocyte biology generated data indicating that the function of these omnipresent glial cells and communication between astrocytes and neurons are governed by purinergic signalling, - first discovered by Geoff Burnstock in the 70's and researched through his entire scientific career.
Collapse
Affiliation(s)
- Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| | - K Michael Spyer
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
32
|
Turk AZ, SheikhBahaei S. Morphometric analysis of astrocytes in vocal production circuits of common marmoset (Callithrix jacchus). J Comp Neurol 2021; 530:574-589. [PMID: 34387357 PMCID: PMC8716418 DOI: 10.1002/cne.25230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/10/2022]
Abstract
Astrocytes, the star-shaped glial cells, are the most abundant non-neuronal cell population in the central nervous system. They play a key role in modulating activities of neural networks, including those involved in complex motor behaviors. Common marmosets (Callithrix jacchus), the most vocal non-human primate (NHP), have been used to study the physiology of vocalization and social vocal production. However, the neural circuitry involved in vocal production is not fully understood. In addition, even less is known about the involvement of astrocytes in this circuit. To understand the role, that astrocytes may play in the complex behavior of vocalization, the initial step may be to study their structural properties in the cortical and subcortical regions that are known to be involved in vocalization. Here, in the common marmoset, we identify all astrocytic subtypes seen in other primate's brains, including intralaminar astrocytes. In addition, we reveal detailed structural characteristics of astrocytes and perform morphometric analysis of astrocytes residing in the cortex and midbrain regions that are associated with vocal production. We found that cortical astrocytes in these regions illustrate a higher level of complexity when compared to those in the midbrain. We hypothesize that this complexity that is expressed in cortical astrocytes may reflect their functions to meet the metabolic/structural needs of these regions.
Collapse
Affiliation(s)
- Ariana Z Turk
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Turk AZ, Lotfi Marchoubeh M, Fritsch I, Maguire GA, SheikhBahaei S. Dopamine, vocalization, and astrocytes. BRAIN AND LANGUAGE 2021; 219:104970. [PMID: 34098250 PMCID: PMC8260450 DOI: 10.1016/j.bandl.2021.104970] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 05/06/2023]
Abstract
Dopamine, the main catecholamine neurotransmitter in the brain, is predominately produced in the basal ganglia and released to various brain regions including the frontal cortex, midbrain and brainstem. Dopamine's effects are widespread and include modulation of a number of voluntary and innate behaviors. Vigilant regulation and modulation of dopamine levels throughout the brain is imperative for proper execution of motor behaviors, in particular speech and other types of vocalizations. While dopamine's role in motor circuitry is widely accepted, its unique function in normal and abnormal speech production is not fully understood. In this perspective, we first review the role of dopaminergic circuits in vocal production. We then discuss and propose the conceivable involvement of astrocytes, the numerous star-shaped glia cells of the brain, in the dopaminergic network modulating normal and abnormal vocal productions.
Collapse
Affiliation(s)
- Ariana Z Turk
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Mahsa Lotfi Marchoubeh
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, 72701 AR, USA
| | - Ingrid Fritsch
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, 72701 AR, USA
| | - Gerald A Maguire
- Department of Psychiatry and Neuroscience, School of Medicine, University of California, Riverside, 92521 CA, USA
| | - Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA.
| |
Collapse
|
34
|
Van Horn MR, Benfey NJ, Shikany C, Severs LJ, Deemyad T. Neuron-astrocyte networking: astrocytes orchestrate and respond to changes in neuronal network activity across brain states and behaviors. J Neurophysiol 2021; 126:627-636. [PMID: 34259027 DOI: 10.1152/jn.00062.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Astrocytes are known to play many important roles in brain function. However, research underscoring the extent to which astrocytes modulate neuronal activity is still underway. Here we review the latest evidence regarding the contribution of astrocytes to neuronal oscillations across the brain, with a specific focus on how astrocytes respond to changes in brain state (e.g., sleep, arousal, stress). We then discuss the general mechanisms by which astrocytes signal to neurons to modulate neuronal activity, ultimately driving changes in behavior, followed by a discussion of how astrocytes contribute to respiratory rhythms in the medulla. Finally, we contemplate the possibility that brain stem astrocytes could modulate brainwide oscillations by communicating the status of oxygenation to higher cortical areas.
Collapse
Affiliation(s)
- Marion R Van Horn
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Nicholas J Benfey
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Colleen Shikany
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Liza J Severs
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Physiology and Biophysics, The University of Washington, Seattle, Washington
| | - Tara Deemyad
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Oliveira LM, Baertsch NA, Moreira TS, Ramirez JM, Takakura AC. Unraveling the Mechanisms Underlying Irregularities in Inspiratory Rhythm Generation in a Mouse Model of Parkinson's Disease. J Neurosci 2021; 41:4732-4747. [PMID: 33863785 PMCID: PMC8260248 DOI: 10.1523/jneurosci.2114-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder anatomically characterized by a progressive loss of dopaminergic neurons in the substantia nigra compacta (SNpc). Much less known, yet clinically very important, are the detrimental effects on breathing associated with this disease. Consistent with the human pathophysiology, the 6-hydroxydopamine hydrochloride (6-OHDA) rodent model of PD shows reduced respiratory frequency (fR) and NK1r-immunoreactivity in the pre-Bötzinger complex (preBötC) and PHOX2B+ neurons in the retrotrapezoid nucleus (RTN). To unravel mechanisms that underlie bradypnea in PD, we employed a transgenic approach to label or stimulate specific neuron populations in various respiratory-related brainstem regions. PD mice were characterized by a pronounced decreased number of putatively rhythmically active excitatory neurons in the preBötC and adjacent ventral respiratory column (VRC). Specifically, the number of Dbx1 and Vglut2 neurons was reduced by 47.6% and 17.3%, respectively. By contrast, inhibitory Vgat+ neurons in the VRC, as well as neurons in other respiratory-related brainstem regions, showed relatively minimal or no signs of neuronal loss. Consistent with these anatomic observations, optogenetic experiments identified deficits in respiratory function that were specific to manipulations of excitatory (Dbx1/Vglut2) neurons in the preBötC. We conclude that the decreased number of this critical population of respiratory neurons is an important contributor to the development of irregularities in inspiratory rhythm generation in this mouse model of PD.SIGNIFICANCE STATEMENT We found a decreased number of a specific population of medullary neurons which contributes to breathing abnormalities in a mouse model of Parkinson's disease (PD).
Collapse
Affiliation(s)
- Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
- Department of Pediatrics, University of Washington, Seattle, Washington 98101
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
- Department of Pediatrics, University of Washington, Seattle, Washington 98101
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
| |
Collapse
|
36
|
Martinez D, Kline DD. The role of astrocytes in the nucleus tractus solitarii in maintaining central control of autonomic function. Am J Physiol Regul Integr Comp Physiol 2021; 320:R418-R424. [PMID: 33439770 PMCID: PMC8238142 DOI: 10.1152/ajpregu.00254.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022]
Abstract
The nucleus tractus solitarii (nTS) is the first central site for the termination and integration of autonomic and respiratory sensory information. Sensory afferents terminating in the nTS as well as the embedded nTS neurocircuitry release and utilize glutamate that is critical for maintenance of baseline cardiorespiratory parameters and initiating cardiorespiratory reflexes, including those activated by bouts of hypoxia. nTS astrocytes contribute to synaptic and neuronal activity through a variety of mechanisms, including gliotransmission and regulation of glutamate in the extracellular space via membrane-bound transporters. Here, we aim to highlight recent evidence for the role of astrocytes within the nTS and their regulation of autonomic and cardiorespiratory processes under normal and hypoxic conditions.
Collapse
Affiliation(s)
- Diana Martinez
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
37
|
Bondi H, Bortolotto V, Canonico PL, Grilli M. Complex and regional-specific changes in the morphological complexity of GFAP + astrocytes in middle-aged mice. Neurobiol Aging 2021; 100:59-71. [PMID: 33493951 DOI: 10.1016/j.neurobiolaging.2020.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/14/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023]
Abstract
During aging, alterations in astrocyte phenotype occur in areas associated with age-related cognitive decline, including hippocampus. Previous work reported subregion-specific changes in surface, volume, and soma size of hippocampal astrocytes during physiological aging. Herein we extensively analyzed, by morphometric analysis, fine morphological features of GFAP+ astrocytes in young (6-month-old) and middle-aged (14-month-old) male mice. We observed remarkable heterogeneity in the astrocytic response to aging in distinct subfields and along the dorsoventral axis of hippocampus and in entorhinal cortex. In middle-aged mice dorsal granule cell and molecular layers, but not hilus, astrocytes underwent remarkable increase in their morphological complexity. These changes were absent in ventral Dentate Gyrus (DG). In addition, in entorhinal cortex, the major input to dorsal DG, astrocytes underwent remarkable atrophic changes in middle-aged mice. Since dorsal DG, and not ventral DG, is involved in cognitive functions, these findings appear worth of further evaluation. Our findings also suggest an additional level of complexity in the structural changes associated with brain aging.
Collapse
Affiliation(s)
- Heather Bondi
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valeria Bortolotto
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.
| |
Collapse
|
38
|
Maguire GA, Yoo BR, SheikhBahaei S. Investigation of Risperidone Treatment Associated With Enhanced Brain Activity in Patients Who Stutter. Front Neurosci 2021; 15:598949. [PMID: 33642973 PMCID: PMC7906995 DOI: 10.3389/fnins.2021.598949] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Stuttering is a childhood onset fluency disorder that leads to impairment in speech. A randomized, double-blinded placebo-controlled study was conducted with 10 adult subjects to observe the effects of risperidone (a dopamine receptor 2/serotonin receptor 2 antagonist) on brain metabolism, using [18F] deoxyglucose as the marker. At baseline and after 6 weeks of taking risperidone (0.5–2.0 mg/day) or a placebo pill, participants were assigned to a solo reading aloud task for 30 min and subsequently underwent a 90-min positron emission tomography scan. Paired t-tests were performed to compare the pre-treatment vs. post-treatment in groups. After imaging and analysis, the blind was broken, which revealed an equal number of subjects of those on risperidone and those on placebo. There were no significant differences in the baseline scans taken before medication randomization. However, scans taken after active treatment demonstrated higher glucose uptake in the specific regions of the brain for those in the risperidone treatment group (p < 0.05). Risperidone treatment was associated with increased metabolism in the left striatum, which consists of the caudate and putamen, and the Broca’s area. The current study strengthens previous research that suggests the role of elevated dopamine activity and striatal hypometabolism in stuttering. We propose that the mechanism of risperidone’s action in stuttering, in part, involves increased metabolism of striatal astrocytes. We conclude that using neuroimaging techniques to visualize changes in the brain of those who stutter can provide valuable insights into the pathophysiology of the disorder and guide the development of future interventions.
Collapse
Affiliation(s)
- Gerald A Maguire
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Bo Ram Yoo
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Shahriar SheikhBahaei
- National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
Du Preez A, Onorato D, Eiben I, Musaelyan K, Egeland M, Zunszain PA, Fernandes C, Thuret S, Pariante CM. Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice. Brain Behav Immun 2021; 91:24-47. [PMID: 32755644 DOI: 10.1016/j.bbi.2020.07.015] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
Unpredictable chronic mild stress (UCMS) is one of the most commonly used, robust and translatable models for studying the neurobiological basis of major depression. Although the model currently has multiple advantages, it does not entirely follow the trajectory of the disorder, whereby depressive symptomology can often present months after exposure to stress. Furthermore, patients with depression are more likely to withdraw in response to their stressful experience, or as a symptom of their depression, and, in turn, this withdrawal/isolation can further exacerbate the stressful experience and the depressive symptomology. Therefore, we investigated the effect(s) of 6 weeks of UCMS followed by another 6 weeks of social isolation (referred to as UCMSI), on behaviour, corticosterone stress responsivity, immune system functioning, and hippocampal neurogenesis, in young adult male mice. We found that UCMSI induced several behavioural changes resembling depression but did not induce peripheral inflammation. However, UCMSI animals showed increased microglial activation in the ventral dentate gyrus (DG) of the hippocampus and astrocyte activation in both the dorsal and ventral DG, with increased GFAP-positive cell immunoreactivity, GFAP-positive cell hypertrophy and process extension, and increased s100β-positive cell density. Moreover, UCMSI animals had significantly reduced neurogenesis in the DG and reduced levels of peripheral vascular endothelial growth factor (VEGF) - a trophic factor produced by astrocytes and that stimulates neurogenesis. Finally, UCMSI mice also had normal baseline corticosterone levels but a smaller increase in corticosterone following acute stress, that is, the Porsolt Swim Test. Our work gives clinically relevant insights into the role that microglial and astrocyte functioning, and hippocampal neurogenesis may play in the context of stress, social isolation and depression, offering a potentially new avenue for therapeutic target.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Diletta Onorato
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inez Eiben
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Martin Egeland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
40
|
Testen A, Kim R, Reissner KJ. High-Resolution Three-Dimensional Imaging of Individual Astrocytes Using Confocal Microscopy. ACTA ACUST UNITED AC 2020; 91:e92. [PMID: 32068976 DOI: 10.1002/cpns.92] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Astrocytes play numerous vital roles in the central nervous system. Accordingly, it is of merit to identify structural and functional properties of astrocytes in both health and disease. The majority of studies examining the morphology of astrocytes have employed immunoassays for markers such as glial fibrillary acidic protein, which are insufficient to encapsulate the considerable structural complexity of these cells. Herein, we describe a method utilizing a commercially available and validated, genetically encoded membrane-associated fluorescent marker of astrocytes, AAV5-GfaABC1D-Lck-GFP. This tool and approach allow for visualization of a single isolated astrocyte in its entirety, including fine peripheral processes. Astrocytes are imaged using confocal microscopy and reconstructed in three dimensions to obtain detailed morphometric data. We further provide an immunohistochemistry procedure to assess colocalization of isolated astrocytes with synaptic markers throughout the z-plane. This technique, which can be utilized via a standard laboratory confocal microscope and Imaris software, allows for detailed analysis of the morphology and synaptic colocalization of astrocytes in fixed tissue. © 2020 by John Wiley & Sons, Inc. Basic Protocol 1: Microinjection of AAV5-GfaABC1D-Lck-GFP into the nucleus accumbens of rats Basic Protocol 2: Tissue processing and immunohistochemistry for post-synaptic density-95 Basic Protocol 3: Single-cell image acquisition Basic Protocol 4: Three-dimensional reconstruction of single cells Basic Protocol 5: Three-dimensional colocalization analysis.
Collapse
Affiliation(s)
- Anze Testen
- Neuroscience Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ronald Kim
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathryn J Reissner
- Neuroscience Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
41
|
Abstract
Astrocytes, the star-shaped brain cells, are known chemosensitive cells in the respiratory system. A new study shows that trafficking of TRPA1 channels in and out of the cell membrane in brainstem astrocytes contributes to their role as central respiratory oxygen sensors.
Collapse
Affiliation(s)
- Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
MacDonald AJ, Ellacott KLJ. Astrocytes in the nucleus of the solitary tract: Contributions to neural circuits controlling physiology. Physiol Behav 2020; 223:112982. [PMID: 32535136 PMCID: PMC7378570 DOI: 10.1016/j.physbeh.2020.112982] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/04/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
The nucleus of the solitary tract (NTS) is the primary brainstem centre for the integration of physiological information from the periphery transmitted via the vagus nerve. In turn, the NTS feeds into downstream circuits regulating physiological parameters. Astrocytes are glial cells which have key roles in maintaining CNS tissue homeostasis and regulating neuronal communication. Recently an increasing number of studies have implicated astrocytes in the regulation of synaptic transmission and physiology. This review aims to highlight evidence for a role for astrocytes in the functions of the NTS. Astrocytes maintain and modulate NTS synaptic transmission contributing to the control of diverse physiological systems namely cardiovascular, respiratory, glucoregulatory, and gastrointestinal. In addition, it appears these cells may have a role in central control of feeding behaviour. As such these cells are a key component of signal processing and physiological control by the NTS.
Collapse
Affiliation(s)
- Alastair J MacDonald
- Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Level 4, RILD, Barrack Rd, Exeter EX2 5DW, UK
| | - Kate L J Ellacott
- Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Level 4, RILD, Barrack Rd, Exeter EX2 5DW, UK.
| |
Collapse
|
43
|
Mastitskaya S, Turovsky E, Marina N, Theparambil SM, Hadjihambi A, Kasparov S, Teschemacher AG, Ramage AG, Gourine AV, Hosford PS. Astrocytes Modulate Baroreflex Sensitivity at the Level of the Nucleus of the Solitary Tract. J Neurosci 2020; 40:3052-3062. [PMID: 32132265 PMCID: PMC7141885 DOI: 10.1523/jneurosci.1438-19.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 12/16/2019] [Accepted: 01/12/2020] [Indexed: 11/21/2022] Open
Abstract
Maintenance of cardiorespiratory homeostasis depends on autonomic reflexes controlled by neuronal circuits of the brainstem. The neurophysiology and neuroanatomy of these reflex pathways are well understood, however, the mechanisms and functional significance of autonomic circuit modulation by glial cells remain largely unknown. In the experiments conducted in male laboratory rats we show that astrocytes of the nucleus of the solitary tract (NTS), the brain area that receives and integrates sensory information from the heart and blood vessels, respond to incoming afferent inputs with [Ca2+]i elevations. Astroglial [Ca2+]i responses are triggered by transmitters released by vagal afferents, glutamate acting at AMPA receptors and 5-HT acting at 5-HT2A receptors. In conscious freely behaving animals blockade of Ca2+-dependent vesicular release mechanisms in NTS astrocytes by virally driven expression of a dominant-negative SNARE protein (dnSNARE) increased baroreflex sensitivity by 70% (p < 0.001). This effect of compromised astroglial function was specific to the NTS as expression of dnSNARE in astrocytes of the ventrolateral brainstem had no effect. ATP is considered the principle gliotransmitter and is released by vesicular mechanisms blocked by dnSNARE expression. Consistent with this hypothesis, in anesthetized rats, pharmacological activation of P2Y1 purinoceptors in the NTS decreased baroreflex gain by 40% (p = 0.031), whereas blockade of P2Y1 receptors increased baroreflex gain by 57% (p = 0.018). These results suggest that glutamate and 5-HT, released by NTS afferent terminals, trigger Ca2+-dependent astroglial release of ATP to modulate baroreflex sensitivity via P2Y1 receptors. These data add to the growing body of evidence supporting an active role of astrocytes in brain information processing.SIGNIFICANCE STATEMENT Cardiorespiratory reflexes maintain autonomic balance and ensure cardiovascular health. Impaired baroreflex may contribute to the development of cardiovascular disease and serves as a robust predictor of cardiovascular and all-cause mortality. The data obtained in this study suggest that astrocytes are integral components of the brainstem mechanisms that process afferent information and modulate baroreflex sensitivity via the release of ATP. Any condition associated with higher levels of "ambient" ATP in the NTS would be expected to decrease baroreflex gain by the mechanism described here. As ATP is the primary signaling molecule of glial cells (astrocytes, microglia), responding to metabolic stress and inflammatory stimuli, our study suggests a plausible mechanism of how the central component of the baroreflex is affected in pathological conditions.
Collapse
Affiliation(s)
- Svetlana Mastitskaya
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Egor Turovsky
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino 142290, Russian Federation
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Shefeeq M Theparambil
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Anna Hadjihambi
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Sergey Kasparov
- Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
- Baltic Federal University, Kaliningrad 236041, Russian Federation, and
| | - Anja G Teschemacher
- Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Andrew G Ramage
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom,
| | - Patrick S Hosford
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, United Kingdom,
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| |
Collapse
|
44
|
Patodia S, Paradiso B, Ellis M, Somani A, Sisodiya SM, Devinsky O, Thom M. Characterisation of medullary astrocytic populations in respiratory nuclei and alterations in sudden unexpected death in epilepsy. Epilepsy Res 2019; 157:106213. [PMID: 31610338 DOI: 10.1016/j.eplepsyres.2019.106213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 12/29/2022]
Abstract
Central failure of respiration during a seizure is one possible mechanism for sudden unexpected death in epilepsy (SUDEP). Neuroimaging studies indicate volume loss in the medulla in SUDEP and a post mortem study has shown reduction in neuromodulatory neuropeptidergic and monoaminergic neurones in medullary respiratory nuclear groups. Specialised glial cells identified in the medulla are considered essential for normal respiratory regulation including astrocytes with pacemaker properties in the pre-Botzinger complex and populations of subpial and perivascular astrocytes, sensitive to increased pCO2, that excite respiratory neurones. Our aim was to explore niches of medullary astrocytes in SUDEP cases compared to controls. In 48 brainstems from three groups, SUDEP (20), epilepsy controls (10) and non-epilepsy controls (18), sections through the medulla were labelled for GFAP, vimentin and functional markers, astrocytic gap junction protein connexin43 (Cx43) and adenosine A1 receptor (A1R). Regions including the ventro-lateral medulla (VLM; for the pre-Bötzinger complex), Median Raphe (MR) and lateral medullary subpial layer (MSPL) were quantified using image analysis for glial cell populations and compared between groups. Findings included morphologically and regionally distinct vimentin/Cx34-positive glial cells in the VLM and MR in close proximity to neurones. We noted a reduction of vimentin-positive glia in the VLM and MSPL and Cx43 glia in the MR in SUDEP cases compared to control groups (p < 0.05-0.005). In addition, we identified vimentin, Cx43 and A1R positive glial cells in the MSPL region which likely correspond to chemosensory glia identified experimentally. In conclusion, altered medullary glial cell populations could contribute to impaired respiratory regulatory capacity and vulnerability to SUDEP and warrant further investigation.
Collapse
Affiliation(s)
- Smriti Patodia
- Departments of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Beatrice Paradiso
- Departments of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom; Cardiovascular Pathology Unit, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padua, Italy
| | - Matthew Ellis
- Departments of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Alyma Somani
- Departments of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Sanjay M Sisodiya
- Departments of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Orrin Devinsky
- New York University School of Medicine, Comprehensive Epilepsy Center, New York, United States
| | - Maria Thom
- Departments of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom; Departments of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom.
| |
Collapse
|
45
|
Astrocyte networks modulate respiration – sniffing glue. Respir Physiol Neurobiol 2019; 265:3-8. [DOI: 10.1016/j.resp.2018.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/17/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
|
46
|
Czeisler CM, Silva TM, Fair SR, Liu J, Tupal S, Kaya B, Cowgill A, Mahajan S, Silva PE, Wang Y, Blissett AR, Göksel M, Borniger JC, Zhang N, Fernandes‐Junior SA, Catacutan F, Alves MJ, Nelson RJ, Sundaresean V, Rekling J, Takakura AC, Moreira TS, Otero JJ. The role of PHOX2B-derived astrocytes in chemosensory control of breathing and sleep homeostasis. J Physiol 2019; 597:2225-2251. [PMID: 30707772 PMCID: PMC6462490 DOI: 10.1113/jp277082] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/28/2019] [Indexed: 01/07/2023] Open
Abstract
KEY POINTS The embryonic PHOX2B-progenitor domain generates neuronal and glial cells which together are involved in chemosensory control of breathing and sleep homeostasis. Ablating PHOX2B-derived astrocytes significantly contributes to secondary hypoxic respiratory depression as well as abnormalities in sleep homeostasis. PHOX2B-derived astrocyte ablation results in axonal pathologies in the retrotrapezoid nucleus. ABSTRACT We identify in mice a population of ∼800 retrotrapezoid nucleus (RTN) astrocytes derived from PHOX2B-positive, OLIG3-negative progenitor cells, that interact with PHOX2B-expressing RTN chemosensory neurons. PHOX2B-derived astrocyte ablation during early life results in adult-onset O2 chemoreflex deficiency. These animals also display changes in sleep homeostasis, including fragmented sleep and disturbances in delta power after sleep deprivation, all without observable changes in anxiety or social behaviours. Ultrastructural evaluation of the RTN demonstrates that PHOX2B-derived astrocyte ablation results in features characteristic of degenerative neuro-axonal dystrophy, including abnormally dilated axon terminals and increased amounts of synapses containing autophagic vacuoles/phagosomes. We conclude that PHOX2B-derived astrocytes are necessary for maintaining a functional O2 chemosensory reflex in the adult, modulate sleep homeostasis, and are key regulators of synaptic integrity in the RTN region, which is necessary for the chemosensory control of breathing. These data also highlight how defects in embryonic development may manifest as neurodegenerative pathology in an adult.
Collapse
Affiliation(s)
| | - Talita M. Silva
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - Summer R. Fair
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jillian Liu
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Srinivasan Tupal
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Behiye Kaya
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Aaron Cowgill
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Salil Mahajan
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Phelipe E. Silva
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - Yangyang Wang
- Department of NeuroscienceThe Ohio State University College of MedicineColumbusOHUSA
- The Ohio State University Mathematical Biosciences InstituteColumbusOHUSA
| | - Angela R. Blissett
- Department of Mechanical and Aerospace EngineeringThe Ohio State University College of EngineeringColumbusOHUSA
| | - Mustafa Göksel
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jeremy C. Borniger
- Department of NeuroscienceThe Ohio State University College of MedicineColumbusOHUSA
| | - Ning Zhang
- Department of NeuroscienceWest Virginia UniversityWVUSA
| | - Silvio A. Fernandes‐Junior
- The Ohio State University Campus Microscopy and Imaging FacilityColumbusOHUSA
- Department of PharmacologyInstitute of Biomedical ScienceUniversity of São PauloSao PauloBrazil
| | - Fay Catacutan
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Michele J. Alves
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | | | - Vishnu Sundaresean
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jens Rekling
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
| | - Ana C. Takakura
- Department of PharmacologyInstitute of Biomedical ScienceUniversity of São PauloSao PauloBrazil
| | - Thiago S. Moreira
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - José J. Otero
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| |
Collapse
|
47
|
SheikhBahaei S, Morris B, Collina J, Anjum S, Znati S, Gamarra J, Zhang R, Gourine AV, Smith JC. Morphometric analysis of astrocytes in brainstem respiratory regions. J Comp Neurol 2018; 526:2032-2047. [PMID: 29888789 PMCID: PMC6158060 DOI: 10.1002/cne.24472] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/07/2018] [Accepted: 05/13/2018] [Indexed: 12/30/2022]
Abstract
Astrocytes, the most abundant and structurally complex glial cells of the central nervous system, are proposed to play an important role in modulating the activities of neuronal networks, including respiratory rhythm‐generating circuits of the preBötzinger complex (preBötC) located in the ventrolateral medulla of the brainstem. However, structural properties of astrocytes residing within different brainstem regions are unknown. In this study astrocytes in the preBötC, an intermediate reticular formation (IRF) region with respiratory‐related function, and a region of the nucleus tractus solitarius (NTS) in adult rats were reconstructed and their morphological features were compared. Detailed morphological analysis revealed that preBötC astrocytes are structurally more complex than those residing within the functionally distinct neighboring IRF region, or the NTS, located at the dorsal aspect of the medulla oblongata. Structural analyses of the brainstem microvasculature indicated no significant regional differences in vascular properties. We hypothesize that high morphological complexity of preBötC astrocytes reflects their functional role in providing structural/metabolic support and modulation of the key neuronal circuits essential for breathing, as well as constraints imposed by arrangements of associated neurons and/or other local structural features of the brainstem parenchyma.
Collapse
Affiliation(s)
- Shahriar SheikhBahaei
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland.,Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Brian Morris
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Jared Collina
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Sommer Anjum
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Sami Znati
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Julio Gamarra
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Ruli Zhang
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Jeffrey C Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS) National Institutes of Health (NIH), Bethesda, Maryland
| |
Collapse
|