1
|
Staehr C, Login H, Melnikova EV, Bakun M, Ziemlinska E, Kisiswa L, Ardestani SB, Nolte SS, Beck HC, Hansen LMB, Postnov D, Verkhratsky A, Malik AR, Nykjaer A, Matchkov VV. SorCS2 Is Important for Astrocytic Function in Neurovascular Signaling. Acta Physiol (Oxf) 2025; 241:e70052. [PMID: 40342271 PMCID: PMC12060088 DOI: 10.1111/apha.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION The receptor SorCS2 is involved in the trafficking of membrane receptors and transporters. It has been implicated in brain disorders and has previously been reported to be indispensable for ionotropic glutamatergic neurotransmission in the hippocampus. AIM We aimed to study the role of SorCS2 in the control of astrocyte-neuron communication, critical for neurovascular coupling. METHODS Brain slices from P8 and 2-month-old wild-type and SorCS2 knockout (Sorcs2-/-) mice were immunostained for SorCS2, GFAP, AQP4, IB4, and CD31. Neurovascular coupling was assessed in vivo using laser speckle contrast imaging and ex vivo in live brain slices loaded with calcium-sensitive dye. Bulk and cell surface fraction proteomics was analyzed on freshly isolated and cultured astrocytes, respectively, and validated with Western blot and qPCR. RESULTS SorCS2 was strongly expressed in astrocytes, primarily in their endfeet, of P8 mice; however, it was sparsely represented in 2-month-old mice. Sorcs2-/- mice demonstrated reduced neurovascular coupling associated with a reduced astrocytic calcium response to neuronal excitation. No differences in vascularization or endothelium-dependent relaxation ex vivo between the 2-month-old groups were observed. Proteomics suggested changes in glutamatergic signaling and suppressed calcium signaling in Sorcs2-/- brains from both P8 and 2-month-old mice. The increased abundance of glutamate metabotropic receptor 3 in Sorcs2-/- astrocytes was validated by PCR and Western blot. In cultured Sorcs2-/- astrocytes, AQP4 abundance was increased in the bulk lysate but reduced in the cell surface fraction, suggesting impaired trafficking. CONCLUSION The results suggest that SorCS2 expression is important for the development of neurovascular coupling, at least in part by modulating glutamatergic and calcium signaling in astrocytes.
Collapse
Affiliation(s)
- Christian Staehr
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Anaesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
| | - Hande Login
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | | | - Magdalena Bakun
- Mass Spectrometry LaboratoryInstitute of Biochemistry and Biophysics, Polish Academy of SciencesWarsawPoland
| | - Ewelina Ziemlinska
- Cellular Neurobiology Research Group, Faculty of Biology, Institute of Developmental Biology and Biomedical SciencesUniversity of WarsawWarsawPoland
| | | | | | | | | | | | - Dmitry Postnov
- Department of Clinical Medicine, Center of Functionally Integrative NeuroscienceAarhus University Hospital SkejbyAarhusDenmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque CountryLeioaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
- International Joint Research Centre on Purinergic Signalling of Sichuan Province Chengdu University of Traditional Chinese MedicineChengduChina
| | - Anna R. Malik
- Cellular Neurobiology Research Group, Faculty of Biology, Institute of Developmental Biology and Biomedical SciencesUniversity of WarsawWarsawPoland
| | - Anders Nykjaer
- Department of BiomedicineAarhus UniversityAarhusDenmark
- PROMEMO and DANDRITEAarhus UniversityAarhusDenmark
- Department of NeurosurgeryAarhus University Hospital SkejbyAarhusDenmark
| | | |
Collapse
|
2
|
Vieira WF, Real CC, Martins DO, Chacur M. The Role of Exercise on Glial Cell Activity in Neuropathic Pain Management. Cells 2025; 14:487. [PMID: 40214441 PMCID: PMC11988158 DOI: 10.3390/cells14070487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic pain is a widespread global health problem with profound socioeconomic implications, affecting millions of people of all ages. Glial cells (GCs) in pain pathways play essential roles in the processing of pain signals. Dysregulation of GC activity contributes to chronic pain states, making them targets for therapeutic interventions. Non-pharmacological approaches, such as exercise, are strongly recommended for effective pain management. This review examines the link between exercise, regular physical activity (PA), and glial cell-mediated pain processing, highlighting its potential as a strategy for managing chronic pain. Exercise not only improves overall health and quality of life but also influences the function of GCs. Recent research highlights the ability of exercise to mitigate neuroinflammatory responses and modulate the activity of GCs by reducing the activation of microglia and astrocytes, as well as modulating the expression biomarkers, thereby attenuating pain hypersensitivity. Here, we summarize new insights into the role of exercise as a non-pharmacological intervention for the relief of chronic pain.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), 2415 Prof. Lineu Prestes Avenue, São Paulo 05508-000, SP, Brazil;
| | - Caroline C. Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | | | - Marucia Chacur
- Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), 2415 Prof. Lineu Prestes Avenue, São Paulo 05508-000, SP, Brazil;
| |
Collapse
|
3
|
Suarez A, Fernandez L, Riera J. Characterizing astrocyte-mediated neurovascular coupling by combining optogenetics and biophysical modeling. J Cereb Blood Flow Metab 2025:271678X241311010. [PMID: 39791314 PMCID: PMC11719438 DOI: 10.1177/0271678x241311010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Vasoactive signaling from astrocytes is an important contributor to the neurovascular coupling (NVC), which aims at providing energy to neurons during brain activation by increasing blood perfusion in the surrounding vasculature. Pharmacological manipulations have been previously combined with experimental techniques (e.g., transgenic mice, uncaging, and multiphoton microscopy) and stimulation paradigms to isolate in vivo individual pathways of the astrocyte-mediated NVC. Unfortunately, these pathways are highly nonlinear and non-additive. To separate these pathways in a unified framework, we combine a comprehensive biophysical model of vasoactive signaling from astrocytes with a unique optogenetic stimulation method that selectively induces astrocytic Ca2+ signaling in a large population of astrocytes. We also use a sensitivity analysis and an optimization technique to estimate key model parameters. Optogenetically-induced Ca2+ signals in astrocytes cause a cerebral blood flow (CBF) response with two major components. Component-1 was rapid and smaller (ΔCBF∼13%, 18 seconds), while component-2 was slowest and highest (ΔCBF ∼18%, 45 seconds). The proposed biophysical model was adequate in reproducing component-2, which was validated with a pharmacological manipulation. Model's predictions were not in contradiction with previous studies. Finally, we discussed scenarios accounting for the existence of component-1, which once validated might be included in our model.
Collapse
Affiliation(s)
- Alejandro Suarez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Lazaro Fernandez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Jorge Riera
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| |
Collapse
|
4
|
Yang L, Zhao W, Kan Y, Ren C, Ji X. From Mechanisms to Medicine: Neurovascular Coupling in the Diagnosis and Treatment of Cerebrovascular Disorders: A Narrative Review. Cells 2024; 14:16. [PMID: 39791717 PMCID: PMC11719775 DOI: 10.3390/cells14010016] [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: 10/30/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Neurovascular coupling (NVC) refers to the process of local changes in cerebral blood flow (CBF) after neuronal activity, which ensures the timely and adequate supply of oxygen, glucose, and substrates to the active regions of the brain. Recent clinical imaging and experimental technology advancements have deepened our understanding of the cellular mechanisms underlying NVC. Pathological conditions such as stroke, subarachnoid hemorrhage, cerebral small vascular disease, and vascular cognitive impairment can disrupt NVC even before clinical symptoms appear. However, the complexity of the underlying mechanism remains unclear. This review discusses basic and clinical experimental evidence on how neural activity sensitively communicates with the vasculature to cause spatial changes in blood flow in cerebrovascular diseases. A deeper understanding of how neurovascular unit-related cells participate in NVC regulation is necessary to better understand blood flow and nerve activity recovery in cerebrovascular diseases.
Collapse
Affiliation(s)
- Lu Yang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
| | - Yuan Kan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
| | - Changhong Ren
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
5
|
Song J. BDNF Signaling in Vascular Dementia and Its Effects on Cerebrovascular Dysfunction, Synaptic Plasticity, and Cholinergic System Abnormality. J Lipid Atheroscler 2024; 13:122-138. [PMID: 38826183 PMCID: PMC11140249 DOI: 10.12997/jla.2024.13.2.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 06/04/2024] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia and is characterized by memory impairment, blood-brain barrier disruption, neuronal cell loss, glia activation, impaired synaptic plasticity, and cholinergic system abnormalities. To effectively prevent and treat VaD a good understanding of the mechanisms underlying its neuropathology is needed. Brain-derived neurotrophic factor (BDNF) is an important neurotrophic factor with multiple functions in the systemic circulation and the central nervous system and is known to regulate neuronal cell survival, synaptic formation, glia activation, and cognitive decline. Recent studies indicate that when compared with normal subjects, patients with VaD have low serum BDNF levels and that BDNF deficiency in the serum and cerebrospinal fluid is an important indicator of VaD. Here, we review current knowledge on the role of BDNF signaling in the pathology of VaD, such as cerebrovascular dysfunction, synaptic dysfunction, and cholinergic system impairment.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
6
|
Feng L, Gao L. The role of neurovascular coupling dysfunction in cognitive decline of diabetes patients. Front Neurosci 2024; 18:1375908. [PMID: 38576869 PMCID: PMC10991808 DOI: 10.3389/fnins.2024.1375908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Neurovascular coupling (NVC) is an important mechanism to ensure adequate blood supply to active neurons in the brain. NVC damage can lead to chronic impairment of neuronal function. Diabetes is characterized by high blood sugar and is considered an important risk factor for cognitive impairment. In this review, we provide fMRI evidence of NVC damage in diabetic patients with cognitive decline. Combined with the exploration of the major mechanisms and signaling pathways of NVC, we discuss the effects of chronic hyperglycemia on the cellular structure of NVC signaling, including key receptors, ion channels, and intercellular connections. Studying these diabetes-related changes in cell structure will help us understand the underlying causes behind diabetes-induced NVC damage and early cognitive decline, ultimately helping to identify the most effective drug targets for treatment.
Collapse
Affiliation(s)
| | - Ling Gao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Renden RB, Institoris A, Sharma K, Tran CHT. Modulatory effects of noradrenergic and serotonergic signaling pathway on neurovascular coupling. Commun Biol 2024; 7:287. [PMID: 38459113 PMCID: PMC10923894 DOI: 10.1038/s42003-024-05996-y] [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: 06/24/2023] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
Dynamic changes in astrocyte Ca2+ are recognized as contributors to functional hyperemia, a critical response to increased neuronal activity mediated by a process known as neurovascular coupling (NVC). Although the critical role of glutamatergic signaling in this process has been extensively investigated, the impact of behavioral state, and the release of behavior-associated neurotransmitters, such as norepinephrine and serotonin, on astrocyte Ca2+ dynamics and functional hyperemia have received less attention. We used two-photon imaging of the barrel cortex in awake mice to examine the role of noradrenergic and serotonergic projections in NVC. We found that both neurotransmitters facilitated sensory stimulation-induced increases in astrocyte Ca2+. Interestingly, while ablation of serotonergic neurons reduced sensory stimulation-induced functional hyperemia, ablation of noradrenergic neurons caused both attenuation and potentiation of functional hyperemia. Our study demonstrates that norepinephrine and serotonin are involved in modulating sensory stimulation-induced astrocyte Ca2+ elevations and identifies their differential effects in regulating functional hyperemia.
Collapse
Affiliation(s)
- Robert B Renden
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV, USA
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kushal Sharma
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV, USA
| | - Cam Ha T Tran
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV, USA.
| |
Collapse
|
8
|
Ozawa K, Nagao M, Konno A, Iwai Y, Vittani M, Kusk P, Mishima T, Hirai H, Nedergaard M, Hirase H. Astrocytic GPCR-Induced Ca 2+ Signaling Is Not Causally Related to Local Cerebral Blood Flow Changes. Int J Mol Sci 2023; 24:13590. [PMID: 37686396 PMCID: PMC10487464 DOI: 10.3390/ijms241713590] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/17/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Activation of Gq-type G protein-coupled receptors (GPCRs) gives rise to large cytosolic Ca2+ elevations in astrocytes. Previous in vitro and in vivo studies have indicated that astrocytic Ca2+ elevations are closely associated with diameter changes in the nearby blood vessels, which astrocytes enwrap with their endfeet. However, the causal relationship between astrocytic Ca2+ elevations and blood vessel diameter changes has been questioned, as mice with diminished astrocytic Ca2+ signaling show normal sensory hyperemia. We addressed this controversy by imaging cortical vasculature while optogenetically elevating astrocyte Ca2+ in a novel transgenic mouse line, expressing Opto-Gq-type GPCR Optoα1AR (Astro-Optoα1AR) in astrocytes. Blue light illumination on the surface of the somatosensory cortex induced Ca2+ elevations in cortical astrocytes and their endfeet in mice under anesthesia. Blood vessel diameter did not change significantly with Optoα1AR-induced Ca2+ elevations in astrocytes, while it was increased by forelimb stimulation. Next, we labeled blood plasma with red fluorescence using AAV8-P3-Alb-mScarlet in Astro-Optoα1AR mice. We were able to identify arterioles that display diameter changes in superficial areas of the somatosensory cortex through the thinned skull. Photo-stimulation of astrocytes in the cortical area did not result in noticeable changes in the arteriole diameters compared with their background strain C57BL/6. Together, compelling evidence for astrocytic Gq pathway-induced vasodiameter changes was not observed. Our results support the notion that short-term (<10 s) hyperemia is not mediated by GPCR-induced astrocytic Ca2+ signaling.
Collapse
Affiliation(s)
- Katsuya Ozawa
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako 351-0106, Saitama, Japan; (K.O.)
| | - Masaki Nagao
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan
- Viral Vector Core, Gunma University, Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Youichi Iwai
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako 351-0106, Saitama, Japan; (K.O.)
| | - Marta Vittani
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Peter Kusk
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Tsuneko Mishima
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan
- Viral Vector Core, Gunma University, Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
9
|
Kozachkov L, Kastanenka KV, Krotov D. Building transformers from neurons and astrocytes. Proc Natl Acad Sci U S A 2023; 120:e2219150120. [PMID: 37579149 PMCID: PMC10450673 DOI: 10.1073/pnas.2219150120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 06/22/2023] [Indexed: 08/16/2023] Open
Abstract
Glial cells account for between 50% and 90% of all human brain cells, and serve a variety of important developmental, structural, and metabolic functions. Recent experimental efforts suggest that astrocytes, a type of glial cell, are also directly involved in core cognitive processes such as learning and memory. While it is well established that astrocytes and neurons are connected to one another in feedback loops across many timescales and spatial scales, there is a gap in understanding the computational role of neuron-astrocyte interactions. To help bridge this gap, we draw on recent advances in AI and astrocyte imaging technology. In particular, we show that neuron-astrocyte networks can naturally perform the core computation of a Transformer, a particularly successful type of AI architecture. In doing so, we provide a concrete, normative, and experimentally testable account of neuron-astrocyte communication. Because Transformers are so successful across a wide variety of task domains, such as language, vision, and audition, our analysis may help explain the ubiquity, flexibility, and power of the brain's neuron-astrocyte networks.
Collapse
Affiliation(s)
- Leo Kozachkov
- Massachusetts Institute of Technology-International Business Machines, Watson Artificial Intelligence Laboratory, IBM Research, Cambridge, MA02142
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA02115
| | - Dmitry Krotov
- Massachusetts Institute of Technology-International Business Machines, Watson Artificial Intelligence Laboratory, IBM Research, Cambridge, MA02142
| |
Collapse
|
10
|
Baranes K, Hastings N, Rahman S, Poulin N, Tavares JM, Kuan W, Syed N, Kunz M, Blighe K, Belgard TG, Kotter MRN. Transcription factor combinations that define human astrocyte identity encode significant variation of maturity and function. Glia 2023; 71:1870-1889. [PMID: 37029764 PMCID: PMC10952910 DOI: 10.1002/glia.24372] [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: 08/16/2021] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 04/09/2023]
Abstract
Increasing evidence indicates that cellular identity can be reduced to the distinct gene regulatory networks controlled by transcription factors (TFs). However, redundancy exists in these states as different combinations of TFs can induce broadly similar cell types. We previously demonstrated that by overcoming gene silencing, it is possible to deterministically reprogram human pluripotent stem cells directly into cell types of various lineages. In the present study we leverage the consistency and precision of our approach to explore four different TF combinations encoding astrocyte identity, based on previously published reports. Analysis of the resulting induced astrocytes (iAs) demonstrated that all four cassettes generate cells with the typical morphology of in vitro astrocytes, which expressed astrocyte-specific markers. The transcriptional profiles of all four iAs clustered tightly together and displayed similarities with mature human astrocytes, although maturity levels differed between cells. Importantly, we found that the TF cassettes induced iAs with distinct differences with regards to their cytokine response and calcium signaling. In vivo transplantation of selected iAs into immunocompromised rat brains demonstrated long term stability and integration. In conclusion, all four TF combinations were able to induce stable astrocyte-like cells that were morphologically similar but showed subtle differences with respect to their transcriptome. These subtle differences translated into distinct differences with regards to cell function, that could be related to maturation state and/or regional identity of the resulting cells. This insight opens an opportunity to precision-engineer cells to meet functional requirements, for example, in the context of therapeutic cell transplantation.
Collapse
Affiliation(s)
- Koby Baranes
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Nataly Hastings
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Saifur Rahman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Noah Poulin
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Joana M. Tavares
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Wei‐Li Kuan
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - Najeeb Syed
- The Bioinformatics CROSanfordFlorida32771USA
| | - Meik Kunz
- The Bioinformatics CROSanfordFlorida32771USA
| | | | | | - Mark R. N. Kotter
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| |
Collapse
|
11
|
Paulson OB, Schousboe A, Hultborn H. The history of Danish neuroscience. Eur J Neurosci 2023; 58:2893-2960. [PMID: 37477973 DOI: 10.1111/ejn.16062] [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: 11/22/2022] [Revised: 05/04/2023] [Accepted: 05/29/2023] [Indexed: 07/22/2023]
Abstract
The history of Danish neuroscience starts with an account of impressive contributions made at the 17th century. Thomas Bartholin was the first Danish neuroscientist, and his disciple Nicolaus Steno became internationally one of the most prominent neuroscientists in this period. From the start, Danish neuroscience was linked to clinical disciplines. This continued in the 19th and first half of the 20th centuries with new initiatives linking basic neuroscience to clinical neurology and psychiatry in the same scientific environment. Subsequently, from the middle of the 20th century, basic neuroscience was developing rapidly within the preclinical university sector. Clinical neuroscience continued and was even reinforced during this period with important translational research and a close co-operation between basic and clinical neuroscience. To distinguish 'history' from 'present time' is not easy, as many historical events continue in present time. Therefore, we decided to consider 'History' as new major scientific developments in Denmark, which were launched before the end of the 20th century. With this aim, scientists mentioned will have been born, with a few exceptions, no later than the early 1960s. However, we often refer to more recent publications in documenting the developments of initiatives launched before the end of the last century. In addition, several scientists have moved to Denmark after the beginning of the present century, and they certainly are contributing to the present status of Danish neuroscience-but, again, this is not the History of Danish neuroscience.
Collapse
Affiliation(s)
- Olaf B Paulson
- Neurobiology Research Unit, Department of Neurology, Rigshospitalet, 9 Blegdamsvej, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Hultborn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Fedotova A, Brazhe A, Doronin M, Toptunov D, Pryazhnikov E, Khiroug L, Verkhratsky A, Semyanov A. Dissociation Between Neuronal and Astrocytic Calcium Activity in Response to Locomotion in Mice. FUNCTION 2023; 4:zqad019. [PMID: 37342415 PMCID: PMC10278990 DOI: 10.1093/function/zqad019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 06/22/2023] Open
Abstract
Locomotion triggers a coordinated response of both neurons and astrocytes in the brain. Here we performed calcium (Ca2+) imaging of these two cell types in the somatosensory cortex in head-fixed mice moving on the airlifted platform. Ca2+ activity in astrocytes significantly increased during locomotion from a low quiescence level. Ca2+ signals first appeared in the distal processes and then propagated to astrocytic somata, where it became significantly larger and exhibited oscillatory behaviour. Thus, astrocytic soma operates as both integrator and amplifier of Ca2+ signal. In neurons, Ca2+ activity was pronounced in quiescent periods and further increased during locomotion. Neuronal Ca2+ concentration ([Ca2+]i) rose almost immediately following the onset of locomotion, whereas astrocytic Ca2+ signals lagged by several seconds. Such a long lag suggests that astrocytic [Ca2+]i elevations are unlikely to be triggered by the activity of synapses among local neurons. Ca2+ responses to pairs of consecutive episodes of locomotion did not significantly differ in neurons, while were significantly diminished in response to the second locomotion in astrocytes. Such astrocytic refractoriness may arise from distinct mechanisms underlying Ca2+ signal generation. In neurons, the bulk of Ca2+ enters through the Ca2+ channels in the plasma membrane allowing for steady-level Ca2+ elevations in repetitive runs. Astrocytic Ca2+ responses originate from the intracellular stores, the depletion of which affects subsequent Ca2+ signals. Functionally, neuronal Ca2+ response reflects sensory input processed by neurons. Astrocytic Ca2+ dynamics is likely to provide metabolic and homeostatic support within the brain active milieu.
Collapse
Affiliation(s)
- Anna Fedotova
- Faculty of Biology, Moscow State University, Moscow 119991, Russia
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
| | - Alexey Brazhe
- Faculty of Biology, Moscow State University, Moscow 119991, Russia
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
| | - Maxim Doronin
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
- College of Medicine, Jiaxing University, Jiaxing, Zhejiang Province, 314001, China
| | | | | | | | | | | |
Collapse
|
13
|
Tesler F, Linne ML, Destexhe A. Modeling the relationship between neuronal activity and the BOLD signal: contributions from astrocyte calcium dynamics. Sci Rep 2023; 13:6451. [PMID: 37081004 PMCID: PMC10119111 DOI: 10.1038/s41598-023-32618-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/30/2023] [Indexed: 04/22/2023] Open
Abstract
Functional magnetic resonance imaging relies on the coupling between neuronal and vascular activity, but the mechanisms behind this coupling are still under discussion. Recent experimental evidence suggests that calcium signaling may play a significant role in neurovascular coupling. However, it is still controversial where this calcium signal is located (in neurons or elsewhere), how it operates and how relevant is its role. In this paper we introduce a biologically plausible model of the neurovascular coupling and we show that calcium signaling in astrocytes can explain main aspects of the dynamics of the coupling. We find that calcium signaling can explain so-far unrelated features such as the linear and non-linear regimes, the negative vascular response (undershoot) and the emergence of a (calcium-driven) Hemodynamic Response Function. These features are reproduced here for the first time by a single model of the detailed neuronal-astrocyte-vascular pathway. Furthermore, we analyze how information is coded and transmitted from the neuronal to the vascular system and we predict that frequency modulation of astrocytic calcium dynamics plays a key role in this process. Finally, our work provides a framework to link neuronal activity to the BOLD signal, and vice-versa, where neuronal activity can be inferred from the BOLD signal. This opens new ways to link known alterations of astrocytic calcium signaling in neurodegenerative diseases (e.g. Alzheimer's and Parkinson's diseases) with detectable changes in the neurovascular coupling.
Collapse
Affiliation(s)
- Federico Tesler
- CNRS, Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, 91400, Saclay, France.
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, 33720, Tampere, Finland
| | - Alain Destexhe
- CNRS, Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, 91400, Saclay, France
| |
Collapse
|
14
|
Hazra R, Novelli EM, Hu X. Astrocytic mitochondrial frataxin-A promising target for ischemic brain injury. CNS Neurosci Ther 2023; 29:783-788. [PMID: 36550598 PMCID: PMC9928550 DOI: 10.1111/cns.14068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
In the ischemic brain, hypoxia leads to mitochondrial dysfunction, insufficient energy production, and astrocyte activation. Yet, most studies investigating mitochondrial dysfunction in cerebral ischemia have focused exclusively on neurons. This review will highlight the importance of the morphological, molecular, and functional heterogeneity of astrocytes in their role in brain injuries and explore how activated astrocytes exhibit calcium imbalance, reactive oxygen species overproduction, and apoptosis. In addition, special focus will be given to the role of the mitochondrial protein frataxin in activated astrocytes during ischemia and its putative role in the pharmacological management of cerebral ischemia.
Collapse
Affiliation(s)
- Rimi Hazra
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Hu
- Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Toth L, Czigler A, Hegedus E, Komaromy H, Amrein K, Czeiter E, Yabluchanskiy A, Koller A, Orsi G, Perlaki G, Schwarcz A, Buki A, Ungvari Z, Toth PJ. Age-related decline in circulating IGF-1 associates with impaired neurovascular coupling responses in older adults. GeroScience 2022; 44:2771-2783. [PMID: 35869380 PMCID: PMC9768079 DOI: 10.1007/s11357-022-00623-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/09/2022] [Indexed: 01/07/2023] Open
Abstract
Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) to the increased oxygen and energy requirements of active brain regions via neurovascular coupling (NVC) contributes to the genesis of age-related cognitive impairment. Aging is associated with marked deficiency in the vasoprotective hormone insulin-like growth factor-1 (IGF-1). Preclinical studies on animal models of aging suggest that circulating IGF-1 deficiency is causally linked to impairment of NVC responses. The present study was designed to test the hypotheses that decreases in circulating IGF-1 levels in older adults also predict the magnitude of age-related decline of NVC responses. In a single-center cross-sectional study, we enrolled healthy young (n = 31, 11 female, 20 male, mean age: 28.4 + / - 4.2 years) and aged volunteers (n = 32, 18 female, 14 male, mean age: 67.9 + / - 4.1 years). Serum IGF-1 level, basal CBF (phase contrast magnetic resonance imaging (MRI)), and NVC responses during the trail making task (with transcranial Doppler sonography) were assessed. We found that circulating IGF-1 levels were significantly decreased with age and associated with decreased basal CBF. Age-related decline in IGF-1 levels predicted the magnitude of age-related decline in NVC responses. In conclusion, our study provides additional evidence in support of the concept that age-related circulating IGF-1 deficiency contributes to neurovascular aging, impairing CBF and functional hyperemia in older adults.
Collapse
Affiliation(s)
- Luca Toth
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andras Czigler
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Emoke Hegedus
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Hedvig Komaromy
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Akos Koller
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Gergely Orsi
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Lóránd Research Network (ELKH), Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Pecs, Hungary
| | - Gabor Perlaki
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Lóránd Research Network (ELKH), Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Pecs, Hungary
| | - Attila Schwarcz
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Andras Buki
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Peter J Toth
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary.
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
16
|
Ladthavorlaphatt K, Surti FBS, Beishon LC, Panerai RB, Robinson TG. Challenging neurovascular coupling through complex and variable duration cognitive paradigms: A subcomponent analysis. Med Eng Phys 2022; 110:103921. [PMID: 36564144 DOI: 10.1016/j.medengphy.2022.103921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/04/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
A similar pattern of cerebral blood velocity (CBv) response has been observed for neurovascular coupling (NVC) assessment with cognitive tasks of varying complexity and duration. This lack of specificity could result from parallel changes in arterial blood pressure (BP) and PaCO2, which could confound the estimates of NVC integrity. Healthy participants (n = 16) underwent recordings at rest (5 min sitting) and during randomized paradigms of different complexity (naming words (NW) beginning with P-, R-, V- words and serial subtractions (SS) of 100-2, 100-7, 1000-17, with durations of 5, 30 and 60 s). Bilateral CBv (middle cerebral arteries, transcranial Doppler), end-tidal CO2 (EtCO2, capnography), blood pressure (BP, Finapres) and heart rate (HR, ECG) were recorded continuously. The bilateral CBv response to all paradigms was classified under objective criteria to select only responders, then the repeated data were averaged between visits. Bilateral CBv change to tasks was decomposed into the relative contributions (subcomponents) of arterial BP (VBP; neurogenic), critical closing pressure (VCrCP; metabolic) and resistance area product (VRAP; myogenic). A temporal effect was demonstrated in bilateral VBP and VRAP during all tasks (p<0.002), increased VBP early (between 0 and 10 s) and followed by decreases of VRAP late (25-35 s) in the response. VCrCP varied by complexity and duration (p<0.046). The main contributions to CBv responses to cognitive tasks of different complexity and duration were VBP and VRAP, whilst a smaller contribution from VCrCP would suggest sensitivity to metabolic demands. Further studies are needed to assess the influence of different paradigms, ageing and cerebrovascular conditions.
Collapse
Affiliation(s)
- Kannaphob Ladthavorlaphatt
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; Medical Diagnostics Unit, Thammasat University Hospital, Thammasat University, Pathumthani, Thailand.
| | - Farhaana B S Surti
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom
| | - Lucy C Beishon
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Ronney B Panerai
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Thompson G Robinson
- Department of Cardiovascular Sciences, College of Life Sciences, Leicester Royal Infirmary, University of Leicester, Level 4, Robert Kilpatrick Clinical Sciences Building, Leicester LE2 7LX, United Kingdom; NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| |
Collapse
|
17
|
Denizot A, Arizono M, Nägerl UV, Berry H, De Schutter E. Control of Ca 2+ signals by astrocyte nanoscale morphology at tripartite synapses. Glia 2022; 70:2378-2391. [PMID: 36097958 PMCID: PMC9825906 DOI: 10.1002/glia.24258] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 01/11/2023]
Abstract
Much of the Ca2+ activity in astrocytes is spatially restricted to microdomains and occurs in fine processes that form a complex anatomical meshwork, the so-called spongiform domain. A growing body of literature indicates that those astrocytic Ca2+ signals can influence the activity of neuronal synapses and thus tune the flow of information through neuronal circuits. Because of technical difficulties in accessing the small spatial scale involved, the role of astrocyte morphology on Ca2+ microdomain activity remains poorly understood. Here, we use computational tools and idealized 3D geometries of fine processes based on recent super-resolution microscopy data to investigate the mechanistic link between astrocytic nanoscale morphology and local Ca2+ activity. Simulations demonstrate that the nano-morphology of astrocytic processes powerfully shapes the spatio-temporal properties of Ca2+ signals and promotes local Ca2+ activity. The model predicts that this effect is attenuated upon astrocytic swelling, hallmark of brain diseases, which we confirm experimentally in hypo-osmotic conditions. Upon repeated neurotransmitter release events, the model predicts that swelling hinders astrocytic signal propagation. Overall, this study highlights the influence of the complex morphology of astrocytes at the nanoscale and its remodeling in pathological conditions on neuron-astrocyte communication at so-called tripartite synapses, where astrocytic processes come into close contact with pre- and postsynaptic structures.
Collapse
Affiliation(s)
- Audrey Denizot
- Computational Neuroscience UnitOkinawa Institute of Science and TechnologyOnna‐SonJapan
| | - Misa Arizono
- Interdisciplinary Institute for NeuroscienceUniversité de BordeauxBordeauxFrance
- Interdisciplinary Institute for NeuroscienceCNRS UMR 5297BordeauxFrance
- Department of PharmacologyKyoto University Graduate School of MedicineKyotoJapan
| | - U. Valentin Nägerl
- Interdisciplinary Institute for NeuroscienceUniversité de BordeauxBordeauxFrance
- Interdisciplinary Institute for NeuroscienceCNRS UMR 5297BordeauxFrance
| | - Hugues Berry
- LIRIS, UMR5205 CNRSUniv LyonVilleurbanneFrance
- INRIAVilleurbanneFrance
| | - Erik De Schutter
- Computational Neuroscience UnitOkinawa Institute of Science and TechnologyOnna‐SonJapan
| |
Collapse
|
18
|
Ding F, Liang S, Li R, Yang Z, He Y, Yang S, Duan Q, Zhang J, Lyu J, Zhou Z, Huang M, Wang H, Li J, Yang C, Wang Y, Gong M, Chen S, Jia H, Chen X, Liao X, Fu L, Zhang K. Astrocytes exhibit diverse Ca2+ changes at subcellular domains during brain aging. Front Aging Neurosci 2022; 14:1029533. [PMID: 36389078 PMCID: PMC9650392 DOI: 10.3389/fnagi.2022.1029533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Astrocytic Ca2+ transients are essential for astrocyte integration into neural circuits. These Ca2+ transients are primarily sequestered in subcellular domains, including primary branches, branchlets and leaflets, and endfeet. In previous studies, it suggests that aging causes functional defects in astrocytes. Until now, it was unclear whether and how aging affects astrocytic Ca2+ transients at subcellular domains. In this study, we combined a genetically encoded Ca2+ sensor (GCaMP6f) and in vivo two-photon Ca2+ imaging to determine changes in Ca2+ transients within astrocytic subcellular domains during brain aging. We showed that aging increased Ca2+ transients in astrocytic primary branches, higher-order branchlets, and terminal leaflets. However, Ca2+ transients decreased within astrocytic endfeet during brain aging, which could be caused by the decreased expressions of Aquaporin-4 (AQP4). In addition, aging-induced changes of Ca2+ transient types were heterogeneous within astrocytic subcellular domains. These results demonstrate that the astrocytic Ca2+ transients within subcellular domains are affected by aging differently. This finding contributes to a better understanding of the physiological role of astrocytes in aging-induced neural circuit degeneration.
Collapse
Affiliation(s)
- Fusheng Ding
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shanshan Liang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Ruijie Li
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Yong He
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shaofan Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Qingtian Duan
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jianxiong Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jing Lyu
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Zhenqiao Zhou
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Mingzhu Huang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Haoyu Wang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jin Li
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Yuxia Wang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shangbin Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Jia
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, China
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Guangyang Bay Laboratory, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
- *Correspondence: Xiang Liao,
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
- Ling Fu,
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Kuan Zhang,
| |
Collapse
|
19
|
Looking to the stars for answers: Strategies for determining how astrocytes influence neuronal activity. Comput Struct Biotechnol J 2022; 20:4146-4156. [PMID: 36016711 PMCID: PMC9379862 DOI: 10.1016/j.csbj.2022.07.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022] Open
Abstract
Astrocytes are critical components of neural circuits positioned in close proximity to the synapse, allowing them to rapidly sense and respond to neuronal activity. One repeatedly observed biomarker of astroglial activation is an increase in intracellular Ca2+ levels. These astroglial Ca2+ signals are often observed spreading throughout various cellular compartments from perisynaptic astroglial processes, to major astrocytic branches and on to the soma or cell body. Here we review recent evidence demonstrating that astrocytic Ca2+ events are remarkably heterogeneous in both form and function, propagate through the astroglial syncytia, and are directly linked to the ability of astroglia to influence local neuronal activity. As many of the cellular functions of astroglia can be linked to intracellular Ca2+ signaling, and the diversity and heterogeneity of these events becomes more apparent, there is an increasing need for novel experimental strategies designed to better understand the how these signals evolve in parallel with neuronal activity. Here we review the recent advances that enable the characterization of both subcellular and population-wide astrocytic Ca2+ dynamics. Additionally, we also outline the experimental design required for simultaneous in vivo Ca2+ imaging in the context of neuronal or astroglial manipulation, highlighting new experimental strategies made possible by recent advances in viral vector, imaging, and quantification technologies. Through combined usage of these reagents and methodologies, we provide a conceptual framework to study how astrocytes functionally integrate into neural circuits and to what extent they influence and direct the synaptic activity underlying behavioral responses.
Collapse
|
20
|
Subclinical cognitive deficits are associated with reduced cerebrovascular response to visual stimulation in mid-sixties men. GeroScience 2022; 44:1905-1923. [PMID: 35648331 DOI: 10.1007/s11357-022-00596-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/22/2022] [Indexed: 11/04/2022] Open
Abstract
Reduced cerebrovascular response to neuronal activation is observed in patients with neurodegenerative disease. In the present study, we examined the correlation between reduced cerebrovascular response to visual activation (ΔCBFVis.Act) and subclinical cognitive deficits in a human population of mid-sixties individuals without neurodegenerative disease. Such a correlation would suggest that impaired cerebrovascular function occurs before overt neurodegenerative disease. A total of 187 subjects (age 64-67 years) of the Metropolit Danish Male Birth Cohort participated in the study. ΔCBFVis.Act was measured using arterial spin labelling (ASL) MRI. ΔCBFVis.Act correlated positively with cognitive performance in: Global cognition (p = 0.046), paired associative memory (p = 0.025), spatial recognition (p = 0.026), planning (p = 0.016), simple processing speed (p < 0.01), and with highly significant correlations with current intelligence (p < 10-5), and more complex processing speed (p < 10-3), the latter two explaining approximately 11-13% of the variance. Reduced ΔCBFVis.Act was independent of brain atrophy. Our findings suggest that inhibited cerebrovascular response to neuronal activation is an early deficit in the ageing brain and associated with subclinical cognitive deficits. Cerebrovascular dysfunction could be an early sign of a trajectory pointing towards the development of neurodegenerative disease. Future efforts should elucidate if maintenance of a healthy cerebrovascular function can protect against the development of dementia.
Collapse
|
21
|
The effects of locomotion on sensory-evoked haemodynamic responses in the cortex of awake mice. Sci Rep 2022; 12:6236. [PMID: 35422473 PMCID: PMC9010417 DOI: 10.1038/s41598-022-10195-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/04/2022] [Indexed: 12/22/2022] Open
Abstract
Investigating neurovascular coupling in awake rodents is becoming ever more popular due, in part, to our increasing knowledge of the profound impacts that anaesthesia can have upon brain physiology. Although awake imaging brings with it many advantages, we still do not fully understand how voluntary locomotion during imaging affects sensory-evoked haemodynamic responses. In this study we investigated how evoked haemodynamic responses can be affected by the amount and timing of locomotion. Using an awake imaging set up, we used 2D-Optical Imaging Spectroscopy (2D-OIS) to measure changes in cerebral haemodynamics within the sensory cortex of the brain during either 2 s whisker stimulation or spontaneous (no whisker stimulation) experiments, whilst animals could walk on a spherical treadmill. We show that locomotion alters haemodynamic responses. The amount and timing of locomotion relative to whisker stimulation is important, and can significantly impact sensory-evoked haemodynamic responses. If locomotion occurred before or during whisker stimulation, the amplitude of the stimulus-evoked haemodynamic response was significantly altered. Therefore, monitoring of locomotion during awake imaging is necessary to ensure that conclusions based on comparisons of evoked haemodynamic responses (e.g., between control and disease groups) are not confounded by the effects of locomotion.
Collapse
|
22
|
Tran CHT. Toolbox for studying neurovascular coupling in vivo, with a focus on vascular activity and calcium dynamics in astrocytes. NEUROPHOTONICS 2022; 9:021909. [PMID: 35295714 PMCID: PMC8920490 DOI: 10.1117/1.nph.9.2.021909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/23/2022] [Indexed: 05/14/2023]
Abstract
Significance: Insights into the cellular activity of each member of the neurovascular unit (NVU) is critical for understanding their contributions to neurovascular coupling (NVC)-one of the key control mechanisms in cerebral blood flow regulation. Advances in imaging and genetic tools have enhanced our ability to observe, manipulate and understand the cellular activity of NVU components, namely neurons, astrocytes, microglia, endothelial cells, vascular smooth muscle cells, and pericytes. However, there are still many unresolved questions. Since astrocytes are considered electrically unexcitable,Ca 2 + signaling is the main parameter used to monitor their activity. It is therefore imperative to study astrocyticCa 2 + dynamics simultaneously with vascular activity using tools appropriate for the question of interest. Aim: To highlight currently available genetic and imaging tools for studying the NVU-and thus NVC-with a focus on astrocyteCa 2 + dynamics and vascular activity, and discuss the utility, technical advantages, and limitations of these tools for elucidating NVC mechanisms. Approach: We draw attention to some outstanding questions regarding the mechanistic basis of NVC and emphasize the role of astrocyticCa 2 + elevations in functional hyperemia. We further discuss commonly used genetic, and optical imaging tools, as well as some newly developed imaging modalities for studying NVC at the cellular level, highlighting their advantages and limitations. Results: We provide an overview of the current state of NVC research, focusing on the role of astrocyticCa 2 + elevations in functional hyperemia; summarize recent advances in genetically engineeredCa 2 + indicators, fluorescence microscopy techniques for studying NVC; and discuss the unmet challenges for future imaging development. Conclusions: Advances in imaging techniques together with improvements in genetic tools have significantly contributed to our understanding of NVC. Many pieces of the puzzle have been revealed, but many more remain to be discovered. Ultimately, optimizing NVC research will require a concerted effort to improve imaging techniques, available genetic tools, and analytical software.
Collapse
Affiliation(s)
- Cam Ha T. Tran
- University of Nevada, Reno School of Medicine, Department of Physiology and Cell Biology, Reno, Nevada, United States
| |
Collapse
|
23
|
Del Franco AP, Chiang PP, Newman EA. Dilation of cortical capillaries is not related to astrocyte calcium signaling. Glia 2022; 70:508-521. [PMID: 34767261 PMCID: PMC8732319 DOI: 10.1002/glia.24119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 12/30/2022]
Abstract
The brain requires an adequate supply of oxygen and nutrients to maintain proper function as neuronal activity varies. This is achieved, in part, through neurovascular coupling mechanisms that mediate local increases in blood flow through the dilation of arterioles and capillaries. The role of astrocytes in mediating this functional hyperemia response is controversial. Specifically, the function of astrocyte Ca2+ signaling is unclear. Cortical arterioles dilate in the absence of astrocyte Ca2+ signaling, but previous work suggests that Ca2+ increases are necessary for capillary dilation. This question has not been fully addressed in vivo, however, and we have reexamined the role of astrocyte Ca2+ signaling in vessel dilation in the barrel cortex of awake, behaving mice. We recorded evoked vessel dilations and astrocyte Ca2+ signaling in response to whisker stimulation. Experiments were carried out on WT and IP3R2 KO mice, a transgenic model where astrocyte Ca2+ signaling is substantially reduced. Compared to WT mice at rest, Ca2+ signaling in astrocyte endfeet contacting capillaries increased by 240% when whisker stimulation evoked running. In contrast, Ca2+ signaling was reduced to 9% of WT values in IP3R2 KO mice. In all three conditions, however, the amplitude of capillary dilation was largely unchanged. In addition, the latency to the onset of astrocyte Ca2+ signaling lagged behind dilation onset in most trials, although a subset of rapid onset Ca2+ events with latencies as short as 0.15 s occurred. In summary, we found that whisker stimulation-evoked capillary dilations occurred independent of astrocyte Ca2+ increases in the cerebral cortex.
Collapse
Affiliation(s)
- Armani P Del Franco
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
24
|
Bach P, Schuster R, Koopmann A, Vollstaedt-Klein S, Spanagel R, Kiefer F. Plasma calcium concentration during detoxification predicts neural cue-reactivity and craving during early abstinence in alcohol-dependent patients. Eur Arch Psychiatry Clin Neurosci 2022; 272:341-348. [PMID: 33630132 PMCID: PMC8866328 DOI: 10.1007/s00406-021-01240-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/12/2021] [Indexed: 11/26/2022]
Abstract
Recent studies on the pathophysiology of alcohol dependence suggest a link between peripheral calcium concentrations and alcohol craving. Here, we investigated the association between plasma calcium concentration, cue-induced brain activation, and alcohol craving. Plasma calcium concentrations were measured at the onset of inpatient detoxification in a sample of N = 115 alcohol-dependent patients. Alcohol cue-reactivity was assessed during early abstinence (mean 11.1 days) using a functional magnetic resonance imaging (fMRI) alcohol cue-reactivity task. Multiple regression analyses and bivariate correlations between plasma calcium concentrations, clinical craving measures and neural alcohol cue-reactivity (CR) were tested. Results show a significant negative correlation between plasma calcium concentrations and compulsive alcohol craving. Higher calcium levels predicted higher alcohol cue-induced brain response in a cluster of frontal brain areas, including the dorsolateral prefrontal cortex (dlPFC), the anterior prefrontal cortex (alPFC), and the inferior (IFG) and middle frontal gyri (MFG). In addition, functional brain activation in those areas correlated negatively with craving for alcohol during fMRI. Higher peripheral calcium concentrations during withdrawal predicted increased alcohol cue-induced brain activation in frontal brain areas, which are associated with craving inhibition and cognitive control functions. This might indicate that higher plasma calcium concentrations at onset of detoxification could modulate craving inhibition during early abstinence.Trial registration number: DRKS00003388; date of registration: 14.12.2011.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Heidelberg University, Central Institute of Mental Health, Square J5, 68159, Mannheim, Germany
- Feuerlein Center on Translational Addiction Medicine, Heidelberg University, Heidelberg, Germany
| | - Rilana Schuster
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Heidelberg University, Central Institute of Mental Health, Square J5, 68159, Mannheim, Germany.
- Feuerlein Center on Translational Addiction Medicine, Heidelberg University, Heidelberg, Germany.
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstraße 200 a, 69118, Heidelberg, Germany.
| | - Anne Koopmann
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Heidelberg University, Central Institute of Mental Health, Square J5, 68159, Mannheim, Germany
- Feuerlein Center on Translational Addiction Medicine, Heidelberg University, Heidelberg, Germany
| | - Sabine Vollstaedt-Klein
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Heidelberg University, Central Institute of Mental Health, Square J5, 68159, Mannheim, Germany
- Feuerlein Center on Translational Addiction Medicine, Heidelberg University, Heidelberg, Germany
| | - Rainer Spanagel
- Medical Faculty Mannheim, Heidelberg University, Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Heidelberg University, Central Institute of Mental Health, Square J5, 68159, Mannheim, Germany
- Feuerlein Center on Translational Addiction Medicine, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
25
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
26
|
Ahmadpour N, Kantroo M, Stobart JL. Extracellular Calcium Influx Pathways in Astrocyte Calcium Microdomain Physiology. Biomolecules 2021; 11:1467. [PMID: 34680100 PMCID: PMC8533159 DOI: 10.3390/biom11101467] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are complex glial cells that play many essential roles in the brain, including the fine-tuning of synaptic activity and blood flow. These roles are linked to fluctuations in intracellular Ca2+ within astrocytes. Recent advances in imaging techniques have identified localized Ca2+ transients within the fine processes of the astrocytic structure, which we term microdomain Ca2+ events. These Ca2+ transients are very diverse and occur under different conditions, including in the presence or absence of surrounding circuit activity. This complexity suggests that different signalling mechanisms mediate microdomain events which may then encode specific astrocyte functions from the modulation of synapses up to brain circuits and behaviour. Several recent studies have shown that a subset of astrocyte microdomain Ca2+ events occur rapidly following local neuronal circuit activity. In this review, we consider the physiological relevance of microdomain astrocyte Ca2+ signalling within brain circuits and outline possible pathways of extracellular Ca2+ influx through ionotropic receptors and other Ca2+ ion channels, which may contribute to astrocyte microdomain events with potentially fast dynamics.
Collapse
Affiliation(s)
| | | | - Jillian L. Stobart
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MG R3E 0T5, Canada; (N.A.); (M.K.)
| |
Collapse
|
27
|
Abstract
The cerebral microcirculation undergoes dynamic changes in parallel with the development of neurons, glia, and their energy metabolism throughout gestation and postnatally. Cerebral blood flow (CBF), oxygen consumption, and glucose consumption are as low as 20% of adult levels in humans born prematurely but eventually exceed adult levels at ages 3 to 11 years, which coincide with the period of continued brain growth, synapse formation, synapse pruning, and myelination. Neurovascular coupling to sensory activation is present but attenuated at birth. By 2 postnatal months, the increase in CBF often is disproportionately smaller than the increase in oxygen consumption, in contrast to the relative hyperemia seen in adults. Vascular smooth muscle myogenic tone increases in parallel with developmental increases in arterial pressure. CBF autoregulatory response to increased arterial pressure is intact at birth but has a more limited range with arterial hypotension. Hypoxia-induced vasodilation in preterm fetal sheep with low oxygen consumption does not sustain cerebral oxygen transport, but the response becomes better developed for sustaining oxygen transport by term. Nitric oxide tonically inhibits vasomotor tone, and glutamate receptor activation can evoke its release in lambs and piglets. In piglets, astrocyte-derived carbon monoxide plays a central role in vasodilation evoked by glutamate, ADP, and seizures, and prostanoids play a large role in endothelial-dependent and hypercapnic vasodilation. Overall, homeostatic mechanisms of CBF regulation in response to arterial pressure, neuronal activity, carbon dioxide, and oxygenation are present at birth but continue to develop postnatally as neurovascular signaling pathways are dynamically altered and integrated. © 2021 American Physiological Society. Compr Physiol 11:1-62, 2021.
Collapse
|
28
|
Boily M, Li L, Vallerand D, Girouard H. Angiotensin II Disrupts Neurovascular Coupling by Potentiating Calcium Increases in Astrocytic Endfeet. J Am Heart Assoc 2021; 10:e020608. [PMID: 34459216 PMCID: PMC8649296 DOI: 10.1161/jaha.120.020608] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/09/2021] [Indexed: 11/16/2022]
Abstract
Background Angiotensin II (Ang II), a critical mediator of hypertension, impairs neurovascular coupling. Since astrocytes are key regulators of neurovascular coupling, we sought to investigate whether Ang II impairs neurovascular coupling through modulation of astrocytic Ca2+ signaling. Methods and Results Using laser Doppler flowmetry, we found that Ang II attenuates cerebral blood flow elevations induced by whisker stimulation or the metabotropic glutamate receptors agonist, 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.01). In acute brain slices, Ang II shifted the vascular response induced by 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid towards vasoconstriction (P<0.05). The resting and 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid-induced Ca2+ levels in the astrocytic endfeet were more elevated in the presence of Ang II (P<0.01). Both effects were reversed by the AT1 receptor antagonist, candesartan (P<0.01 for diameter and P<0.05 for calcium levels). Using photolysis of caged Ca2+ in astrocytic endfeet or pre-incubation of 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis (acetoxymethyl ester), we demonstrated the link between potentiated Ca2+ elevation and impaired vascular response in the presence of Ang II (P<0.001 and P<0.05, respectively). Both intracellular Ca2+ mobilization and Ca2+ influx through transient receptor potential vanilloid 4 mediated Ang II-induced astrocytic Ca2+ elevation, since blockade of these pathways significantly prevented the intracellular Ca2+ in response to 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.05). Conclusions These results suggest that Ang II through its AT1 receptor potentiates the astrocytic Ca2+ responses to a level that promotes vasoconstriction over vasodilation, thus altering cerebral blood flow increases in response to neuronal activity.
Collapse
Affiliation(s)
- Michaël Boily
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Lin Li
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Diane Vallerand
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
| | - Hélène Girouard
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
- Centre de Recherche de l’Institut de Gériatrie de MontréalMontréalQuébecCanada
| |
Collapse
|
29
|
Haidey JN, Peringod G, Institoris A, Gorzo KA, Nicola W, Vandal M, Ito K, Liu S, Fielding C, Visser F, Nguyen MD, Gordon GR. Astrocytes regulate ultra-slow arteriole oscillations via stretch-mediated TRPV4-COX-1 feedback. Cell Rep 2021; 36:109405. [PMID: 34348138 DOI: 10.1016/j.celrep.2021.109405] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/22/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022] Open
Abstract
Very-low-frequency oscillations in microvascular diameter cause fluctuations in oxygen delivery that are important for fueling the brain and for functional imaging. However, little is known about how the brain regulates ongoing oscillations in cerebral blood flow. In mouse and rat cortical brain slice arterioles, we find that selectively enhancing tone is sufficient to recruit a TRPV4-mediated Ca2+ elevation in adjacent astrocyte endfeet. This endfoot Ca2+ signal triggers COX-1-mediated "feedback vasodilators" that limit the extent of evoked vasoconstriction, as well as constrain fictive vasomotion in slices. Astrocyte-Ptgs1 knockdown in vivo increases the power of arteriole oscillations across a broad range of very low frequencies (0.01-0.3 Hz), including ultra-slow vasomotion (∼0.1 Hz). Conversely, clamping astrocyte Ca2+in vivo reduces the power of vasomotion. These data demonstrate bidirectional communication between arterioles and astrocyte endfeet to regulate oscillatory microvasculature activity.
Collapse
Affiliation(s)
- Jordan N Haidey
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Govind Peringod
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kelsea A Gorzo
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Wilten Nicola
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Physics and Astronomy, Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Milène Vandal
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kenichi Ito
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shiying Liu
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cameron Fielding
- Centre for Genome Engineering, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Frank Visser
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
30
|
Lohr C, Beiersdorfer A, Fischer T, Hirnet D, Rotermund N, Sauer J, Schulz K, Gee CE. Using Genetically Encoded Calcium Indicators to Study Astrocyte Physiology: A Field Guide. Front Cell Neurosci 2021; 15:690147. [PMID: 34177468 PMCID: PMC8226001 DOI: 10.3389/fncel.2021.690147] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/06/2021] [Indexed: 01/14/2023] Open
Abstract
Ca2+ imaging is the most frequently used technique to study glial cell physiology. While chemical Ca2+ indicators served to visualize and measure changes in glial cell cytosolic Ca2+ concentration for several decades, genetically encoded Ca2+ indicators (GECIs) have become state of the art in recent years. Great improvements have been made since the development of the first GECI and a large number of GECIs with different physical properties exist, rendering it difficult to select the optimal Ca2+ indicator. This review discusses some of the most frequently used GECIs and their suitability for glial cell research.
Collapse
Affiliation(s)
- Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | | | - Timo Fischer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Daniela Hirnet
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Natalie Rotermund
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Jessica Sauer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Kristina Schulz
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Christine E Gee
- Institute of Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
31
|
Sancho L, Contreras M, Allen NJ. Glia as sculptors of synaptic plasticity. Neurosci Res 2021; 167:17-29. [PMID: 33316304 PMCID: PMC8513541 DOI: 10.1016/j.neures.2020.11.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
Glial cells are non-neuronal cells in the nervous system that are crucial for proper brain development and function. Three major classes of glia in the central nervous system (CNS) include astrocytes, microglia and oligodendrocytes. These cells have dynamic morphological and functional properties and constantly surveil neural activity throughout life, sculpting synaptic plasticity. Astrocytes form part of the tripartite synapse with neurons and perform many homeostatic functions essential to proper synaptic function including clearing neurotransmitter and regulating ion balance; they can modify these properties, in addition to additional mechanisms such as gliotransmitter release, to influence short- and long-term plasticity. Microglia, the resident macrophage of the CNS, monitor synaptic activity and can eliminate synapses by phagocytosis or modify synapses by release of cytokines or neurotrophic factors. Oligodendrocytes regulate speed of action potential conduction and efficiency of information exchange through the formation of myelin, having important consequences for the plasticity of neural circuits. A deeper understanding of how glia modulate synaptic and circuit plasticity will further our understanding of the ongoing changes that take place throughout life in the dynamic environment of the CNS.
Collapse
Affiliation(s)
- Laura Sancho
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Minerva Contreras
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
32
|
Lim D, Semyanov A, Genazzani A, Verkhratsky A. Calcium signaling in neuroglia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:1-53. [PMID: 34253292 DOI: 10.1016/bs.ircmb.2021.01.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glial cells exploit calcium (Ca2+) signals to perceive the information about the activity of the nervous tissue and the tissue environment to translate this information into an array of homeostatic, signaling and defensive reactions. Astrocytes, the best studied glial cells, use several Ca2+ signaling generation pathways that include Ca2+ entry through plasma membrane, release from endoplasmic reticulum (ER) and from mitochondria. Activation of metabotropic receptors on the plasma membrane of glial cells is coupled to an enzymatic cascade in which a second messenger, InsP3 is generated thus activating intracellular Ca2+ release channels in the ER endomembrane. Astrocytes also possess store-operated Ca2+ entry and express several ligand-gated Ca2+ channels. In vivo astrocytes generate heterogeneous Ca2+ signals, which are short and frequent in distal processes, but large and relatively rare in soma. In response to neuronal activity intracellular and inter-cellular astrocytic Ca2+ waves can be produced. Astrocytic Ca2+ signals are involved in secretion, they regulate ion transport across cell membranes, and are contributing to cell morphological plasticity. Therefore, astrocytic Ca2+ signals are linked to fundamental functions of the central nervous system ranging from synaptic transmission to behavior. In oligodendrocytes, Ca2+ signals are generated by plasmalemmal Ca2+ influx, or by release from intracellular stores, or by combination of both. Microglial cells exploit Ca2+ permeable ionotropic purinergic receptors and transient receptor potential channels as well as ER Ca2+ release. In this contribution, basic morphology of glial cells, glial Ca2+ signaling toolkit, intracellular Ca2+ signals and Ca2+-regulated functions are discussed with focus on astrocytes.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Faculty of Biology, Moscow State University, Moscow, Russia; Sechenov First Moscow State Medical University, Moscow, Russia
| | - Armando Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Alexei Verkhratsky
- Sechenov First Moscow State Medical University, Moscow, Russia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
33
|
Tarantini S, Balasubramanian P, Yabluchanskiy A, Ashpole NM, Logan S, Kiss T, Ungvari A, Nyúl-Tóth Á, Schwartzman ML, Benyo Z, Sonntag WE, Csiszar A, Ungvari Z. IGF1R signaling regulates astrocyte-mediated neurovascular coupling in mice: implications for brain aging. GeroScience 2021; 43:901-911. [PMID: 33674953 PMCID: PMC8110646 DOI: 10.1007/s11357-021-00350-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is associated with a significant deficiency in circulating insulin-like growth factor-1 (IGF-1), which has an important role in the pathogenesis of age-related vascular cognitive impairment (VCI). Impairment of moment-to-moment adjustment of regional cerebral blood flow via neurovascular coupling (NVC) importantly contributes to VCI. Previous studies established a causal link between circulating IGF-1 deficiency and neurovascular dysfunction. Release of vasodilator mediators from activated astrocytes plays a key role in NVC. To determine the impact of impaired IGF-1 signaling on astrocytic function, astrocyte-mediated NVC responses were studied in a novel mouse model of astrocyte-specific knockout of IGF1R (GFAP-CreERT2/Igf1rf/f) and accelerated neurovascular aging. We found that mice with disrupted astrocytic IGF1R signaling exhibit impaired NVC responses, decreased stimulated release of the vasodilator gliotransmitter epoxy-eicosatrienoic acids (EETs), and upregulation of soluble epoxy hydrolase (sEH), which metabolizes and inactivates EETs. Collectively, our findings provide additional evidence that IGF-1 promotes astrocyte health and maintains normal NVC, protecting cognitive health.
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nicole M Ashpole
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Pharmacology Division, Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, Oxford, MS, USA
| | - Sreemathi Logan
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Rehabilitation Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
- Vascular Cognitive Impairment and Neurodegeneration Program/HCEMM, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Michal L Schwartzman
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, NY, USA
| | - Zoltan Benyo
- Vascular Cognitive Impairment and Neurodegeneration Program/HCEMM, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - William E Sonntag
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
- Vascular Cognitive Impairment and Neurodegeneration Program/HCEMM, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Reynolds Oklahoma Center on Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
- Vascular Cognitive Impairment and Neurodegeneration Program/HCEMM, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
34
|
Vaidyanathan TV, Collard M, Yokoyama S, Reitman ME, Poskanzer KE. Cortical astrocytes independently regulate sleep depth and duration via separate GPCR pathways. eLife 2021; 10:63329. [PMID: 33729913 PMCID: PMC7968927 DOI: 10.7554/elife.63329] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Non-rapid eye movement (NREM) sleep, characterized by slow-wave electrophysiological activity, underlies several critical functions, including learning and memory. However, NREM sleep is heterogeneous, varying in duration, depth, and spatially across the cortex. While these NREM sleep features are thought to be largely independently regulated, there is also evidence that they are mechanistically coupled. To investigate how cortical NREM sleep features are controlled, we examined the astrocytic network, comprising a cortex-wide syncytium that influences population-level neuronal activity. We quantified endogenous astrocyte activity in mice over natural sleep and wake, then manipulated specific astrocytic G-protein-coupled receptor (GPCR) signaling pathways in vivo. We find that astrocytic Gi- and Gq-coupled GPCR signaling separately control NREM sleep depth and duration, respectively, and that astrocytic signaling causes differential changes in local and remote cortex. These data support a model in which the cortical astrocyte network serves as a hub for regulating distinct NREM sleep features. Sleep has many roles, from strengthening new memories to regulating mood and appetite. While we might instinctively think of sleep as a uniform state of reduced brain activity, the reality is more complex. First, over the course of the night, we cycle between a number of different sleep stages, which reflect different levels of sleep depth. Second, the amount of sleep depth is not necessarily even across the brain but can vary between regions. These sleep stages consist of either rapid eye movement (REM) sleep or non-REM (NREM) sleep. REM sleep is when most dreaming occurs, whereas NREM sleep is particularly important for learning and memory and can vary in duration and depth. During NREM sleep, large groups of neurons synchronize their firing to create rhythmic waves of activity known as slow waves. The more synchronous the activity, the deeper the sleep. Vaidyanathan et al. now show that brain cells called astrocytes help regulate NREM sleep. Astrocytes are not neurons but belong to a group of specialized cells called glia. They are the largest glia cell type in the brain and display an array of proteins on their surfaces called G-protein-coupled receptors (GPCRs). These enable them to sense sleep-wake signals from other parts of the brain and to generate their own signals. In fact, each astrocyte can communicate with thousands of neurons at once. They are therefore well-poised to coordinate brain activity during NREM sleep. Using innovative tools, Vaidyanathan et al. visualized astrocyte activity in mice as the animals woke up or fell asleep. The results showed that astrocytes change their activity just before each sleep–wake transition. They also revealed that astrocytes control both the depth and duration of NREM sleep via two different types of GPCR signals. Increasing one of these signals (Gi-GPCR) made the mice sleep more deeply but did not change sleep duration. Decreasing the other (Gq-GPCR) made the mice sleep for longer but did not affect sleep depth. Sleep problems affect many people at some point in their lives, and often co-exist with other conditions such as mental health disorders. Understanding how the brain regulates different features of sleep could help us develop better – and perhaps more specific – treatments for sleep disorders. The current study suggests that manipulating GPCRs on astrocytes might increase sleep depth, for example. But before work to test this idea can begin, we must first determine whether findings from sleeping mice also apply to people.
Collapse
Affiliation(s)
- Trisha V Vaidyanathan
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Max Collard
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Sae Yokoyama
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Michael E Reitman
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Kira E Poskanzer
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, San Francisco, United States
| |
Collapse
|
35
|
Verisokin AY, Verveyko DV, Postnov DE, Brazhe AR. Modeling of Astrocyte Networks: Toward Realistic Topology and Dynamics. Front Cell Neurosci 2021; 15:645068. [PMID: 33746715 PMCID: PMC7973220 DOI: 10.3389/fncel.2021.645068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal firing and neuron-to-neuron synaptic wiring are currently widely described as orchestrated by astrocytes—elaborately ramified glial cells tiling the cortical and hippocampal space into non-overlapping domains, each covering hundreds of individual dendrites and hundreds thousands synapses. A key component to astrocytic signaling is the dynamics of cytosolic Ca2+ which displays multiscale spatiotemporal patterns from short confined elemental Ca2+ events (puffs) to Ca2+ waves expanding through many cells. Here, we synthesize the current understanding of astrocyte morphology, coupling local synaptic activity to astrocytic Ca2+ in perisynaptic astrocytic processes and morphology-defined mechanisms of Ca2+ regulation in a distributed model. To this end, we build simplified realistic data-driven spatial network templates and compile model equations as defined by local cell morphology. The input to the model is spatially uncorrelated stochastic synaptic activity. The proposed modeling approach is validated by statistics of simulated Ca2+ transients at a single cell level. In multicellular templates we observe regular sequences of cell entrainment in Ca2+ waves, as a result of interplay between stochastic input and morphology variability between individual astrocytes. Our approach adds spatial dimension to the existing astrocyte models by employment of realistic morphology while retaining enough flexibility and scalability to be embedded in multiscale heterocellular models of neural tissue. We conclude that the proposed approach provides a useful description of neuron-driven Ca2+-activity in the astrocyte syncytium.
Collapse
Affiliation(s)
| | - Darya V Verveyko
- Department of Theoretical Physics, Kursk State University, Kursk, Russia
| | - Dmitry E Postnov
- Department of Optics and Biophotonics, Saratov State University, Saratov, Russia
| | - Alexey R Brazhe
- Department of Biophysics, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia.,Department of Molecular Neurobiology, Institute of Bioorganic Chemistry RAS, Russian Federation, Moscow, Russia
| |
Collapse
|
36
|
Moshkforoush A, Balachandar L, Moncion C, Montejo KA, Riera J. Unraveling ChR2-driven stochastic Ca2+ dynamics in astrocytes: A call for new interventional paradigms. PLoS Comput Biol 2021; 17:e1008648. [PMID: 33566841 PMCID: PMC7875401 DOI: 10.1371/journal.pcbi.1008648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023] Open
Abstract
Optogenetic targeting of astrocytes provides a robust experimental model to differentially induce Ca2+ signals in astrocytes in vivo. However, a systematic study quantifying the response of optogenetically modified astrocytes to light is yet to be performed. Here, we propose a novel stochastic model of Ca2+ dynamics in astrocytes that incorporates a light sensitive component-channelrhodopsin 2 (ChR2). Utilizing this model, we investigated the effect of different light stimulation paradigms on cells expressing select variants of ChR2 (wild type, ChETA, and ChRET/TC). Results predict that depending on paradigm specification, astrocytes might undergo drastic changes in their basal Ca2+ level and spiking probability. Furthermore, we performed a global sensitivity analysis to assess the effect of variation in parameters pertinent to the shape of the ChR2 photocurrent on astrocytic Ca2+ dynamics. Results suggest that directing variants towards the first open state of the ChR2 photocycle (o1) enhances spiking activity in astrocytes during optical stimulation. Evaluation of the effect of Ca2+ buffering and coupling coefficient in a network of ChR2-expressing astrocytes demonstrated basal level elevations in the stimulated region and propagation of calcium activity to unstimulated cells. Buffering reduced the diffusion range of Ca2+ within the network, thereby limiting propagation and influencing the activity of astrocytes. Collectively, the framework presented in this study provides valuable information for the selection of light stimulation paradigms that elicit desired astrocytic activity using existing ChR2 constructs, as well as aids in the engineering of future application-oriented optogenetic variants.
Collapse
Affiliation(s)
- Arash Moshkforoush
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Lakshmini Balachandar
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Carolina Moncion
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
| | - Karla A. Montejo
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jorge Riera
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
37
|
Howarth C, Mishra A, Hall CN. More than just summed neuronal activity: how multiple cell types shape the BOLD response. Philos Trans R Soc Lond B Biol Sci 2021; 376:20190630. [PMID: 33190598 PMCID: PMC7116385 DOI: 10.1098/rstb.2019.0630] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Functional neuroimaging techniques are widely applied to investigations of human cognition and disease. The most commonly used among these is blood oxygen level-dependent (BOLD) functional magnetic resonance imaging. The BOLD signal occurs because neural activity induces an increase in local blood supply to support the increased metabolism that occurs during activity. This supply usually outmatches demand, resulting in an increase in oxygenated blood in an active brain region, and a corresponding decrease in deoxygenated blood, which generates the BOLD signal. Hence, the BOLD response is shaped by an integration of local oxygen use, through metabolism, and supply, in the blood. To understand what information is carried in BOLD signals, we must understand how several cell types in the brain-local excitatory neurons, inhibitory neurons, astrocytes and vascular cells (pericytes, vascular smooth muscle and endothelial cells), and their modulation by ascending projection neurons-contribute to both metabolism and haemodynamic changes. Here, we review the contributions of each cell type to the regulation of cerebral blood flow and metabolism, and discuss situations where a simplified interpretation of the BOLD response as reporting local excitatory activity may misrepresent important biological phenomena, for example with regards to arousal states, ageing and neurological disease. This article is part of the theme issue 'Key relationships between non-invasive functional neuroimaging and the underlying neuronal activity'.
Collapse
Affiliation(s)
- Clare Howarth
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|
38
|
Sharma K, Gordon GRJ, Tran CHT. Heterogeneity of Sensory-Induced Astrocytic Ca 2+ Dynamics During Functional Hyperemia. Front Physiol 2020; 11:611884. [PMID: 33362585 PMCID: PMC7758506 DOI: 10.3389/fphys.2020.611884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022] Open
Abstract
Astrocytic Ca2+ fluctuations associated with functional hyperemia have typically been measured from large cellular compartments such as the soma, the whole arbor and the endfoot. The most prominent Ca2+ event is a large magnitude, delayed signal that follows vasodilation. However, previous work has provided little information about the spatio-temporal properties of such Ca2+ transients or their heterogeneity. Here, using an awake, in vivo two-photon fluorescence-imaging model, we performed detailed profiling of delayed astrocytic Ca2+ signals across astrocytes or within individual astrocyte compartments using small regions of interest next to penetrating arterioles and capillaries along with vasomotor responses to vibrissae stimulation. We demonstrated that while a 5-s air puff that stimulates all whiskers predominantly generated reproducible functional hyperemia in the presence or absence of astrocytic Ca2+ changes, whisker stimulation inconsistently produced astrocytic Ca2+ responses. More importantly, these Ca2+ responses were heterogeneous among subcellular structures of the astrocyte and across different astrocytes that resided within the same field of view. Furthermore, we found that whisker stimulation induced discrete Ca2+ “hot spots” that spread regionally within the endfoot. These data reveal that astrocytic Ca2+ dynamics associated with the microvasculature are more complex than previously thought, and highlight the importance of considering the heterogeneity of astrocytic Ca2+ activity to fully understanding neurovascular coupling.
Collapse
Affiliation(s)
- Kushal Sharma
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Grant R J Gordon
- Department of Physiology and Pharmacology, School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Cam Ha T Tran
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| |
Collapse
|
39
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
40
|
Okubo Y. Astrocytic Ca2+ signaling mediated by the endoplasmic reticulum in health and disease. J Pharmacol Sci 2020; 144:83-88. [DOI: 10.1016/j.jphs.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
|
41
|
Do CT, Manjaly ZM, Heinzle J, Schöbi D, Kasper L, Pruessmann KP, Stephan KE, Frässle S. Hemodynamic modeling of long-term aspirin effects on blood oxygenated level dependent responses at 7 Tesla in patients at cardiovascular risk. Eur J Neurosci 2020; 53:1262-1278. [PMID: 32936980 DOI: 10.1111/ejn.14970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 08/06/2020] [Accepted: 09/02/2020] [Indexed: 11/29/2022]
Abstract
Aspirin is considered a potential confound for functional magnetic resonance imaging (fMRI) studies. This is because aspirin affects the synthesis of prostaglandin, a vasoactive mediator centrally involved in neurovascular coupling, a process underlying blood oxygenated level dependent (BOLD) responses. Aspirin-induced changes in BOLD signal are a potential confound for fMRI studies of at-risk individuals or patients (e.g. with cardiovascular conditions or stroke) who receive low-dose aspirin prophylactically and are compared to healthy controls without aspirin. To examine the severity of this potential confound, we combined high field (7 Tesla) MRI during a simple hand movement task with a biophysically informed hemodynamic model. We compared elderly individuals receiving aspirin for primary or secondary prophylactic purposes versus age-matched volunteers without aspirin medication, testing for putative differences in BOLD responses. Specifically, we fitted hemodynamic models to BOLD responses from 14 regions activated by the task and examined whether model parameter estimates were significantly altered by aspirin. While our analyses indicate that hemodynamics differed across regions, consistent with the known regional variability of BOLD responses, we neither found a significant main effect of aspirin (i.e., an average effect across brain regions) nor an expected drug × region interaction. While our sample size is not sufficiently large to rule out small-to-medium global effects of aspirin, we had adequate statistical power for detecting the expected interaction. Altogether, our analysis suggests that patients with cardiovascular risk receiving low-dose aspirin for primary or secondary prophylactic purposes do not show strongly altered BOLD signals when compared to healthy controls without aspirin.
Collapse
Affiliation(s)
- Cao-Tri Do
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Zina-Mary Manjaly
- Department of Neurology, Schulthess Clinic, Zurich, Switzerland.,Department of Health Science and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
| | - Jakob Heinzle
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Dario Schöbi
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Lars Kasper
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.,MR Technology Group, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Klaas P Pruessmann
- MR Technology Group, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.,Wellcome Centre for Human Neuroimaging, University College London, London, UK.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Stefan Frässle
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Verkhratsky A, Semyanov A, Zorec R. Physiology of Astroglial Excitability. FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa016. [PMID: 35330636 PMCID: PMC8788756 DOI: 10.1093/function/zqaa016] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023]
Abstract
Classic physiology divides all neural cells into excitable neurons and nonexcitable neuroglia. Neuroglial cells, chiefly responsible for homeostasis and defense of the nervous tissue, coordinate their complex homeostatic responses with neuronal activity. This coordination reflects a specific form of glial excitability mediated by complex changes in intracellular concentration of ions and second messengers organized in both space and time. Astrocytes are equipped with multiple molecular cascades, which are central for regulating homeostasis of neurotransmitters, ionostasis, synaptic connectivity, and metabolic support of the central nervous system. Astrocytes are further provisioned with multiple receptors for neurotransmitters and neurohormones, which upon activation trigger intracellular signals mediated by Ca2+, Na+, and cyclic AMP. Calcium signals have distinct organization and underlying mechanisms in different astrocytic compartments thus allowing complex spatiotemporal signaling. Signals mediated by fluctuations in cytosolic Na+ are instrumental for coordination of Na+ dependent astrocytic transporters with tissue state and homeostatic demands. Astroglial ionic excitability may also involve K+, H+, and Cl-. The cyclic AMP signalling system is, in comparison to ions, much slower in targeting astroglial effector mechanisms. This evidence review summarizes the concept of astroglial intracellular excitability.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK,Achucarro Center for Neuroscience, Ikerbasque, 48011 Bilbao, Spain,Address correspondence to A.V. (e-mail: )
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia,Faculty of Biology, Moscow State University, Moscow, Russia,Sechenov First Moscow State Medical University, Moscow, Russia
| | - Robert Zorec
- Celica Biomedical, Ljubljana 1000, Slovenia,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| |
Collapse
|
43
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
44
|
The capillary Kir channel as sensor and amplifier of neuronal signals: Modeling insights on K +-mediated neurovascular communication. Proc Natl Acad Sci U S A 2020; 117:16626-16637. [PMID: 32601236 DOI: 10.1073/pnas.2000151117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuronal activity leads to an increase in local cerebral blood flow (CBF) to allow adequate supply of oxygen and nutrients to active neurons, a process termed neurovascular coupling (NVC). We have previously shown that capillary endothelial cell (cEC) inwardly rectifying K+ (Kir) channels can sense neuronally evoked increases in interstitial K+ and induce rapid and robust dilations of upstream parenchymal arterioles, suggesting a key role of cECs in NVC. The requirements of this signal conduction remain elusive. Here, we utilize mathematical modeling to investigate how small outward currents in stimulated cECs can elicit physiologically relevant spread of vasodilatory signals within the highly interconnected brain microvascular network to increase local CBF. Our model shows that the Kir channel can act as an "on-off" switch in cECs to hyperpolarize the cell membrane as extracellular K+ increases. A local hyperpolarization can be amplified by the voltage-dependent activation of Kir in neighboring cECs. Sufficient Kir density enables robust amplification of the hyperpolarizing stimulus and produces responses that resemble action potentials in excitable cells. This Kir-mediated excitability can remain localized in the stimulated region or regeneratively propagate over significant distances in the microvascular network, thus dramatically increasing the efficacy of K+ for eliciting local hyperemia. Modeling results show how changes in cEC transmembrane current densities and gap junctional resistances can affect K+-mediated NVC and suggest a key role for Kir as a sensor of neuronal activity and an amplifier of retrograde electrical signaling in the cerebral vasculature.
Collapse
|
45
|
Compound Chai Jin Jie Yu Tablets, Acts as An Antidepressant by Promoting Synaptic Function in the Hippocampal Neurons. DIGITAL CHINESE MEDICINE 2020. [DOI: 10.1016/j.dcmed.2020.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
46
|
Negri S, Faris P, Pellavio G, Botta L, Orgiu M, Forcaia G, Sancini G, Laforenza U, Moccia F. Group 1 metabotropic glutamate receptors trigger glutamate-induced intracellular Ca 2+ signals and nitric oxide release in human brain microvascular endothelial cells. Cell Mol Life Sci 2020; 77:2235-2253. [PMID: 31473770 PMCID: PMC11104941 DOI: 10.1007/s00018-019-03284-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/02/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022]
Abstract
Neurovascular coupling (NVC) is the mechanism whereby an increase in neuronal activity causes an increase in local cerebral blood flow (CBF) to ensure local supply of oxygen and nutrients to the activated areas. The excitatory neurotransmitter glutamate gates post-synaptic N-methyl-D-aspartate receptors to mediate extracellular Ca2+ entry and stimulate neuronal nitric oxide (NO) synthase to release NO, thereby triggering NVC. Recent work suggested that endothelial Ca2+ signals could underpin NVC by recruiting the endothelial NO synthase. For instance, acetylcholine induced intracellular Ca2+ signals followed by NO release by activating muscarinic 5 receptors in hCMEC/D3 cells, a widely employed model of human brain microvascular endothelial cells. Herein, we sought to assess whether also glutamate elicits metabotropic Ca2+ signals and NO release in hCMEC/D3 cells. Glutamate induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+]i) that was blocked by α-methyl-4-carboxyphenylglycine and phenocopied by trans-1-amino-1,3-cyclopentanedicarboxylic acid, which, respectively, block and activate group 1 metabotropic glutamate receptors (mGluRs). Accordingly, hCMEC/D3 expressed both mGluR1 and mGluR5 and the Ca2+ response to glutamate was inhibited by their pharmacological blockade with, respectively, CPCCOEt and MTEP hydrochloride. The Ca2+ response to glutamate was initiated by endogenous Ca2+ release from the endoplasmic reticulum and endolysosomal Ca2+ store through inositol-1,4,5-trisphosphate receptors and two-pore channels, respectively, and sustained by store-operated Ca2+ entry. In addition, glutamate induced robust NO release that was suppressed by pharmacological blockade of the accompanying increase in [Ca2+]i. These data demonstrate for the first time that glutamate may induce metabotropic Ca2+ signals in human brain microvascular endothelial cells. The Ca2+ response to glutamate is likely to support NVC during neuronal activity, thereby reinforcing the emerging role of brain microvascular endothelial cells in the regulation of CBF.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
- Research Center, Salahaddin University, Erbil, Kurdistan-Region of Iraq, Iraq
| | - Giorgia Pellavio
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Botta
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Matteo Orgiu
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Greta Forcaia
- Department of Experimental Medicine, University of Milano-Bicocca, Monza, Italy
| | - Giulio Sancini
- Department of Experimental Medicine, University of Milano-Bicocca, Monza, Italy
| | - Umberto Laforenza
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100, Pavia, Italy.
| |
Collapse
|
47
|
Lee L, Boorman L, Glendenning E, Christmas C, Sharp P, Redgrave P, Shabir O, Bracci E, Berwick J, Howarth C. Key Aspects of Neurovascular Control Mediated by Specific Populations of Inhibitory Cortical Interneurons. Cereb Cortex 2020; 30:2452-2464. [PMID: 31746324 PMCID: PMC7174996 DOI: 10.1093/cercor/bhz251] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 01/21/2023] Open
Abstract
Inhibitory interneurons can evoke vasodilation and vasoconstriction, making them potential cellular drivers of neurovascular coupling. However, the specific regulatory roles played by particular interneuron subpopulations remain unclear. Our purpose was therefore to adopt a cell-specific optogenetic approach to investigate how somatostatin (SST) and neuronal nitric oxide synthase (nNOS)-expressing interneurons might influence the neurovascular relationship. In mice, specific activation of SST- or nNOS-interneurons was sufficient to evoke hemodynamic changes. In the case of nNOS-interneurons, robust hemodynamic changes occurred with minimal changes in neural activity, suggesting that the ability of blood oxygen level dependent functional magnetic resonance imaging (BOLD fMRI) to reliably reflect changes in neuronal activity may be dependent on type of neuron recruited. Conversely, activation of SST-interneurons produced robust changes in evoked neural activity with shallow cortical excitation and pronounced deep layer cortical inhibition. Prolonged activation of SST-interneurons often resulted in an increase in blood volume in the centrally activated area with an accompanying decrease in blood volume in the surrounding brain regions, analogous to the negative BOLD signal. These results demonstrate the role of specific populations of cortical interneurons in the active control of neurovascular function.
Collapse
Affiliation(s)
- L Lee
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - L Boorman
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - E Glendenning
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - C Christmas
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - P Sharp
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - P Redgrave
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - O Shabir
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - E Bracci
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - J Berwick
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| | - C Howarth
- Department of Psychology, University of Sheffield, Sheffield S1 2LT, UK
| |
Collapse
|
48
|
King CM, Bohmbach K, Minge D, Delekate A, Zheng K, Reynolds J, Rakers C, Zeug A, Petzold GC, Rusakov DA, Henneberger C. Local Resting Ca 2+ Controls the Scale of Astroglial Ca 2+ Signals. Cell Rep 2020; 30:3466-3477.e4. [PMID: 32160550 PMCID: PMC7068654 DOI: 10.1016/j.celrep.2020.02.043] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/21/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Astroglia regulate neurovascular coupling while engaging in signal exchange with neurons. The underlying cellular machinery is thought to rely on astrocytic Ca2+ signals, but what controls their amplitude and waveform is poorly understood. Here, we employ time-resolved two-photon excitation fluorescence imaging in acute hippocampal slices and in cortex in vivo to find that resting [Ca2+] predicts the scale (amplitude) and the maximum (peak) of astroglial Ca2+ elevations. We bidirectionally manipulate resting [Ca2+] by uncaging intracellular Ca2+ or Ca2+ buffers and use ratiometric imaging of a genetically encoded Ca2+ indicator to establish that alterations in resting [Ca2+] change co-directionally the peak level and anti-directionally the amplitude of local Ca2+ transients. This relationship holds for spontaneous and for induced (for instance by locomotion) Ca2+ signals. Our findings uncover a basic generic rule of Ca2+ signal formation in astrocytes, thus also associating the resting Ca2+ level with the physiological "excitability" state of astroglia.
Collapse
Affiliation(s)
- Claire M King
- Institute of Neurology, University College London, London, UK
| | - Kirsten Bohmbach
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Daniel Minge
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kaiyu Zheng
- Institute of Neurology, University College London, London, UK
| | - James Reynolds
- Institute of Neurology, University College London, London, UK
| | - Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, University Hospital Bonn, Bonn, Germany
| | | | - Christian Henneberger
- Institute of Neurology, University College London, London, UK; Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
49
|
Kozachkov L, Michmizos KP. Sequence Learning in Associative Neuronal-Astrocytic Networks. Brain Inform 2020. [DOI: 10.1007/978-3-030-59277-6_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
50
|
Gu X, Chen W, Volkow ND, Koretsky AP, Du C, Pan Y. Synchronized Astrocytic Ca 2+ Responses in Neurovascular Coupling during Somatosensory Stimulation and for the Resting State. Cell Rep 2019; 23:3878-3890. [PMID: 29949771 PMCID: PMC7469112 DOI: 10.1016/j.celrep.2018.05.091] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/05/2018] [Accepted: 05/27/2018] [Indexed: 02/08/2023] Open
Abstract
The role of astrocytes in neurovascular coupling (NVC) is unclear. Here, we applied a multimodality imaging approach to concomitantly measure synchronized neuronal or astrocytic Ca2+ and hemodynamic changes in the mouse somatosensory cortex at rest and during sensory electrical stimulation. Strikingly, we found that low-frequency stimulation (0.3-1 Hz), which consistently evokes fast neuronal Ca2+ transients (6.0 ± 2.7 ms latency) that always precede vascular responses, does not always elicit astrocytic Ca2+ transients (313 ± 65 ms latency). However, the magnitude of the hemodynamic response is increased when astrocytic transients occur, suggesting a facilitatory role of astrocytes in NVC. High-frequency stimulation (5-10 Hz) consistently evokes a large, delayed astrocytic Ca2+ accumulation (3.48 ± 0.09 s latency) that is temporarily associated with vasoconstriction, suggesting a role for astrocytes in resetting NVC. At rest, neuronal, but not astrocytic, Ca2+ fluctuations correlate with hemodynamic low-frequency oscillations. Taken together, these results support a role for astrocytes in modulating, but not triggering, NVC.
Collapse
Affiliation(s)
- Xiaochun Gu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA; Jiangsu Key Laboratory of Molecular and Functional Imaging, Key Laboratory of Developmental Genes and Human Diseases, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, #87 Dingjiaqiao Road, Nanjing 210009, China
| | - Wei Chen
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20857, USA
| | - Alan P Koretsky
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|