51
|
Liang Y, Walczak P. Long term intravital single cell tracking under multiphoton microscopy. J Neurosci Methods 2020; 349:109042. [PMID: 33340557 DOI: 10.1016/j.jneumeth.2020.109042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Visualizing and tracking cells over time in a living organism has been a much-coveted dream before the invention of intravital microscopy. The opaque nature of tissue was a major hurdle that was remedied by the multiphoton microscopy. With the advancement of optical imaging and fluorescent labeling tools, intravital high resolution imaging has become increasingly accessible over the past few years. Long-term intravital tracking of single cells (LIST) under multiphoton microscopy provides a unique opportunity to gain insight into the longitudinal changes in the morphology, migration, or function of cells or subcellular structures. It is particularly suitable for studying slow-evolving cellular and molecular events during normal development or disease progression, without losing the opportunity of catching fast events such as calcium signals. Here, we review the application of LIST under 2-photon microscopy in various fields of neurobiology and discuss challenges and new directions in labeling and imaging methods for LIST. Overall, this review provides an overview of current applications of LIST in mammals, which is an emerging field that will contribute to a better understanding of essential molecular and cellular events in health and disease.
Collapse
Affiliation(s)
- Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
52
|
de Souza Melo CG, Nicolai EN, Alcaino C, Cassmann TJ, Whiteman ST, Wright AM, Miller KE, Gibbons SJ, Beyder A, Linden DR. Identification of intrinsic primary afferent neurons in mouse jejunum. Neurogastroenterol Motil 2020; 32:e13989. [PMID: 32986284 PMCID: PMC8114175 DOI: 10.1111/nmo.13989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The gut is the only organ system with intrinsic neural reflexes. Intrinsic primary afferent neurons (IPANs) of the enteric nervous system initiate intrinsic reflexes, form gut-brain connections, and undergo considerable neuroplasticity to cause digestive diseases. They remain inaccessible to study in mice in the absence of a selective marker. Advillin is used as a marker for primary afferent neurons in dorsal root ganglia. The aim of this study was to test the hypothesis that advillin is expressed in IPANs of the mouse jejunum. METHODS Advillin expression was assessed with immunohistochemistry and using transgenic mice expressing an inducible Cre recombinase under the advillin promoter were used to drive tdTomato and the genetically encoded calcium indicator GCaMP5. These mice were used to characterize the morphology and physiology of advillin-expressing enteric neurons using confocal microscopy, calcium imaging, and whole-cell patch-clamp electrophysiology. KEY RESULTS Advillin is expressed in about 25% of myenteric neurons of the mouse jejunum, and these neurons demonstrate the requisite properties of IPANs. Functionally, they demonstrate calcium responses following mechanical stimuli of the mucosa and during antidromic action potentials. They have Dogiel type II morphology with neural processes that mostly remain within the myenteric plexus, but also project to the mucosa and express NeuN and calcitonin gene-related peptide (CGRP), but not nNOS. CONCLUSIONS AND INFERENCES Advillin marks jejunal IPANs providing accessibility to this important neuronal population to study and model digestive disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - David R. Linden
- Address of Correspondence: David R. Linden, Ph.D., Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN U.S.A. 55905, Phone: 507-538-4090
- Fax: 507-284-0266,
| |
Collapse
|
53
|
Liu L, Kearns KN, Eli I, Sharifi KA, Soldozy S, Carlson EW, Scott KW, Sluzewski MF, Acton ST, Stauderman KA, Kalani MYS, Park M, Tvrdik P. Microglial Calcium Waves During the Hyperacute Phase of Ischemic Stroke. Stroke 2020; 52:274-283. [PMID: 33161850 DOI: 10.1161/strokeaha.120.032766] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Ischemic injury triggers multiple pathological responses in the brain tissue, including spreading depolarizations across the cerebral cortex (cortical spreading depolarizations [CSD]). Microglia have been recently shown to play a significant role in the propagation of CSD. However, the intracellular responses of myeloid cells during ischemic stroke have not been investigated. METHODS We have studied intracellular calcium activity in cortical microglia in the stroke model of the middle cerebral artery occlusion, using the murine Polr2a-based and Cre-dependent GCaMP5 and tdTomato reporter (PC::G5-tdT). High-speed 2-photon microscopy through cranial windows was employed to record signals from genetically encoded indicators of calcium. Inflammatory stimuli and pharmacological inhibition were used to modulate microglial calcium responses in the somatosensory cortex. RESULTS In vivo imaging revealed periodical calcium activity in microglia during the hyperacute phase of ischemic stroke. This activity was more frequent during the first 6 hours after occlusion, but the amplitudes of calcium transients became larger at later time points. Consistent with CSD nature of these events, we reproducibly triggered comparable calcium transients with microinjections of potassium chloride (KCl) into adjacent cortical areas. Furthermore, lipopolysaccharide-induced peripheral inflammation, mimicking sterile inflammation during ischemic stroke, produced significantly greater microglial calcium transients during CSD. Finally, in vivo pharmacological analysis with CRAC (calcium release-activated channel) inhibitor CM-EX-137 demonstrated that CSD-associated microglial calcium transients after KCl microinjections are mediated at least in part by the CRAC mechanism. CONCLUSIONS Our findings demonstrate that microglia participate in ischemic brain injury via previously undetected mechanisms, which may provide new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Lei Liu
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Kathryn N Kearns
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Ilyas Eli
- Department of Neurosurgery (I.E., E.W.C.), University of Utah School of Medicine, Salt Lake City
| | - Khadijeh A Sharifi
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| | - Sauson Soldozy
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Elizabeth W Carlson
- Department of Neurosurgery (I.E., E.W.C.), University of Utah School of Medicine, Salt Lake City
| | - Kyle W Scott
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - M Filip Sluzewski
- Department of Electrical and Computer Engineering (M.F.S., S.T.A.), University of Virginia Health System, Charlottesville
| | - Scott T Acton
- Department of Electrical and Computer Engineering (M.F.S., S.T.A.), University of Virginia Health System, Charlottesville
| | | | - M Yashar S Kalani
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| | - Min Park
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Petr Tvrdik
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| |
Collapse
|
54
|
Semyanov A, Henneberger C, Agarwal A. Making sense of astrocytic calcium signals — from acquisition to interpretation. Nat Rev Neurosci 2020; 21:551-564. [DOI: 10.1038/s41583-020-0361-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
55
|
Holman HA, Wan Y, Rabbitt RD. Developmental GAD2 Expression Reveals Progenitor-like Cells with Calcium Waves in Mammalian Crista Ampullaris. iScience 2020; 23:101407. [PMID: 32771977 PMCID: PMC7415930 DOI: 10.1016/j.isci.2020.101407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 01/26/2023] Open
Abstract
Sense of motion, spatial orientation, and balance in vertebrates relies on sensory hair cells in the inner ear vestibular system. Vestibular supporting cells can regenerate hair cells that are lost from aging, ototoxicity, and trauma, although not all factors or specific cell types are known. Here we report a population of GAD2-positive cells in the mouse crista ampullaris and trace GAD2 progenitor-like cells that express pluripotent transcription factors SOX2, PROX1, and CTBP2. GAD2 progenitor-like cells organize into rosettes around a central branched structure in the eminentia cruciatum (EC) herein named the EC plexus. GCaMP5G calcium indicator shows spontaneous and acetylcholine-evoked whole-cell calcium waves in neonatal and adult mice. We present a hypothetical model that outlines the lineage and potential regenerative capacity of GAD2 cells in the mammalian vestibular neuroepithelium.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Yong Wan
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Graduate Program in Neuroscience, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology-Head & Neck Surgery, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
56
|
Bernier LP, Bohlen CJ, York EM, Choi HB, Kamyabi A, Dissing-Olesen L, Hefendehl JK, Collins HY, Stevens B, Barres BA, MacVicar BA. Nanoscale Surveillance of the Brain by Microglia via cAMP-Regulated Filopodia. Cell Rep 2020; 27:2895-2908.e4. [PMID: 31167136 DOI: 10.1016/j.celrep.2019.05.010] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/14/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.
Collapse
Affiliation(s)
- Louis-Philippe Bernier
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| | - Christopher J Bohlen
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Elisa M York
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hyun B Choi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Alireza Kamyabi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Lasse Dissing-Olesen
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Jasmin K Hefendehl
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hannah Y Collins
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Ben A Barres
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Brian A MacVicar
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
57
|
Zhang N, Zhang Z, He R, Li H, Ding S. GLAST-CreER T2 mediated deletion of GDNF increases brain damage and exacerbates long-term stroke outcomes after focal ischemic stroke in mouse model. Glia 2020; 68:2395-2414. [PMID: 32497340 DOI: 10.1002/glia.23848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human death. Glial scar formation largely caused by reactive astrogliosis in peri-infarct region (PIR) is the hallmark of FIS. Glial cell-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line supernatant and is a potent survival neurotrophic factor. Here, using CreERT2 -LoxP recombination technology, we generated inducible and astrocyte-specific GDNF conditional knockout (cKO), that is, GLAST-GDNF-/- cKO mice to investigate the effect of reactive astrocytes (RAs)-derived GDNF on neuronal death, brain damage, oxidative stress and motor function recovery after photothrombosis (PT)-induced FIS. Under non-ischemic conditions, we found that adult GLAST-GDNF-/- cKO mice exhibited significant lower numbers of Brdu+, Ki67+ cells, and DCX+ cells in the dentate gyrus (DG) in hippocampus than GDNF floxed (GDNFf/f ) control (Ctrl) mice, indicating endogenous astrocytic GDNF can promote adult neurogenesis. Under ischemic conditions, GLAST-GDNF-/- cKO mice had a significant increase in infarct volume, hippocampal damage and FJB+ degenerating neurons after PT as compared with the Ctrl mice. GLAST-GDNF-/- cKO mice also had lower densities of Brdu+ and Ki67+ cells in the PIR and exhibited larger behavioral deficits than the Ctrl mice. Mechanistically, GDNF deficiency in astrocytes increased oxidative stress through the downregulation of glucose-6-phosphate dehydrogenase (G6PD) in RAs. In summary, our study indicates that RAs-derived endogenous GDNF plays important roles in reducing brain damage and promoting brain recovery after FIS through neural regeneration and suggests that promoting anti-oxidant mechanism in RAs is a potential strategy in stroke therapy.
Collapse
Affiliation(s)
- Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Zhe Zhang
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Rui He
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Hailong Li
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
58
|
Hu C, Lakshmipathi J, Stuart D, Peti-Peterdi J, Gyarmati G, Hao CM, Hansell P, Kohan DE. Renomedullary Interstitial Cell Endothelin A Receptors Regulate BP and Renal Function. J Am Soc Nephrol 2020; 31:1555-1568. [PMID: 32487560 DOI: 10.1681/asn.2020020232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/06/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The physiologic role of renomedullary interstitial cells, which are uniquely and abundantly found in the renal inner medulla, is largely unknown. Endothelin A receptors regulate multiple aspects of renomedullary interstitial cell function in vitro. METHODS To assess the effect of targeting renomedullary interstitial cell endothelin A receptors in vivo, we generated a mouse knockout model with inducible disruption of renomedullary interstitial cell endothelin A receptors at 3 months of age. RESULTS BP and renal function were similar between endothelin A receptor knockout and control mice during normal and reduced sodium or water intake. In contrast, on a high-salt diet, compared with control mice, the knockout mice had reduced BP; increased urinary sodium, potassium, water, and endothelin-1 excretion; increased urinary nitrite/nitrate excretion associated with increased noncollecting duct nitric oxide synthase-1 expression; increased PGE2 excretion associated with increased collecting duct cyclooxygenase-1 expression; and reduced inner medullary epithelial sodium channel expression. Water-loaded endothelin A receptor knockout mice, compared with control mice, had markedly enhanced urine volume and reduced urine osmolality associated with increased urinary endothelin-1 and PGE2 excretion, increased cyclooxygenase-2 protein expression, and decreased inner medullary aquaporin-2 protein content. No evidence of endothelin-1-induced renomedullary interstitial cell contraction was observed. CONCLUSIONS Disruption of renomedullary interstitial cell endothelin A receptors reduces BP and increases salt and water excretion associated with enhanced production of intrinsic renal natriuretic and diuretic factors. These studies indicate that renomedullary interstitial cells can modulate BP and renal function under physiologic conditions.
Collapse
Affiliation(s)
- Chunyan Hu
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | | | - Deborah Stuart
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | - Janos Peti-Peterdi
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Georgina Gyarmati
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter Hansell
- Department of Medical Cell Biology, Section of Integrative Physiology, Uppsala University Biomedical Center, Uppsala, Sweden
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| |
Collapse
|
59
|
Ghilardi SJ, O'Reilly BM, Sgro AE. Intracellular signaling dynamics and their role in coordinating tissue repair. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1479. [PMID: 32035001 PMCID: PMC7187325 DOI: 10.1002/wsbm.1479] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/20/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022]
Abstract
Tissue repair is a complex process that requires effective communication and coordination between cells across multiple tissues and organ systems. Two of the initial intracellular signals that encode injury signals and initiate tissue repair responses are calcium and extracellular signal-regulated kinase (ERK). However, calcium and ERK signaling control a variety of cellular behaviors important for injury repair including cellular motility, contractility, and proliferation, as well as the activity of several different transcription factors, making it challenging to relate specific injury signals to their respective repair programs. This knowledge gap ultimately hinders the development of new wound healing therapies that could take advantage of native cellular signaling programs to more effectively repair tissue damage. The objective of this review is to highlight the roles of calcium and ERK signaling dynamics as mechanisms that link specific injury signals to specific cellular repair programs during epithelial and stromal injury repair. We detail how the signaling networks controlling calcium and ERK can now also be dissected using classical signal processing techniques with the advent of new biosensors and optogenetic signal controllers. Finally, we advocate the importance of recognizing calcium and ERK dynamics as key links between injury detection and injury repair programs that both organize and execute a coordinated tissue repair response between cells across different tissues and organs. This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Biological Mechanisms > Cell Signaling Laboratory Methods and Technologies > Imaging Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models.
Collapse
Affiliation(s)
- Samuel J. Ghilardi
- Department of Biomedical Engineering and the Biological Design CenterBoston UniversityBostonMassachusetts
| | - Breanna M. O'Reilly
- Department of Biomedical Engineering and the Biological Design CenterBoston UniversityBostonMassachusetts
| | - Allyson E. Sgro
- Department of Biomedical Engineering and the Biological Design CenterBoston UniversityBostonMassachusetts
| |
Collapse
|
60
|
Zhou T, Gao B, Fan Y, Liu Y, Feng S, Cong Q, Zhang X, Zhou Y, Yadav PS, Lin J, Wu N, Zhao L, Huang D, Zhou S, Su P, Yang Y. Piezo1/2 mediate mechanotransduction essential for bone formation through concerted activation of NFAT-YAP1-ß-catenin. eLife 2020; 9:52779. [PMID: 32186512 PMCID: PMC7112954 DOI: 10.7554/elife.52779] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/17/2020] [Indexed: 12/15/2022] Open
Abstract
Mechanical forces are fundamental regulators of cell behaviors. However, molecular regulation of mechanotransduction remain poorly understood. Here, we identified the mechanosensitive channels Piezo1 and Piezo2 as key force sensors required for bone development and osteoblast differentiation. Loss of Piezo1, or more severely Piezo1/2, in mesenchymal or osteoblast progenitor cells, led to multiple spontaneous bone fractures in newborn mice due to inhibition of osteoblast differentiation and increased bone resorption. In addition, loss of Piezo1/2 rendered resistant to further bone loss caused by unloading in both bone development and homeostasis. Mechanistically, Piezo1/2 relayed fluid shear stress and extracellular matrix stiffness signals to activate Ca2+ influx to stimulate Calcineurin, which promotes concerted activation of NFATc1, YAP1 and ß-catenin transcription factors by inducing their dephosphorylation as well as NFAT/YAP1/ß-catenin complex formation. Yap1 and ß-catenin activities were reduced in the Piezo1 and Piezo1/2 mutant bones and such defects were partially rescued by enhanced ß-catenin activities.
Collapse
Affiliation(s)
- Taifeng Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Bo Gao
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Fan
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Xiaolei Zhang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yaxing Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Prem S Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Jiachen Lin
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopedic Surgery and Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Wu
- Department of Orthopedic Surgery and Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Zhao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Dongsheng Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, United States
| | - Peiqiang Su
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| |
Collapse
|
61
|
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
|
62
|
Eosinophil-platelet interactions promote atherosclerosis and stabilize thrombosis with eosinophil extracellular traps. Blood 2020; 134:1859-1872. [PMID: 31481482 DOI: 10.1182/blood.2019000518] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
Clinical observations implicate a role of eosinophils in cardiovascular diseases because markers of eosinophil activation are elevated in atherosclerosis and thrombosis. However, their contribution to atherosclerotic plaque formation and arterial thrombosis remains unclear. In these settings, we investigated how eosinophils are recruited and activated through an interplay with platelets. Here, we provide evidence for a central importance of eosinophil-platelet interactions in atherosclerosis and thrombosis. We show that eosinophils support atherosclerotic plaque formation involving enhanced von Willebrand factor exposure on endothelial cells and augmented platelet adhesion. During arterial thrombosis, eosinophils are quickly recruited in an integrin-dependent manner and engage in interactions with platelets leading to eosinophil activation as we show by intravital calcium imaging. These direct interactions induce the formation of eosinophil extracellular traps (EETs), which are present in human thrombi and constitute a substantial part of extracellular traps in murine thrombi. EETs are decorated with the granule protein major basic protein, which causes platelet activation by eosinophils. Consequently, targeting of EETs diminished thrombus formation in vivo, which identifies this approach as a novel antithrombotic concept. Finally, in our clinical analysis of coronary artery thrombi, we identified female patients with stent thrombosis as the population that might derive the greatest benefit from an eosinophil-inhibiting strategy. In summary, eosinophils contribute to atherosclerotic plaque formation and thrombosis through an interplay with platelets, resulting in mutual activation. Therefore, eosinophils are a promising new target in the prevention and therapy of atherosclerosis and thrombosis.
Collapse
|
63
|
Zhu Y, Li S, Sun Y, Wu J, Xiong Z, Jin T, Jia H, Yang X. IL1R1 Polymorphisms are Associated with Lumbar Disc Herniation Risk in the Northwestern Chinese Han Population. Med Sci Monit 2019; 25:3728-3738. [PMID: 31104063 PMCID: PMC6540620 DOI: 10.12659/msm.913563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background The aim of this study was to assess the association of single-nucleotide polymorphisms (SNPs) in IL1R1 with the risk of lumbar disc herniation (LDH) in the Han population in northwest China. Material/Methods To estimate the association of IL1R1 polymorphisms with LDH risk, Agena MassARRAY was used to determine the genotypes of 498 LDH patients and 463 controls. The association between IL1R1 variants and LDH risk was examined by logistic regression analysis with adjustments for age and gender. Stratification analysis was observed between gender and age with polymorphisms of IL1R1. Haplotype construction and analysis in IL1R1 were also applied to detect the potential association. Results The mutant homozygous genotype in codominant model (AA versus GG, OR=2.37, 95% CI: 1.08–5.21, P=0.001) and in recessive model (AA versus GG/GA, OR=2.82, 95% CI: 1.30–6.12, P=0.005) of rs956730 were associated with an increased LDH risk in males, while rs956730 heterozygous genotype under codominant model (AG versus GG, OR=0.65, 95% CI: 0.46–0.92, P=0.001) was a protective genotype in males. In addition, the recessive model (CT/CC versus TT, OR=3.43, 95% CI: 1.11–10.57, P=0.020) of rs10490571 was associated with an increased LDH risk among people older than 50 years of age. Conclusions This study demonstrated that genetic variants in the IL1R1 genes were associated with LDH risk in the Han population of northwestern China.
Collapse
Affiliation(s)
- Yong Zhu
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Shunan Li
- The Hohhot First Hospital, Hohhot, Inner Mongolia, China (mainland)
| | - Yao Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, Shaanxi, China (mainland)
| | - Jiamin Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, Shaanxi, China (mainland)
| | - Zichao Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, Shaanxi, China (mainland)
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, Shaanxi, China (mainland)
| | - Haiyu Jia
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Xuejun Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| |
Collapse
|
64
|
Holman HA, Poppi LA, Frerck M, Rabbitt RD. Spontaneous and Acetylcholine Evoked Calcium Transients in the Developing Mouse Utricle. Front Cell Neurosci 2019; 13:186. [PMID: 31133810 PMCID: PMC6514437 DOI: 10.3389/fncel.2019.00186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Spontaneous calcium transients are present during early postnatal development in the mouse retina and cochlea, and play an important role in maturation of the sensory organs and neural circuits in the central nervous system (CNS). It is not known whether similar calcium transients occur during postnatal development in the vestibular sensory organs. Here we demonstrate spontaneous intracellular calcium transients in sensory hair cells (HCs) and supporting cells (SCs) in the murine utricular macula during the first two postnatal weeks. Calcium transients were monitored using a genetically encoded calcium indicator, GCaMP5G (G5), at 100 ms-frame−1 in excised utricle sensory epithelia, including HCs, SCs, and neurons. The reporter line expressed G5 and tdTomato (tdT) in a Gad2-Cre dependent manner within a subset of utricular HCs, SCs and neurons. Kinetics of the G5 reporter limited temporal resolution to calcium events lasting longer than 200 ms. Spontaneous calcium transients lasting 1-2 s were observed in the expressing population of HCs at birth and slower spontaneous transients lasting 10-30 s appeared in SCs by P3. Beginning at P5, calcium transients could be modulated by application of the efferent neurotransmitter acetylcholine (ACh). In mature mice, calcium transients in the utricular macula occurred spontaneously, had a duration 1-2 s, and could be modulated by the exogenous application of acetylcholine (ACh) or muscarine. Long-lasting calcium transients evoked by ACh in mature mice were blocked by atropine, consistent with previous reports describing the role of muscarinic receptors expressed in calyx bearing afferents in efferent control of vestibular sensation. Large spontaneous and ACh evoked transients were reversibly blocked by the inositol trisphosphate receptor (IP3R) antagonist aminoethoxydiphenyl borate (2-APB). Results demonstrate long-lasting calcium transients are present in the utricular macula during the first postnatal week, and that responses to ACh mature over this same time period.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Lauren A Poppi
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,School of Biomedical Science and Pharmacy, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Micah Frerck
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,Neuroscience Program, University of Utah, Salt Lake City, UT, United States.,Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
65
|
Okubo Y, Iino M. Visualization of astrocytic intracellular Ca 2+ mobilization. J Physiol 2019; 598:1671-1681. [PMID: 30825213 DOI: 10.1113/jp277609] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/06/2019] [Indexed: 11/08/2022] Open
Abstract
Astrocytes generate robust intracellular Ca2+ concentration changes (Ca2+ signals), which are assumed to regulate astrocytic functions that play crucial roles in the regulation of brain functions. One frequently used strategy for exploring the role of astrocytic Ca2+ signalling is the use of mice deficient in the type 2 inositol 1,4,5-trisphosphate receptor (IP3 R2). These IP3 R2-knockout (KO) mice are reportedly devoid of Ca2+ mobilization from the endoplasmic reticulum (ER) in astrocytes. However, they have shown no functional deficits in several studies, causing a heated debate as to the functional relevance of ER-mediated Ca2+ signalling in astrocytes. Recently, the assumption that Ca2+ mobilization from the ER is absent in IP3 R2-KO astrocytes has been re-evaluated using intraorganellar Ca2+ imaging techniques. The new results indicated that IP3 R2-independent Ca2+ release may generate Ca2+ nanodomains around the ER, which may help explain the absence of functional deficits in IP3 R2-KO mice.
Collapse
Affiliation(s)
- Yohei Okubo
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, 133-0033, Japan
| | - Masamitsu Iino
- Division of Cellular and Molecular Pharmacology, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| |
Collapse
|
66
|
Repetitive Diffuse Mild Traumatic Brain Injury Causes an Atypical Astrocyte Response and Spontaneous Recurrent Seizures. J Neurosci 2019; 39:1944-1963. [PMID: 30665946 DOI: 10.1523/jneurosci.1067-18.2018] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 11/21/2022] Open
Abstract
Focal traumatic brain injury (TBI) induces astrogliosis, a process essential to protecting uninjured brain areas from secondary damage. However, astrogliosis can cause loss of astrocyte homeostatic functions and possibly contributes to comorbidities such as posttraumatic epilepsy (PTE). Scar-forming astrocytes seal focal injuries off from healthy brain tissue. It is these glial scars that are associated with epilepsy originating in the cerebral cortex and hippocampus. However, the vast majority of human TBIs also present with diffuse brain injury caused by acceleration-deceleration forces leading to tissue shearing. The resulting diffuse tissue damage may be intrinsically different from focal lesions that would trigger glial scar formation. Here, we used mice of both sexes in a model of repetitive mild/concussive closed-head TBI, which only induced diffuse injury, to test the hypothesis that astrocytes respond uniquely to diffuse TBI and that diffuse TBI is sufficient to cause PTE. Astrocytes did not form scars and classic astrogliosis characterized by upregulation of glial fibrillary acidic protein was limited. Surprisingly, an unrelated population of atypical reactive astrocytes was characterized by the lack of glial fibrillary acidic protein expression, rapid and sustained downregulation of homeostatic proteins and impaired astrocyte coupling. After a latency period, a subset of mice developed spontaneous recurrent seizures reminiscent of PTE in human TBI patients. Seizing mice had larger areas of atypical astrocytes compared with nonseizing mice, suggesting that these atypical astrocytes might contribute to epileptogenesis after diffuse TBI.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a leading cause of acquired epilepsies. Reactive astrocytes have long been associated with seizures and epilepsy in patients, particularly after focal/lesional brain injury. However, most TBIs also include nonfocal, diffuse injuries. Here, we showed that repetitive diffuse TBI is sufficient for the development of spontaneous recurrent seizures in a subset of mice. We identified an atypical response of astrocytes induced by diffuse TBI characterized by the rapid loss of homeostatic proteins and lack of astrocyte coupling while reactive astrocyte markers or glial scar formation was absent. Areas with atypical astrocytes were larger in animals that later developed seizures suggesting that this response may be one root cause of epileptogenesis after diffuse TBI.
Collapse
|
67
|
Schiessl IM, Fremter K, Burford JL, Castrop H, Peti-Peterdi J. Long-Term Cell Fate Tracking of Individual Renal Cells Using Serial Intravital Microscopy. Methods Mol Biol 2019; 2150:25-44. [PMID: 31087287 DOI: 10.1007/7651_2019_232] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intravital multiphoton microscopy of the kidney is a powerful technique to study alterations in tissue morphology and function simultaneously in the living animal and represents a dynamic and developing research tool in the field. Recent technological advances include serial intravital multiphoton microscopy of the same kidney regions over several weeks and combined with ex vivo histology for cellular biomarker expression of the same cells, which had been subject to serial imaging before. Thus, serial intravital multiphoton microscopy followed by ex vivo histology provides unique tools to perform long-term cell fate tracing of the same renal cells during physiological and pathophysiological conditions, thereby allowing the detection of structural changes of the same renal cells over time. Examples include renal cell migration and proliferation while linking these events to local functional alterations and eventually to the expression of distinct cellular biomarkers. Here, we provide a detailed step-by-step protocol to facilitate serial intravital multiphoton microscopy for long-term in vivo tracking of renal cells and subsequent ex vivo histology for immunohistological staining of the same cells in the fixed tissue.
Collapse
Affiliation(s)
- Ina Maria Schiessl
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - James L Burford
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, USA
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
68
|
|
69
|
Abstract
Astrocytes are glial cells carrying out complex homeostatic functions in the healthy and diseased central nervous system (CNS). It has so far been impossible to reliably culture adult astrocytes and the results of studies on astrocytes outside of their normal environment are challenging to interpret. Consequently, most culture studies use astrocytes isolated from postnatal rodents. Yet cultured astrocytes do not display their complex three-dimensional in vivo morphology, and transcriptomes of cultured astrocytes vary significantly from those of acutely isolated astrocytes (Cahoy et al., J Neurosci 28:264-278, 2008). Astrocyte isolation for culture experiments, and the cutting of acute brain slices, induces astrocyte reactivity similar to a severe acute injury. In response to CNS injury, such as moderate or severe focal traumatic brain injury (TBI), astrocytes can change in cell number, physiological state, gene and protein expression, secretome, and morphology, in a process termed reactive astrogliosis. This makes the use of methods that inherently induce astrogliosis (e.g., dissociation of brain tissue for culture or sectioning of brains for acute brain slices) challenging, especially when conditions are studied that present with changes in astrocyte function that are milder and/or of a different nature.In this methods chapter, we will describe a technical approach that allows one to study astrocytes in the intact brain using two-photon in vivo imaging. We will use mild TBI as an example of how to use this approach to compare astrocyte function in the same animal before and after an injury.Here we describe the use of a noninvasive label-free method (Choi et al., J Biomed Opt 16:075003, 2011) to increase astrocyte Ca2+ using optical femtosecond pulsed laser activation. We will provide systematic instruction of the surgical technique, which when done properly, allows in vivo astrocyte imaging in the same experimental animal before the injury as well as over the course of days, weeks, and even months after injury. We will also elaborate on challenges in astrocytic Ca2+ imaging and how different image acquisition settings can affect the readout of astrocyte Ca2+ oscillations.
Collapse
|
70
|
Tvrdik P, Kearns KN, Sharifi KA, Sluzewski MF, Acton ST, Kalani MYS. Calcium Imaging of Microglial Network Activity in Stroke. Methods Mol Biol 2019; 2034:267-279. [PMID: 31392691 DOI: 10.1007/978-1-4939-9658-2_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Calcium signaling plays a significant role in microglial activation. Genetically encoded calcium indicators (GECI) have been widely used for calcium imaging studies in many brain cell types, including neurons, astrocytes, and oligodendrocytes. However, microglial calcium imaging approaches have been hampered by idiosyncrasies of their gene expression and malleable cell properties. The generation of PC::G5-tdT, a Polr2a locus-based conditional mouse reporter of calcium, facilitated the deployment of GECI in microglia. When crossed with the Iba1(Aif1)-IRES-Cre line, all brain microglia of the progeny are labeled with the calcium indicator variant GCaMP5G and the red fluorescent protein tdTomato. This reporter system has enabled in vivo studies of intracellular calcium in large microglial cell populations in cerebral pathologies such as ischemic stroke. In this chapter, we outline specific guidelines for genetic, surgical, imaging, and data analysis aspects of microglial calcium monitoring of the ischemic cortex following middle cerebral artery occlusion.
Collapse
Affiliation(s)
- Petr Tvrdik
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Kathryn N Kearns
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Khadijeh A Sharifi
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - M Filip Sluzewski
- Department of Electrical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Scott T Acton
- Department of Electrical Engineering, University of Virginia, Charlottesville, VA, USA
| | - M Yashar S Kalani
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
71
|
Abstract
Genetically encoded calcium indicators (GECIs) have become widely used for Ca2+ imaging in cultured cells as well as in living organisms. Transduction of microglia with viral vectors encoding GECIs provides a convenient means to label microglia for in vivo Ca2+ imaging. We describe a method using microglia-specific microRNA-9-regulated viral vector, to label microglial cells with a ratiometric GECI (Twitch-2B). This method enables longitudinal recording of both transient and sustained elevations of Ca2+ in microglia in live animals.
Collapse
Affiliation(s)
- Yajie Liang
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
72
|
Conditional Knock-out of mGluR5 from Astrocytes during Epilepsy Development Impairs High-Frequency Glutamate Uptake. J Neurosci 2018; 39:727-742. [PMID: 30504280 DOI: 10.1523/jneurosci.1148-18.2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 11/11/2018] [Accepted: 11/18/2018] [Indexed: 11/21/2022] Open
Abstract
Astrocyte expression of metabotropic glutamate receptor 5 (mGluR5) is consistently observed in resected tissue from patients with epilepsy and is equally prevalent in animal models of epilepsy. However, little is known about the functional signaling properties or downstream consequences of astrocyte mGluR5 activation during epilepsy development. In the rodent brain, astrocyte mGluR5 expression is developmentally regulated and confined in expression/function to the first weeks of life, with similar observations made in human control tissue. Herein, we demonstrate that mGluR5 expression and function dramatically increase in a mouse model of temporal lobe epilepsy. Interestingly, in both male and female mice, mGluR5 function persists in the astrocyte throughout the process of epileptogenesis following status epilepticus. However, mGluR5 expression and function are transient in animals that do not develop epilepsy over an equivalent time period, suggesting that patterns of mGluR5 expression may signify continuing epilepsy development or its resolution. We demonstrate that, during epileptogenesis, astrocytes reacquire mGluR5-dependent calcium transients following agonist application or synaptic glutamate release, a feature of astrocyte-neuron communication absent since early development. Finally, we find that the selective and conditional knock-out of mGluR5 signaling from astrocytes during epilepsy development slows the rate of glutamate clearance through astrocyte glutamate transporters under high-frequency stimulation conditions, a feature that suggests astrocyte mGluR5 expression during epileptogenesis may recapitulate earlier developmental roles in regulating glutamate transporter function.SIGNIFICANCE STATEMENT In development, astrocyte mGluR5 signaling plays a critical role in regulating structural and functional interactions between astrocytes and neurons at the tripartite synapse. Notably, mGluR5 signaling is a positive regulator of astrocyte glutamate transporter expression and function, an essential component of excitatory signaling regulation in hippocampus. After early development, astrocyte mGluR5 expression is downregulated, but reemerges in animal models of temporal lobe epilepsy (TLE) development and patient epilepsy samples. We explored the hypothesis that astrocyte mGluR5 reemergence recapitulates earlier developmental roles during TLE acquisition. Our work demonstrates that astrocytes with mGluR5 signaling during TLE development perform faster glutamate uptake in hippocampus, revealing a previously unexplored role for astrocyte mGluR5 signaling in hypersynchronous pathology.
Collapse
|
73
|
p53 in AgRP neurons is required for protection against diet-induced obesity via JNK1. Nat Commun 2018; 9:3432. [PMID: 30143607 PMCID: PMC6109113 DOI: 10.1038/s41467-018-05711-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity. Emerging studies suggest that p53 is an important regulator of energy metabolism, yet there is little known about the metabolic function of this tumor suppressor in the hypothalamus. Here, authors illustrate that p53, specifically in AgRP neurons, is required for adaptation to diet-induced obesity.
Collapse
|
74
|
Stobart JL, Ferrari KD, Barrett MJP, Stobart MJ, Looser ZJ, Saab AS, Weber B. Long-term In Vivo Calcium Imaging of Astrocytes Reveals Distinct Cellular Compartment Responses to Sensory Stimulation. Cereb Cortex 2018; 28:184-198. [PMID: 28968832 DOI: 10.1093/cercor/bhw366] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/02/2016] [Indexed: 01/28/2023] Open
Abstract
Localized, heterogeneous calcium transients occur throughout astrocytes, but the characteristics and long-term stability of these signals, particularly in response to sensory stimulation, remain unknown. Here, we used a genetically encoded calcium indicator and an activity-based image analysis scheme to monitor astrocyte calcium activity in vivo. We found that different subcellular compartments (processes, somata, and endfeet) displayed distinct signaling characteristics. Closer examination of individual signals showed that sensory stimulation elevated the number of specific types of calcium peaks within astrocyte processes and somata, in a cortical layer-dependent manner, and that the signals became more synchronous upon sensory stimulation. Although mice genetically lacking astrocytic IP3R-dependent calcium signaling (Ip3r2-/-) had fewer signal peaks, the response to sensory stimulation was sustained, suggesting other calcium pathways are also involved. Long-term imaging of astrocyte populations revealed that all compartments reliably responded to stimulation over several months, but that the location of the response within processes may vary. These previously unknown characteristics of subcellular astrocyte calcium signals provide new insights into how astrocytes may encode local neuronal circuit activity.
Collapse
Affiliation(s)
- Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Matthew J P Barrett
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Michael J Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
75
|
Hierro-Bujalance C, Bacskai BJ, Garcia-Alloza M. In Vivo Imaging of Microglia With Multiphoton Microscopy. Front Aging Neurosci 2018; 10:218. [PMID: 30072888 PMCID: PMC6060250 DOI: 10.3389/fnagi.2018.00218] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023] Open
Abstract
Neuroimaging has become an unparalleled tool to understand the central nervous system (CNS) anatomy, physiology and neurological diseases. While an altered immune function and microglia hyperactivation are common neuropathological features for many CNS disorders and neurodegenerative diseases, direct assessment of the role of microglial cells remains a challenging task. Non-invasive neuroimaging techniques, including magnetic resonance imaging (MRI), positron emission tomography (PET) and single positron emission computed tomography (SPECT) are widely used for human clinical applications, and a variety of ligands are available to detect neuroinflammation. In animal models, intravital imaging has been largely used, and minimally invasive multiphoton microcopy (MPM) provides high resolution detection of single microglia cells, longitudinally, in living brain. In this study, we review in vivo real-time MPM approaches to assess microglia in preclinical studies, including individual cell responses in surveillance, support, protection and restoration of brain tissue integrity, synapse formation, homeostasis, as well as in different pathological situations. We focus on in vivo studies that assess the role of microglia in mouse models of Alzheimer’s disease (AD), analyzing microglial motility and recruitment, as well as the role of microglia in anti-amyloid-β treatment, as a key therapeutic approach to treat AD. Altogether, MPM provides a high contrast and high spatial resolution approach to follow microglia chronically in vivo in complex models, supporting MPM as a powerful tool for deep intravital tissue imaging.
Collapse
Affiliation(s)
- Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Instituto de Investigación e Innovación en Ciencias Biomedicas de la Provincia de Cadiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Brian J Bacskai
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Instituto de Investigación e Innovación en Ciencias Biomedicas de la Provincia de Cadiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| |
Collapse
|
76
|
Reinhart KM, Shuttleworth CW. Ketamine reduces deleterious consequences of spreading depolarizations. Exp Neurol 2018; 305:121-128. [PMID: 29653188 PMCID: PMC6261532 DOI: 10.1016/j.expneurol.2018.04.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/31/2018] [Accepted: 04/08/2018] [Indexed: 01/12/2023]
Abstract
Recent work has implicated spreading depolarization (SD) as a key contributor the progression of acute brain injuries, however development of interventions selectively targeting SD has lagged behind. Initial clinical intervention efforts have focused on observations that relatively high doses of the sedative agent ketamine can completely suppress SD. However, blocking propagation of SD could theoretically prevent beneficial effects of SD in surrounding brain regions. Selective targeting of deleterious consequences of SD (rather than abolition) could be a useful adjunct approach, and be achieved with lower ketamine concentrations. We utilized a brain slice model to test whether deleterious consequences of SD could be prevented by ketamine, using concentrations that did not prevent the initiation and propagation of SD. Studies were conducted using murine brain slices, with focal KCl as an SD stimulus. Consequences of SD were assessed with electrophysiological and imaging measures of ionic and synaptic recovery. Under control conditions, ketamine (up to 30 μM) did not prevent SD, but significantly reduced neuronal Ca2+ loading and the duration of associated extracellular potential shifts. Recovery of postsynaptic potentials after SD was also significantly accelerated. When SD was evoked on a background of mild metabolic compromise, neuronal recovery was substantially impaired. Under compromised conditions, the same concentrations of ketamine reduced ionic and metabolic loading during SD, sufficient to preserve functional recovery after repetitive SDs. These results suggest that lower concentrations of ketamine could be utilized to prevent damaging consequences of SD, while not blocking them outright and thereby preserving potentially protective effects of SD.
Collapse
Affiliation(s)
- Katelyn M Reinhart
- Department of Neurosciences, University of New Mexico School of Medicine, United States
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, United States.
| |
Collapse
|
77
|
Hansen KR, DeWalt GJ, Mohammed AI, Tseng HA, Abdulkerim ME, Bensussen S, Saligrama V, Nazer B, Eldred WD, Han X. Mild Blast Injury Produces Acute Changes in Basal Intracellular Calcium Levels and Activity Patterns in Mouse Hippocampal Neurons. J Neurotrauma 2018; 35:1523-1536. [PMID: 29343209 PMCID: PMC5998839 DOI: 10.1089/neu.2017.5029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mild traumatic brain injury (mTBI) represents a serious public health concern. Although much is understood about long-term changes in cell signaling and anatomical pathologies associated with mTBI, little is known about acute changes in neuronal function. Using large scale Ca2+ imaging in vivo, we characterized the intracellular Ca2+ dynamics in thousands of individual hippocampal neurons using a repetitive mild blast injury model in which blasts were directed onto the cranium of unanesthetized mice on two consecutive days. Immediately following each blast event, neurons exhibited two types of changes in Ca2+ dynamics at different time scales. One was a reduction in slow Ca2+ dynamics that corresponded to shifts in basal intracellular Ca2+ levels at a time scale of minutes, suggesting a disruption of biochemical signaling. The second was a reduction in the rates of fast transient Ca2+ fluctuations at the sub-second time scale, which are known to be closely linked to neural activity. Interestingly, the blast-induced changes in basal Ca2+ levels were independent of the changes in the rates of fast Ca2+ transients, suggesting that blasts had heterogeneous effects on different cell populations. Both types of changes recovered after ∼1 h. Together, our results demonstrate that mTBI induced acute, heterogeneous changes in neuronal function, altering intracellular Ca2+ dynamics across different time scales, which may contribute to the initiation of longer-term pathologies.
Collapse
Affiliation(s)
- Kyle R. Hansen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Ali I. Mohammed
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Hua-an Tseng
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Moona E. Abdulkerim
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Seth Bensussen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Venkatesh Saligrama
- Department of Electrical and Computer Engineering, Boston University, Boston, Massachusetts
| | - Bobak Nazer
- Department of Electrical and Computer Engineering, Boston University, Boston, Massachusetts
| | | | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| |
Collapse
|
78
|
Delvalle NM, Dharshika C, Morales-Soto W, Fried DE, Gaudette L, Gulbransen BD. Communication Between Enteric Neurons, Glia, and Nociceptors Underlies the Effects of Tachykinins on Neuroinflammation. Cell Mol Gastroenterol Hepatol 2018; 6:321-344. [PMID: 30116771 PMCID: PMC6091443 DOI: 10.1016/j.jcmgh.2018.05.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/18/2018] [Indexed: 12/18/2022]
Abstract
Background & Aims Tachykinins are involved in physiological and pathophysiological mechanisms in the gastrointestinal tract. The major sources of tachykinins in the gut are intrinsic enteric neurons in the enteric nervous system and extrinsic nerve fibers from the dorsal root and vagal ganglia. Although tachykinins are important mediators in the enteric nervous system, how they contribute to neuroinflammation through effects on neurons and glia is not fully understood. Here, we tested the hypothesis that tachykinins contribute to enteric neuroinflammation through mechanisms that involve intercellular neuron-glia signaling. Methods We used immunohistochemistry and quantitative real-time polymerase chain reaction, and studied cellular activity using transient-receptor potential vanilloid-1 (TRPV1)tm1(cre)Bbm/J::Polr2atm1(CAG-GCaMP5g,-tdTomato)Tvrd and Sox10CreERT2::Polr2atm1(CAG-GCaMP5g,-tdTomato)Tvrd mice or Fluo-4. We used the 2,4-di-nitrobenzene sulfonic acid (DNBS) model of colitis to study neuroinflammation, glial reactivity, and neurogenic contractility. We used Sox10::CreERT2+/-/Rpl22tm1.1Psam/J mice to selectively study glial transcriptional changes. Results Tachykinins are expressed predominantly by intrinsic neuronal varicosities whereas neurokinin-2 receptors (NK2Rs) are expressed predominantly by enteric neurons and TRPV1-positive neuronal varicosities. Stimulation of NK2Rs drives responses in neuronal varicosities that are propagated to enteric glia and neurons. Antagonizing NK2R signaling enhanced recovery from colitis and prevented the development of reactive gliosis, neuroinflammation, and enhanced neuronal contractions. Inflammation drove changes in enteric glial gene expression and function, and antagonizing NK2R signaling mitigated these changes. Neurokinin A-induced neurodegeneration requires glial connexin-43 hemichannel activity. Conclusions Our results show that tachykinins drive enteric neuroinflammation through a multicellular cascade involving enteric neurons, TRPV1-positive neuronal varicosities, and enteric glia. Therapies targeting components of this pathway could broadly benefit the treatment of dysmotility and pain after acute inflammation in the intestine.
Collapse
Key Words
- BzATP, 2’(3’)-O-(4-benzoylbenzoyl)adenosine 5’-triphosphate triethylammonium salt
- Ca2+, calcium
- Colitis
- Cx43, connexin-43
- DMEM, Dulbecco's modified Eagle medium
- DNBS, dinitrobenzene sulfonic acid
- EFS, electrical field stimulation
- ENS, enteric nervous system
- Enteric Nervous System
- FGID, functional gastrointestinal disorder
- GFAP, glial fibrillary acidic protein
- GI, gastrointestinal
- Glia
- HA, hemagglutinin
- IPAN, intrinsic primarily afferent neuron
- LMMP, longitudinal muscle–myenteric plexus
- MSU, Michigan State University
- NK1R, neurokinin-1 receptor
- NK2R, neurokinin-2 receptor
- NKA, neurokinin A
- Neurokinins
- SP, substance P
- TRPV1, transient receptor potential vanilloid-1
- mRNA, messenger RNA
Collapse
Affiliation(s)
| | - Christine Dharshika
- Genetics Program, Michigan State University, East Lansing, Michigan
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | | | - David E. Fried
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Lukas Gaudette
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Brian D. Gulbransen
- Neuroscience Program, Michigan State University, East Lansing, Michigan
- Department of Physiology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
79
|
Stobart JL, Ferrari KD, Barrett MJP, Glück C, Stobart MJ, Zuend M, Weber B. Cortical Circuit Activity Evokes Rapid Astrocyte Calcium Signals on a Similar Timescale to Neurons. Neuron 2018; 98:726-735.e4. [PMID: 29706581 DOI: 10.1016/j.neuron.2018.03.050] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/12/2018] [Accepted: 03/30/2018] [Indexed: 12/22/2022]
Abstract
Sensory stimulation evokes intracellular calcium signals in astrocytes; however, the timing of these signals is disputed. Here, we used novel combinations of genetically encoded calcium indicators for concurrent two-photon imaging of cortical astrocytes and neurons in awake mice during whisker deflection. We identified calcium responses in both astrocyte processes and endfeet that rapidly followed neuronal events (∼120 ms after). These fast astrocyte responses were largely independent of IP3R2-mediated signaling and known neuromodulator activity (acetylcholine, serotonin, and norepinephrine), suggesting that they are evoked by local synaptic activity. The existence of such rapid signals implies that astrocytes are fast enough to play a role in synaptic modulation and neurovascular coupling. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Matthew J P Barrett
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Michael J Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Marc Zuend
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center, University and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
80
|
Yang Y, Liu N, He Y, Liu Y, Ge L, Zou L, Song S, Xiong W, Liu X. Improved calcium sensor GCaMP-X overcomes the calcium channel perturbations induced by the calmodulin in GCaMP. Nat Commun 2018; 9:1504. [PMID: 29666364 PMCID: PMC5904127 DOI: 10.1038/s41467-018-03719-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 03/09/2018] [Indexed: 01/09/2023] Open
Abstract
GCaMP, one popular type of genetically-encoded Ca2+ indicator, has been associated with various side-effects. Here we unveil the intrinsic problem prevailing over different versions and applications, showing that GCaMP containing CaM (calmodulin) interferes with both gating and signaling of L-type calcium channels (CaV1). GCaMP acts as an impaired apoCaM and Ca2+/CaM, both critical to CaV1, which disrupts Ca2+ dynamics and gene expression. We then design and implement GCaMP-X, by incorporating an extra apoCaM-binding motif, effectively protecting CaV1-dependent excitation–transcription coupling from perturbations. GCaMP-X resolves the problems of detrimental nuclear accumulation, acute and chronic Ca2+ dysregulation, and aberrant transcription signaling and cell morphogenesis, while still demonstrating excellent Ca2+-sensing characteristics partly inherited from GCaMP. In summary, CaM/CaV1 gating and signaling mechanisms are elucidated for GCaMP side-effects, while allowing the development of GCaMP-X to appropriately monitor cytosolic, submembrane or nuclear Ca2+, which is also expected to guide the future design of CaM-based molecular tools. The popular genetically-encoded Ca2+ indicator, GCaMP, has several side-effects. Here the authors show that GCaMP containing CaM interferes with gating and signaling of L-type calcium channels, which disrupts Ca2+ dynamics and gene expression, and develop GCaMP-X to overcome these limitations.
Collapse
Affiliation(s)
- Yaxiong Yang
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China.,School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 102402, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Nan Liu
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Yunan University, Kunming, 650091, China
| | - Yuanyuan He
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China
| | - Yuxia Liu
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China
| | - Lin Ge
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China
| | - Linzhi Zou
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Sen Song
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Wei Xiong
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaodong Liu
- Department of Biomedical Engineering, School of Medicine, X-Lab for Transmembrane Signaling Research, Tsinghua University, Beijing, 100084, China. .,School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 102402, China. .,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
81
|
Schiessl IM, Grill A, Fremter K, Steppan D, Hellmuth MK, Castrop H. Renal Interstitial Platelet-Derived Growth Factor Receptor- β Cells Support Proximal Tubular Regeneration. J Am Soc Nephrol 2018; 29:1383-1396. [PMID: 29444905 DOI: 10.1681/asn.2017101069] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/16/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The kidney is considered to be a structurally stable organ with limited baseline cellular turnover. Nevertheless, single cells must be constantly replaced to conserve the functional integrity of the organ. PDGF chain B (PDGF-BB) signaling through fibroblast PDGF receptor-β (PDGFRβ) contributes to interstitial-epithelial cell communication and facilitates regenerative functions in several organs. However, the potential role of interstitial cells in renal tubular regeneration has not been examined. METHODS In mice with fluorescent protein expression in renal tubular cells and PDGFRβ-positive interstitial cells, we ablated single tubular cells by high laser exposure. We then used serial intravital multiphoton microscopy with subsequent three-dimensional reconstruction and ex vivo histology to evaluate the cellular and molecular processes involved in tubular regeneration. RESULTS Single-tubular cell ablation caused the migration and division of dedifferentiated tubular epithelial cells that preceded tubular regeneration. Moreover, tubular cell ablation caused immediate calcium responses in adjacent PDGFRβ-positive interstitial cells and the rapid migration thereof toward the injury. These PDGFRβ-positive cells enclosed the injured epithelium before the onset of tubular cell dedifferentiation, and the later withdrawal of these PDGFRβ-positive cells correlated with signs of tubular cell redifferentiation. Intraperitoneal administration of trapidil to block PDGFRβ impeded PDGFRβ-positive cell migration to the tubular injury site and compromised the recovery of tubular function. CONCLUSIONS Ablated tubular cells are exclusively replaced by resident tubular cell proliferation in a process dependent on PDGFRβ-mediated communication between the renal interstitium and the tubular system.
Collapse
Affiliation(s)
- Ina Maria Schiessl
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and .,Department of Physiology and Biophysics, University of Southern California, Los Angeles, California
| | - Alexandra Grill
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Dominik Steppan
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | | | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| |
Collapse
|
82
|
Saito K, Shigetomi E, Yasuda R, Sato R, Nakano M, Tashiro K, Tanaka KF, Ikenaka K, Mikoshiba K, Mizuta I, Yoshida T, Nakagawa M, Mizuno T, Koizumi S. Aberrant astrocyte Ca 2+ signals "AxCa signals" exacerbate pathological alterations in an Alexander disease model. Glia 2018; 66:1053-1067. [PMID: 29383757 DOI: 10.1002/glia.23300] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/12/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022]
Abstract
Alexander disease (AxD) is a rare neurodegenerative disorder caused by gain of function mutations in the glial fibrillary acidic protein (GFAP) gene. Accumulation of GFAP proteins and formation of Rosenthal fibers (RFs) in astrocytes are hallmarks of AxD. However, malfunction of astrocytes in the AxD brain is poorly understood. Here, we show aberrant Ca2+ responses in astrocytes as playing a causative role in AxD. Transcriptome analysis of astrocytes from a model of AxD showed age-dependent upregulation of GFAP, several markers for neurotoxic reactive astrocytes, and downregulation of Ca2+ homeostasis molecules. In situ AxD model astrocytes produced aberrant extra-large Ca2+ signals "AxCa signals", which increased with age, correlated with GFAP upregulation, and were dependent on stored Ca2+ . Inhibition of AxCa signals by deletion of inositol 1,4,5-trisphosphate type 2 receptors (IP3R2) ameliorated AxD pathogenesis. Taken together, AxCa signals in the model astrocytes would contribute to AxD pathogenesis.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan.,Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan
| | - Rei Yasuda
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryuichi Sato
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji F Tanaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Japan
| | - Ikuko Mizuta
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomokatsu Yoshida
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Nakagawa
- Department of Neurology, North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan
| |
Collapse
|
83
|
Role of Purinergic Receptor P2Y1 in Spatiotemporal Ca 2+ Dynamics in Astrocytes. J Neurosci 2018; 38:1383-1395. [PMID: 29305530 DOI: 10.1523/jneurosci.2625-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Accepted: 12/18/2017] [Indexed: 01/08/2023] Open
Abstract
Fine processes of astrocytes enwrap synapses and are well positioned to sense neuronal information via synaptic transmission. In rodents, astrocyte processes sense synaptic transmission via Gq-protein coupled receptors (GqPCR), including the P2Y1 receptor (P2Y1R), to generate Ca2+ signals. Astrocytes display numerous spontaneous microdomain Ca2+ signals; however, it is not clear whether such signals are due to local synaptic transmission and/or in what timeframe astrocytes sense local synaptic transmission. To ask whether GqPCRs mediate microdomain Ca2+ signals, we engineered mice (both sexes) to specifically overexpress P2Y1Rs in astrocytes, and we visualized Ca2+ signals via a genetically encoded Ca2+ indicator, GCaMP6f, in astrocytes from adult mice. Astrocytes overexpressing P2Y1Rs showed significantly larger Ca2+ signals in response to exogenously applied ligand and to repetitive electrical stimulation of axons compared with controls. However, we found no evidence of increased microdomain Ca2+ signals. Instead, Ca2+ waves appeared and propagated to occupy areas that were up to 80-fold larger than microdomain Ca2+ signals. These Ca2+ waves accounted for only 2% of total Ca2+ events, but they were 1.9-fold larger and 2.9-fold longer in duration than microdomain Ca2+ signals at processes. Ca2+ waves did not require action potentials for their generation and occurred in a probenecid-sensitive manner, indicating that the endogenous ligand for P2Y1R is elevated independently of synaptic transmission. Our data suggest that spontaneous microdomain Ca2+ signals occur independently of P2Y1R activation and that astrocytes may not encode neuronal information in response to synaptic transmission at a point source of neurotransmitter release.SIGNIFICANCE STATEMENT Astrocytes are thought to enwrap synapses with their processes to receive neuronal information via Gq-protein coupled receptors (GqPCRs). Astrocyte processes display numerous microdomain Ca2+ signals that occur spontaneously. To determine whether GqPCRs play a role in microdomain Ca2+ signals and the timeframe in which astrocytes sense neuronal information, we engineered mice whose astrocytes specifically overexpress the P2Y1 receptor, a major GqPCR in astrocytes. We found that overexpression of P2Y1 receptors in astrocytes did not increase microdomain Ca2+ signals in astrocyte processes but caused Ca2+ wavelike signals. Our data indicate that spontaneous microdomain Ca2+ signals do not require activation of P2Y1 receptors.
Collapse
|
84
|
Adams KL, Gallo V. The diversity and disparity of the glial scar. Nat Neurosci 2017; 21:9-15. [PMID: 29269757 DOI: 10.1038/s41593-017-0033-9] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 10/17/2017] [Indexed: 01/02/2023]
Abstract
Injury or disease to the CNS results in multifaceted cellular and molecular responses. One such response, the glial scar, is a structural formation of reactive glia around an area of severe tissue damage. While traditionally viewed as a barrier to axon regeneration, beneficial functions of the glial scar have also been recently identified. In this Perspective, we discuss the divergent roles of the glial scar during CNS regeneration and explore the possibility that these disparities are due to functional heterogeneity within the cells of the glial scar-specifically, astrocytes, NG2 glia and microglia.
Collapse
Affiliation(s)
- Katrina L Adams
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, USA.
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, USA.
| |
Collapse
|
85
|
Dong TX, Othy S, Jairaman A, Skupsky J, Zavala A, Parker I, Dynes JL, Cahalan MD. T-cell calcium dynamics visualized in a ratiometric tdTomato-GCaMP6f transgenic reporter mouse. eLife 2017; 6:32417. [PMID: 29239725 PMCID: PMC5747524 DOI: 10.7554/elife.32417] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Calcium is an essential cellular messenger that regulates numerous functions in living organisms. Here, we describe development and characterization of ‘Salsa6f’, a fusion of GCaMP6f and tdTomato optimized for cell tracking while monitoring cytosolic Ca2+, and a transgenic Ca2+ reporter mouse with Salsa6f targeted to the Rosa26 locus for Cre-dependent expression in specific cell types. The development and function of T cells was unaffected in Cd4-Salsa6f mice. We describe Ca2+ signals reported by Salsa6f during T cell receptor activation in naive T cells, helper Th17 T cells and regulatory T cells, and Ca2+ signals mediated in T cells by an activator of mechanosensitive Piezo1 channels. Transgenic expression of Salsa6f enables ratiometric imaging of Ca2+ signals in complex tissue environments found in vivo. Two-photon imaging of migrating T cells in the steady-state lymph node revealed both cell-wide and localized sub-cellular Ca2+ transients (‘sparkles’) as cells migrate. To help protect the body from disease, small immune cells called T lymphocytes move rapidly, searching for signs of infection. These signs are antigens – processed pieces of proteins from invading bacteria and viruses – which are displayed on the surface of so-called antigen-presenting cells. To visit as many different antigen-presenting cells as possible, T cells move quickly from one to the next in an apparently random manner. How T cells are programmed to move in this way is largely unknown. The entry of calcium ions into cells, through channel proteins, triggers characteristic actions in many cells throughout the body. As such it is possible that the T cells’ movements are related to calcium signals too. However, it was technically challenging to directly measure the amount of calcium in moving cells within the body. To overcome this issue, Dong, Othy et al. genetically engineered mice to produce a new calcium-sensitive reporter protein in their T cells. The reporter, which was named Salsa6f, consisted of a red fluorescent protein fused to another protein that glows green when it binds to calcium ions. Measuring the ratio of red and green fluorescence gives a measure of the concentration of calcium ions inside the cell. In the absence of calcium signaling, the cells can still be tracked via the red fluorescence of Salsa6f. Importantly, the reporter did not affect the development or activity of the T cells in the mice. In a related study, Dong, Othy et al. then used their transgenic mice to ask whether calcium signals guide moving T cells as they search for antigens. Future studies could use these transgenic mice to track the calcium ion concentration in numerous cell types. This would enable new approaches to relate the inner workings of cells to their behaviors in many different organ systems throughout the body.
Collapse
Affiliation(s)
- Tobias X Dong
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Amit Jairaman
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Jonathan Skupsky
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Department of Medicine, University of California, Irvine, United States
| | - Angel Zavala
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Department of Neurobiology & Behavior, University of California, Irvine, United States
| | - Joseph L Dynes
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Institute for Immunology, University of California, Irvine, United States
| |
Collapse
|
86
|
Tvrdik P, Kalani MYS. In Vivo Imaging of Microglial Calcium Signaling in Brain Inflammation and Injury. Int J Mol Sci 2017; 18:ijms18112366. [PMID: 29117112 PMCID: PMC5713335 DOI: 10.3390/ijms18112366] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/20/2022] Open
Abstract
Microglia, the innate immune sentinels of the central nervous system, are the most dynamic cells in the brain parenchyma. They are the first responders to insult and mediate neuroinflammation. Following cellular damage, microglia extend their processes towards the lesion, modify their morphology, release cytokines and other mediators, and eventually migrate towards the damaged area and remove cellular debris by phagocytosis. Intracellular Ca2+ signaling plays important roles in many of these functions. However, Ca2+ in microglia has not been systematically studied in vivo. Here we review recent findings using genetically encoded Ca2+ indicators and two-photon imaging, which have enabled new insights into Ca2+ dynamics and signaling pathways in large populations of microglia in vivo. These new approaches will help to evaluate pre-clinical interventions and immunomodulation for pathological brain conditions such as stroke and neurodegenerative diseases.
Collapse
Affiliation(s)
- Petr Tvrdik
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - M Yashar S Kalani
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
87
|
Taheri M, Handy G, Borisyuk A, White JA. Diversity of Evoked Astrocyte Ca 2+ Dynamics Quantified through Experimental Measurements and Mathematical Modeling. Front Syst Neurosci 2017; 11:79. [PMID: 29109680 PMCID: PMC5660282 DOI: 10.3389/fnsys.2017.00079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/04/2017] [Indexed: 01/06/2023] Open
Abstract
Astrocytes are a major cell type in the mammalian brain. They are not electrically excitable, but generate prominent Ca2+ signals related to a wide variety of critical functions. The mechanisms driving these Ca2+ events remain incompletely understood. In this study, we integrate Ca2+ imaging, quantitative data analysis, and mechanistic computational modeling to study the spatial and temporal heterogeneity of cortical astrocyte Ca2+ transients evoked by focal application of ATP in mouse brain slices. Based on experimental results, we tune a single-compartment mathematical model of IP3-dependent Ca2+ responses in astrocytes and use that model to study response heterogeneity. Using information from the experimental data and the underlying bifurcation structure of our mathematical model, we categorize all astrocyte Ca2+ responses into four general types based on their temporal characteristics: Single-Peak, Multi-Peak, Plateau, and Long-Lasting responses. We find that the distribution of experimentally-recorded response types depends on the location within an astrocyte, with somatic responses dominated by Single-Peak (SP) responses and large and small processes generating more Multi-Peak responses. On the other hand, response kinetics differ more between cells and trials than with location within a given astrocyte. We use the computational model to elucidate possible sources of Ca2+ response variability: (1) temporal dynamics of IP3, and (2) relative flux rates through Ca2+ channels and pumps. Our model also predicts the effects of blocking Ca2+ channels/pumps; for example, blocking store-operated Ca2+ (SOC) channels in the model eliminates Plateau and Long-Lasting responses (consistent with previous experimental observations). Finally, we propose that observed differences in response type distributions between astrocyte somas and processes can be attributed to systematic differences in IP3 rise durations and Ca2+ flux rates.
Collapse
Affiliation(s)
- Marsa Taheri
- Department of Bioengineering, University of Utah, Salt Lake City, UT, United States
| | - Gregory Handy
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States
| | - Alla Borisyuk
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States
| | - John A White
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
88
|
Brawek B, Liang Y, Savitska D, Li K, Fomin-Thunemann N, Kovalchuk Y, Zirdum E, Jakobsson J, Garaschuk O. A new approach for ratiometric in vivo calcium imaging of microglia. Sci Rep 2017; 7:6030. [PMID: 28729628 PMCID: PMC5519759 DOI: 10.1038/s41598-017-05952-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/07/2017] [Indexed: 11/09/2022] Open
Abstract
Microglia, resident immune cells of the brain, react to the presence of pathogens/danger signals with a large repertoire of functional responses including morphological changes, proliferation, chemotaxis, production/release of cytokines, and phagocytosis. In vitro studies suggest that many of these effector functions are Ca2+-dependent, but our knowledge about in vivo Ca2+ signalling in microglia is rudimentary. This is mostly due to technical reasons, as microglia largely resisted all attempts of in vivo labelling with Ca2+ indicators. Here, we introduce a novel approach, utilizing a microglia-specific microRNA-9-regulated viral vector, enabling the expression of a genetically-encoded ratiometric Ca2+ sensor Twitch-2B in microglia. The Twitch-2B-assisted in vivo imaging enables recording of spontaneous and evoked microglial Ca2+ signals and allows for the first time to monitor the steady state intracellular Ca2+ levels in microglia. Intact in vivo microglia show very homogenous and low steady state intracellular Ca2+ levels. However, the levels increase significantly after acute slice preparation and cell culturing along with an increase in the expression of activation markers CD68 and IL-1β. These data identify the steady state intracellular Ca2+ level as a versatile microglial activation marker, which is highly sensitive to the cell's environment.
Collapse
Affiliation(s)
- Bianca Brawek
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | - Yajie Liang
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | - Daria Savitska
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | - Kaizhen Li
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | | | - Yury Kovalchuk
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | - Elizabeta Zirdum
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, 221 84, Sweden
| | - Olga Garaschuk
- Institute of Physiology II, University of Tübingen, 72074, Tübingen, Germany.
| |
Collapse
|
89
|
Abstract
Rapid advances in Ca2+ imaging techniques enable us to simultaneously monitor the activities of hundreds of astrocytes in the intact brain, thus providing a powerful tool for understanding the functions of both host and engrafted astrocytes in sensory processing in vivo. These techniques include both improved Ca2+ indicators and advanced optical recording methods. Astrocytes in multiple cortical and sub-cortical areas are able to respond to the corresponding sensory modalities. These sensory stimuli produce astrocytic Ca2+ responses through different cellular mechanisms. In addition, it has been suggested that astrocytic gene deficiencies in various sensory systems cause impairments in sensory circuits and cognition. Therefore, glial transplantation would be a potentially interesting approach for the cell-based therapy for glia-related disorders. There are multiple cell sources for glial transplantation, including neural stem cells, glial progenitors, and pluripotent stem cells. Both in vitro and in vivo studies have shown that engrafted astrocytes derived from these cell sources are capable of responding to sensory stimulation by elevating the intracellular Ca2+ concentration. These results indicate that engrafted astrocytes not only morphologically but also functionally integrate into the host neural network. Until now, many animal studies have proven that glial transplantation would be a good choice for treating multiple glial disorders. Together, these studies on the sensory responses of host and engrafted astrocytes have provided us a novel perspective in both neuron-glia circuit functions and future treatment strategies for glial disorders.
Collapse
Affiliation(s)
- Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
90
|
Brawek B, Garaschuk O. Monitoring in vivo function of cortical microglia. Cell Calcium 2017; 64:109-117. [DOI: 10.1016/j.ceca.2017.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 02/01/2023]
|
91
|
Oheim M, Schmidt E, Hirrlinger J. Local energy on demand: Are 'spontaneous' astrocytic Ca 2+-microdomains the regulatory unit for astrocyte-neuron metabolic cooperation? Brain Res Bull 2017; 136:54-64. [PMID: 28450076 DOI: 10.1016/j.brainresbull.2017.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/18/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022]
Abstract
Astrocytes are a neural cell type critically involved in maintaining brain energy homeostasis as well as signaling. Like neurons, astrocytes are a heterogeneous cell population. Cortical astrocytes show a complex morphology with a highly branched aborization and numerous fine processes ensheathing the synapses of neighboring neurons, and typically extend one process connecting to blood vessels. Recent studies employing genetically encoded fluorescent calcium (Ca2+) indicators have described 'spontaneous' localized Ca2+-transients in the astrocyte periphery that occur asynchronously, independently of signals in other parts of the cells, and that do not involve somatic Ca2+ transients; however, neither it is known whether these Ca2+-microdomains occur at or near neuronal synapses nor have their molecular basis nor downstream effector(s) been identified. In addition to Ca2+ microdomains, sodium (Na+) transients occur in astrocyte subdomains, too, most likely as a consequence of Na+ co-transport with the neurotransmitter glutamate, which also regulates mitochondrial movements locally - as do cytoplasmic Ca2+ levels. In this review, we cover various aspects of these local signaling events and discuss how structural and biophysical properties of astrocytes might foster such compartmentation. Astrocytes metabolically interact with neurons by providing energy substrates to active neurons. As a single astrocyte branch covers hundreds to thousands of synapses, it is tempting to speculate that these metabolic interactions could occur localized to specific subdomains of astrocytes, perhaps even at the level of small groups of synapses. We discuss how astrocytic metabolism might be regulated at this scale and which signals might contribute to its regulation. We speculate that the astrocytic structures that light up transiently as Ca2+-microdomains might be the functional units of astrocytes linking signaling and metabolic processes to adapt astrocytic function to local energy demands. The understanding of these local regulatory and metabolic interactions will be fundamental to fully appreciate the complexity of brain energy homeostasis as well as its failure in disease and may shed new light on the controversy about neuron-glia bi-directional signaling at the tripartite synapse.
Collapse
Affiliation(s)
- Martin Oheim
- CNRS UMR 8118, Brain Physiology Laboratory, F-75006 Paris, France; Fédération de Recherche en Neurosciences FR3636, Faculté de Sciences Fondamentales et Biomédicales, Université Paris Descartes, PRES Université Sorbonne Paris Cité (USPC), F-75006 Paris, France.
| | - Elke Schmidt
- CNRS UMR 8118, Brain Physiology Laboratory, F-75006 Paris, France; Fédération de Recherche en Neurosciences FR3636, Faculté de Sciences Fondamentales et Biomédicales, Université Paris Descartes, PRES Université Sorbonne Paris Cité (USPC), F-75006 Paris, France
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, D-04103 Leipzig, Germany; Dept. of Neurogenetics, Max-Planck-Institute for Experimental Medicine, D-37075 Göttingen, Germany.
| |
Collapse
|
92
|
Cho JH, Swanson CJ, Chen J, Li A, Lippert LG, Boye SE, Rose K, Sivaramakrishnan S, Chuong CM, Chow RH. The GCaMP-R Family of Genetically Encoded Ratiometric Calcium Indicators. ACS Chem Biol 2017; 12:1066-1074. [PMID: 28195691 PMCID: PMC5572679 DOI: 10.1021/acschembio.6b00883] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report on GCaMP-Rs, a new family of genetically encoded ratiometric calcium indicators that extend the virtues of the GCaMP proteins to ratiometric measurements. We have engineered a tandem construct of calcium-dependent GCaMP and calcium-independent mCherry fluorescent proteins. The tandem design assures that the two proteins localize in the same cellular compartment(s) and facilitates pixelwise ratiometric measurements; however, Förster resonance energy transfer (FRET) between the fluorophores reduces brightness of the sensor by up to half (depending on the GCaMP variant). To eliminate FRET, we introduced a rigid α-helix, the ER/K helix, between GCaMP and mCherry. Avoiding FRET significantly increases the brightness (notably, even at low calcium concentrations), the signal-to-noise ratio, and the dynamic range.
Collapse
Affiliation(s)
- Jung-Hwa Cho
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| | - Carter J. Swanson
- Biophysics Program, University of Michigan, 930 N. University, Room 4028, Ann Arbor, Michigan 48109, United States
| | - Jeannie Chen
- Department of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| | - Ang Li
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| | - Lisa G. Lippert
- Department of Genetics, Cell Biology & Development, University of Minnesota Twin Cities, 4-130 MCB, 420 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Shannon E. Boye
- Department of Ophthalmology, University of Florida, 2000 SW Archer Rd, Rm R3-128, Gainesville, Florida 32611, United States
| | - Kasey Rose
- Department of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology & Development, University of Minnesota Twin Cities, 4-130 MCB, 420 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| | - Robert H. Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
- Department of Biomedical Engineering, Zilkha Neurogenetic Institute, University of Southern California, Room 323, Keck School of Medicine, Los Angeles, California 90089, United States
| |
Collapse
|
93
|
Tufail Y, Cook D, Fourgeaud L, Powers CJ, Merten K, Clark CL, Hoffman E, Ngo A, Sekiguchi KJ, O'Shea CC, Lemke G, Nimmerjahn A. Phosphatidylserine Exposure Controls Viral Innate Immune Responses by Microglia. Neuron 2017; 93:574-586.e8. [PMID: 28111081 DOI: 10.1016/j.neuron.2016.12.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/17/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023]
Abstract
Microglia are the intrinsic immune sentinels of the central nervous system. Their activation restricts tissue injury and pathogen spread, but in some settings, including viral infection, this response can contribute to cell death and disease. Identifying mechanisms that control microglial responses is therefore an important objective. Using replication-incompetent adenovirus 5 (Ad5)-based vectors as a model, we investigated the mechanisms through which microglia recognize and respond to viral uptake. Transgenic, immunohistochemical, molecular-genetic, and fluorescence imaging approaches revealed that phosphatidylserine (PtdSer) exposure on the outer leaflet of transduced cells triggers their engulfment by microglia through TAM receptor-dependent mechanisms. We show that inhibition of phospholipid scramblase 1 (PLSCR1) activity reduces intracellular calcium dysregulation, prevents PtdSer externalization, and enables months-long protection of vector-transduced, transgene-expressing cells from microglial phagocytosis. Our study identifies PLSCR1 as a potent target through which the innate immune response to viral vectors, and potentially other stimuli, may be controlled.
Collapse
Affiliation(s)
- Yusuf Tufail
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniela Cook
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lawrence Fourgeaud
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Colin J Powers
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Katharina Merten
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Charles L Clark
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Elizabeth Hoffman
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alexander Ngo
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kohei J Sekiguchi
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Clodagh C O'Shea
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Greg Lemke
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Immunobiology and Microbial Pathogenesis Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
94
|
Shigetomi E, Koizumi S. Visualization of diversity of calcium signals in astrocytes. Nihon Yakurigaku Zasshi 2017; 148:75-80. [PMID: 27478045 DOI: 10.1254/fpj.148.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
95
|
Garré JM, Yang G, Bukauskas FF, Bennett MVL. An Acute Mouse Spinal Cord Slice Preparation for Studying Glial Activation ex vivo. Bio Protoc 2017; 7:e2102. [PMID: 28503634 DOI: 10.21769/bioprotoc.2102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Pathological conditions such as amyotrophic lateral sclerosis, spinal cord injury and chronic pain are characterized by activation of astrocytes and microglia in spinal cord and have been modeled in rodents. In vivo imaging at cellular level in these animal models is limited due to the spinal cord's highly myelinated funiculi. The preparation of acute slices may offer an alternative and valuable strategy to collect structural and functional information in vitro from dorsal, lateral and ventral regions of spinal cord. Here, we describe a procedure for preparing acute slices from mouse spinal cord (Garré et al., 2016). This preparation should allow further understanding of how glial cells in spinal cord respond acutely to various inflammatory challenges.
Collapse
Affiliation(s)
- Juan Mauricio Garré
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, USA
| | - Guang Yang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, USA
| | - Feliksas F Bukauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, USA.,Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
96
|
McClain JL, Gulbransen BD. The acute inhibition of enteric glial metabolism with fluoroacetate alters calcium signaling, hemichannel function, and the expression of key proteins. J Neurophysiol 2016; 117:365-375. [PMID: 27784805 DOI: 10.1152/jn.00507.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/25/2016] [Indexed: 11/22/2022] Open
Abstract
Glia play key roles in the regulation of neurotransmission in the nervous system. Fluoroacetate (FA) is a metabolic poison widely used to study glial functions by disrupting the tricarboxylic acid cycle enzyme aconitase. Despite the widespread use of FA, the effects of FA on essential glial functions such as calcium (Ca2+) signaling and hemichannel function remain unknown. Therefore, our goal was to assess specifically the impact of FA on essential glial cell functions that are involved with neurotransmission in the enteric nervous system. To this end, we generated a new optogenetic mouse model to study specifically the effects of FA on enteric glial Ca2+ signaling by crossing PC::G5-tdTomato mice with Sox10::creERT2 mice. FA did not change the peak glial Ca2+ response when averaged across all glia within a ganglion. However, FA decreased the percent of responding glia by 30% (P < 0.05) and increased the peak Ca2+ response of the glial cells that still exhibited a response by 26% (P < 0.01). Disruption of Ca2+ signaling with FA impaired the activity-dependent uptake of ethidium bromide through connexin-43 (Cx43) hemichannels (P < 0.05) but did not affect baseline Cx43-dependent dye uptake. FA did not cause overt glial or neurodegeneration, but glial cells significantly increased glial fibrillary acid protein by 56% (P < 0.05) following treatment with FA. Together, these data show that the acute impairment of glial metabolism with FA causes key changes in glial functions associated with their roles in neurotransmission and phenotypic changes indicative of reactive gliosis. NEW & NOTEWORTHY Our study shows that the acute impairment of enteric glial metabolism with fluoroacetate (FA) alters specific glial functions that are associated with the modification of neurotransmission in the gut. These include subtle changes to glial agonist-evoked calcium signaling, the subsequent disruption of connexin-43 hemichannels, and changes in protein expression that are consistent with a transition to reactive glia. These changes in glial function offer a mechanistic explanation for the effects of FA on peripheral neuronal networks.
Collapse
Affiliation(s)
- Jonathon L McClain
- Department of Physiology, Michigan State University, East Lansing, Michigan; and
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan; and .,Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
97
|
Xu J, Bernstein AM, Wong A, Lu XH, Khoja S, Yang XW, Davies DL, Micevych P, Sofroniew MV, Khakh BS. P2X4 Receptor Reporter Mice: Sparse Brain Expression and Feeding-Related Presynaptic Facilitation in the Arcuate Nucleus. J Neurosci 2016; 36:8902-20. [PMID: 27559172 PMCID: PMC4995303 DOI: 10.1523/jneurosci.1496-16.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED P2X4 receptors are ATP-gated cation channels that are widely expressed in the nervous system. To identify P2X4 receptor-expressing cells, we generated BAC transgenic mice expressing tdTomato under the control of the P2X4 receptor gene (P2rx4). We found sparse populations of tdTomato-positive neurons in most brain areas with patterns that matched P2X4 mRNA distribution. tdTomato expression within microglia was low but was increased by an experimental manipulation that triggered microglial activation. We found surprisingly high tdTomato expression in the hypothalamic arcuate nucleus (Arc) (i.e., within parts of the neural circuitry controlling feeding). Immunohistochemistry and genetic crosses of P2rx4 tdTomato mice with cell-specific GFP reporter lines showed that the tdTomato-expressing cells were mainly AgRP-NPY neurons and tanycytes. There was no electrophysiological evidence for functional expression of P2X4 receptors on AgRP-NPY neuron somata, but instead, we found clear evidence for functional presynaptic P2X4 receptor-mediated responses in terminals of AgRP-NPY neurons onto two of their postsynaptic targets (Arc POMC and paraventricular nucleus neurons), where ATP dramatically facilitated GABA release. The presynaptic responses onto POMC neurons, and the expression of tdTomato in AgRP-NPY neurons and tanycytes, were significantly decreased by food deprivation in male mice in a manner that was partially reversed by the satiety-related peptide leptin. Overall, we provide well-characterized tdTomato reporter mice to study P2X4-expressing cells in the brain, new insights on feeding-related regulation of presynaptic P2X4 receptor responses, and the rationale to explore extracellular ATP signaling in the control of feeding behaviors. SIGNIFICANCE STATEMENT Cells expressing ATP-gated P2X4 receptors have proven problematic to identify and study in brain slice preparations because P2X4 expression is sparse. To address this limitation, we generated and characterized BAC transgenic P2rx4 tdTomato reporter mice. We report the distribution of tdTomato-expressing cells throughout the brain and particularly strong expression in the hypothalamic arcuate nucleus. Together, our studies provide a new, well-characterized tool with which to study P2X4 receptor-expressing cells. The electrophysiological studies enabled by this mouse suggest previously unanticipated roles for ATP and P2X4 receptors in the neural circuitry controlling feeding.
Collapse
Affiliation(s)
- Ji Xu
- Departments of Physiology and
| | - Alexander M Bernstein
- Neurobiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Angela Wong
- Neurobiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Xiao-Hong Lu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California-Los Angeles, Los Angeles, California 90095
| | - Sheraz Khoja
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089
| | - X William Yang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California-Los Angeles, Los Angeles, California 90095, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095, and
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Paul Micevych
- Neurobiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Michael V Sofroniew
- Neurobiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Baljit S Khakh
- Departments of Physiology and Neurobiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095,
| |
Collapse
|
98
|
Huang L, Merson TD, Bourne JA. In vivo whole brain, cellular and molecular imaging in nonhuman primate models of neuropathology. Neurosci Biobehav Rev 2016; 66:104-18. [PMID: 27151822 DOI: 10.1016/j.neubiorev.2016.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/31/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Rodents have been the principal model to study brain anatomy and function due to their well-mapped brain architecture, rapid reproduction and amenability to genetic modification. However, there are clear limitations, for example their simpler neocortex, necessitating the need to adopt a model that is closer to humans in order to understand human cognition and brain conditions. Nonhuman primates (NHPs) are ideally suited as they are our closest relatives in the animal kingdom but in vivo imaging technologies to study brain structure and function in these species can be challenging. With the surge in NHP research in recent years, scientists have begun adapting imaging technologies, such as two-photon microscopy, for these species. Here we review the various NHP models that exist as well as their use in advanced microscopic and mesoscopic studies. We discuss the challenges in the field and investigate the opportunities that lie ahead.
Collapse
Affiliation(s)
- Lieven Huang
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia
| | - Tobias D Merson
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria, 3052, Australia
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia.
| |
Collapse
|
99
|
Shigetomi E, Patel S, Khakh BS. Probing the Complexities of Astrocyte Calcium Signaling. Trends Cell Biol 2016; 26:300-312. [PMID: 26896246 PMCID: PMC4946798 DOI: 10.1016/j.tcb.2016.01.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 01/08/2023]
Abstract
Astrocytes are abundant glial cells that tile the entire central nervous system and mediate well-established functions for neurons, blood vessels, and other glia. These ubiquitous cells display intracellular Ca(2+) signals, which have been intensely studied for 25 years. Recently, the use of improved methods has unearthed the panoply of astrocyte Ca(2+) signals and a variable landscape of basal Ca(2+) levels. In vivo studies have started to reveal the settings under which astrocytes display behaviorally relevant Ca(2+) signaling. Studies in mice have emphasized how astrocyte Ca(2+) signaling is altered in distinct neurodegenerative diseases. Progress in the past few years, fueled by methodological advances, has thus reignited interest in astrocyte Ca(2+) signaling for brain function and dysfunction.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
100
|
Nooshabadi F, Yang HJ, Bixler JN, Kong Y, Cirillo JD, Maitland KC. Intravital Fluorescence Excitation in Whole-Animal Optical Imaging. PLoS One 2016; 11:e0149932. [PMID: 26901051 PMCID: PMC4762773 DOI: 10.1371/journal.pone.0149932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/06/2016] [Indexed: 12/22/2022] Open
Abstract
Whole-animal fluorescence imaging with recombinant or fluorescently-tagged pathogens or cells enables real-time analysis of disease progression and treatment response in live animals. Tissue absorption limits penetration of fluorescence excitation light, particularly in the visible wavelength range, resulting in reduced sensitivity to deep targets. Here, we demonstrate the use of an optical fiber bundle to deliver light into the mouse lung to excite fluorescent bacteria, circumventing tissue absorption of excitation light in whole-animal imaging. We present the use of this technology to improve detection of recombinant reporter strains of tdTomato-expressing Mycobacterium bovis BCG (Bacillus Calmette Guerin) bacteria in the mouse lung. A microendoscope was integrated into a whole-animal fluorescence imager to enable intravital excitation in the mouse lung with whole-animal detection. Using this technique, the threshold of detection was measured as 103 colony forming units (CFU) during pulmonary infection. In comparison, the threshold of detection for whole-animal fluorescence imaging using standard epi-illumination was greater than 106 CFU.
Collapse
Affiliation(s)
- Fatemeh Nooshabadi
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Hee-Jeong Yang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Joel N. Bixler
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Ying Kong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Kristen C. Maitland
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|