1
|
Lohr C. Role of P2Y receptors in astrocyte physiology and pathophysiology. Neuropharmacology 2023; 223:109311. [PMID: 36328064 DOI: 10.1016/j.neuropharm.2022.109311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
Astrocytes are active constituents of the brain that manage ion homeostasis and metabolic support of neurons and directly tune synaptic transmission and plasticity. Astrocytes express all known P2Y receptors. These regulate a multitude of physiological functions such as cell proliferation, Ca2+ signalling, gliotransmitter release and neurovascular coupling. In addition, P2Y receptors are fundamental in the transition of astrocytes into reactive astrocytes, as occurring in many brain disorders such as neurodegenerative diseases, neuroinflammation and epilepsy. This review summarizes the current literature addressing the function of P2Y receptors in astrocytes in the healthy brain as well as in brain diseases.
Collapse
Affiliation(s)
- Christian Lohr
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Germany.
| |
Collapse
|
2
|
Abstract
The evolution of the nervous system progressed through cellular diversification and specialization of functions. Conceptually, the nervous system is composed of electrically excitable neuronal networks connected by chemical synapses and nonexcitable glial cells that provide for homeostasis and defense. The evolution of neuroglia began with the emergence of the centralized nervous system and proceeded through a continuous increase in their complexity. In the primate brain, especially in the brain of humans, the astrocyte lineage is exceedingly complex, with the emergence of new types of astroglial cells possibly involved in interlayer communication and integration.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Achucarro Basque Center for Neuroscience, Leioa, Spain.,IKERBASQUE Basque Foundation for Science, Bilbao, Spain.,Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.,Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Amaia M Arranz
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| | - Katarzyna Ciuba
- Dioscuri Centre of Chromatin Biology and Epigenomics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Aleksandra Pękowska
- Dioscuri Centre of Chromatin Biology and Epigenomics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
3
|
Yang R, Song Y, Wang H, Chen C, Bai F, Li C. BmK DKK13, A Scorpion Toxin, Alleviates Pain Behavior in a Rat Model of Trigeminal Neuralgia by Modulating Voltage-Gated Sodium Channels and MAPKs/CREB Pathway. Mol Neurobiol 2022; 59:4535-4549. [PMID: 35579847 DOI: 10.1007/s12035-022-02855-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/27/2022] [Indexed: 12/01/2022]
Abstract
BmK DKK13 (DKK13) is a mutated recombinant peptide, which has a significant antinociception in a rat model of the inflammatory pain. The purpose of this study was to evaluate the antinociceptive effect of DKK13 on trigeminal neuralgia (TN) in rats. Male Sprague-Dawley (SD) rats were treated with the chronic constriction injury of the infraorbital nerve (IoN-CCI) model to induce stable symptoms of TN. DKK13 (1.0 mg/kg, 2.0 mg/kg and 4.0 mg/kg, i.v.) or morphine (4.0 mg/kg, i.v.) was administered by tail vein once on day 14 after IoN-CCI injury. Behavioral tests, electrophysiology and western blotting were performed to investigate the role and underlying mechanisms of DKK13 on IoN-CCI model. Behavioral test results showed that DKK13 could significantly increase the mechanical pain and thermal radiation pain thresholds of IoN-CCI rats and inhibit the asymmetric spontaneous pain scratching behavior. Electrophysiological results showed that DKK13 could significantly reduce the current density of Nav1.8 in the ipsilateral side of trigeminal ganglion (TG) neurons in IoN-CCI rats, and the steady-state activation and inactivation curves of Nav1.8 shifted, respectively, to the direction of hyperpolarization and depolarization. Western blotting results showed that DKK13 significantly reduced the expression of Nav1.8 and the phosphorylation levels of key proteins of MAPKs/CREB pathway in TG tissues of IoN-CCI rats. In brief, DKK13 has a significant antinociceptive effect on IoN-CCI rats, which may be achieved by changing the dynamic characteristics of Nav1.8 channel and regulating the protein phosphorylation in MAPKs/CREB pathway.
Collapse
Affiliation(s)
- Ran Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongbo Song
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Haipeng Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chunyun Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Fei Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chunli Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
4
|
Huang CT, Chen SH, Chang CF, Lin SC, Lue JH, Tsai YJ. Melatonin reduces neuropathic pain behavior and glial activation through MT 2 melatonin receptor modulation in a rat model of lysophosphatidylcholine-induced demyelination neuropathy. Neurochem Int 2020; 140:104827. [PMID: 32853748 DOI: 10.1016/j.neuint.2020.104827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022]
Abstract
In this study, we investigated whether melatonin treatment prevents development of neuropathic pain via suppression of glial mitogen-activated protein kinases (MAPKs) activation in the cuneate nucleus (CN) in a lysophosphatidylcholine (LPC)-induced median nerve demyelination neuropathy model. Rats were fed orally with melatonin once a day at a dose of 37.5, 75, or 150 mg/kg 30 min before until 3 days after LPC treatment. Subsequently, behavioral tests were conducted on these animals, and immunohistochemistry and immunoblotting were used for qualitative and quantitative analysis of glia and MAPKs, including ERK, JNK, and p38, activation. Enzyme-linked immunosorbent assays were applied to measure pro-inflammatory cytokine responses. Furthermore, intra-CN microinjection of S26131 (MT1 receptor antagonist), 4P-PDOT (MT2 receptor antagonist), or prazosin (MT3 receptor antagonist) were performed to investigate the association between melatonin receptor subtypes and effects of melatonin on demyelination neuropathy. LPC treatment of the median nerve induced a significant increase in glial fibrillary acidic protein (GFAP; an astrocyte marker) and ED1 (an activated microglia marker) immunoreactivity in the ipsilateral CN and led to development of neuropathic pain behavior. Inspection of GFAP-immunoreactive astrocytes revealed that astrocytic hypertrophy, but not proliferation, contributed to increased GFAP immunoreactivity. Double immunofluorescence showed that both GFAP-immunoreactive astrocytes and ED1-immunoreactive microglia co-expressed p-ERK, p-JNK, and p-p38 immunoreactivity. Melatonin administration dose-dependently reduced neuropathic pain behavior, decreased glial and MAPKs activation, and diminished the release of pro-inflammatory cytokines in the ipsilateral CN after LPC treatment. Furthermore, 4P-PDOT, but not S26131 or prazosin, antagonized the therapeutic effects of melatonin. In conclusion, administration of melatonin, via its cognate MT2 receptor, inhibited activation of glial MAPKs, production of pro-inflammatory cytokines, and development of demyelination-induced neuropathic pain behavior.
Collapse
Affiliation(s)
- Chun-Ta Huang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Seu-Hwa Chen
- Department of Anatomy, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Chang Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - June-Horng Lue
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Abstract
As the nervous system evolved from the diffused to centralised form, the neurones were joined by the appearance of the supportive cells, the neuroglia. Arguably, these non-neuronal cells evolve into a more diversified cell family than the neurones are. The first ancestral neuroglia appeared in flatworms being mesenchymal in origin. In the nematode C. elegans proto-astrocytes/supportive glia of ectodermal origin emerged, albeit the ensheathment of axons by glial cells occurred later in prawns. The multilayered myelin occurred by convergent evolution of oligodendrocytes and Schwann cells in vertebrates above the jawless fishes. Nutritive partitioning of the brain from the rest of the body appeared in insects when the hemolymph-brain barrier, a predecessor of the blood-brain barrier was formed. The defensive cellular mechanism required specialisation of bona fide immune cells, microglia, a process that occurred in the nervous system of leeches, bivalves, snails, insects and above. In ascending phylogeny, new type of glial cells, such as scaffolding radial glia, appeared and as the bran sizes enlarged, the glia to neurone ratio increased. Humans possess some unique glial cells not seen in other animals.
Collapse
|
6
|
Huang CT, Chen SH, Lin SC, Chen WT, Lue JH, Tsai YJ. Erythropoietin reduces nerve demyelination, neuropathic pain behavior and microglial MAPKs activation through erythropoietin receptors on Schwann cells in a rat model of peripheral neuropathy. Glia 2018; 66:2299-2315. [PMID: 30417431 DOI: 10.1002/glia.23461] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 04/15/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022]
Abstract
Neuroprotective effects of erythropoietin (EPO) on peripheral nerve injury remain uncertain. This study investigated the efficacy of EPO in attenuating median nerve chronic constriction injury (CCI)-induced neuropathy. Animals received an intraneural injection of EPO at doses of 1,000, 3,000, or 5,000 units/kg 15 min before median nerve CCI. Afterwards, the behavioral and electrophysiological tests were conducted. Immunohistochemistry and immunoblotting were used for qualitative and quantitative analysis of microglial and mitogen-activated protein kinases (MAPKs), including p38, JNK, and ERK, activation. Enzyme-linked immunosorbent assay and microdialysis were applied to measure pro-inflammatory cytokine and glutamate responses, respectively. EPO pre-treatment dose-dependently ameliorated neuropathic pain behavior, decreased microglial and MAPKs activation, and diminished the release of pro-inflammatory cytokines and glutamate in the ipsilateral cuneate nucleus after CCI. Moreover, EPO pre-treatment preserved myelination of the injured median nerve on morphological investigation and suppressed injury-induced discharges. We also observed that EPO receptor (EPOR) expression was up-regulated in the injured nerve after CCI. Double immunofluorescence showed that EPOR was localized to Schwann cells. Furthermore, siRNA-mediated knockdown of EPOR expression eliminated the therapeutic effects of EPO on attenuating the microglial and MAPKs activation, pro-inflammatory cytokine responses, injury discharges, and neuropathic pain behavior in CCI rats. In conclusion, binding of EPO to its receptors on Schwann cells maintains myelin integrity and blocks ectopic discharges in the injured median nerve, that in the end contribute to attenuation of neuropathic pain via reducing glutamate release from primary afferents and inhibiting activation of microglial MAPKs and production of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Chun-Ta Huang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Seu-Hwa Chen
- Department of Anatomy, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Chang Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Wei-Ting Chen
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - June-Horng Lue
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
7
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
8
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 964] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
9
|
Verkhratsky A, Zorec R, Parpura V. Stratification of astrocytes in healthy and diseased brain. Brain Pathol 2017; 27:629-644. [PMID: 28805002 PMCID: PMC5599174 DOI: 10.1111/bpa.12537] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/03/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
Astrocytes, a subtype of glial cells, come in variety of forms and functions. However, overarching role of these cell is in the homeostasis of the brain, be that regulation of ions, neurotransmitters, metabolism or neuronal synaptic networks. Loss of homeostasis represents the underlying cause of all brain disorders. Thus, astrocytes are likely involved in most if not all of the brain pathologies. We tabulate astroglial homeostatic functions along with pathological condition that arise from dysfunction of these glial cells. Classification of astrocytes is presented with the emphasis on evolutionary trails, morphological appearance and numerical preponderance. We note that, even though astrocytes from a variety of mammalian species share some common features, human astrocytes appear to be the largest and most complex of all astrocytes studied thus far. It is then an imperative to develop humanized models to study the role of astrocytes in brain pathologies, which is perhaps most abundantly clear in the case of glioblastoma multiforme.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Division of Neuroscience & Experimental PsychologyThe University of ManchesterManchesterUnited Kingdom
- Achúcarro Basque Center for NeuroscienceIKERBASQUE, Basque Foundation for Science48011 BilbaoSpain
- Department of NeuroscienceUniversity of the Basque Country UPV/EHU and CIBERNED48940 LeioaSpain
| | - Robert Zorec
- Laboratory of Cell EngineeringCelica BIOMEDICAL, Tehnološki park 24, Ljubljana 1000SloveniaEurope
- Laboratory of Neuroendocrinology‐Molecular Cell PhysiologyInstitute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana 1000SloveniaEurope
| | - Vladimir Parpura
- Department of Neurobiology, Civitan International Research Center and Center for Glial Biology in Medicine, Evelyn F. McKnight Brain Institute, Atomic Force Microscopy & Nanotechnology Laboratories, 1719 6th Avenue South, CIRC 429University of Alabama at BirminghamBirminghamAL 35294‐0021
| |
Collapse
|
10
|
Neurosteroid Allopregnanolone Suppresses Median Nerve Injury–induced Mechanical Hypersensitivity and Glial Extracellular Signal–regulated Kinase Activation through γ-Aminobutyric Acid Type A Receptor Modulation in the Rat Cuneate Nucleus. Anesthesiology 2016; 125:1202-1218. [DOI: 10.1097/aln.0000000000001360] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Abstract
Background
Mechanisms underlying neuropathic pain relief by the neurosteroid allopregnanolone remain uncertain. We investigated if allopregnanolone attenuates glial extracellular signal-regulated kinase (ERK) activation in the cuneate nucleus (CN) concomitant with neuropathic pain relief in median nerve chronic constriction injury (CCI) model rats.
Methods
We examined the time course and cellular localization of phosphorylated ERK (p-ERK) in CN after CCI. We subsequently employed microinjection of a mitogen-activated protein kinase kinase (ERK kinase) inhibitor, PD98059, to clarify the role of ERK phosphorylation in neuropathic pain development. Furthermore, we explored the effects of allopregnanolone (by mouth), intra-CN microinjection of γ-aminobutyric acid type A receptor antagonist (bicuculline) or γ-aminobutyric acid type B receptor antagonist (phaclofen) plus allopregnanolone, and allopregnanolone synthesis inhibitor (medroxyprogesterone; subcutaneous) on ERK activation and CCI-induced behavioral hypersensitivity.
Results
At 7 days post-CCI, p-ERK levels in ipsilateral CN were significantly increased and reached a peak. PD98059 microinjection into the CN 1 day after CCI dose-dependently attenuated injury-induced behavioral hypersensitivity (withdrawal threshold [mean ± SD], 7.4 ± 1.1, 8.7 ± 1.0, and 10.3 ± 0.8 g for 2.0, 2.5, and 3.0 mM PD98059, respectively, at 7 days post-CCI; n = 6 for each dose). Double immunofluorescence showed that p-ERK was localized to both astrocytes and microglia. Allopregnanolone significantly diminished CN p-ERK levels, glial activation, proinflammatory cytokines, and behavioral hypersensitivity after CCI. Bicuculline, but not phaclofen, blocked all effects of allopregnanolone. Medroxyprogesterone treatment reduced endogenous CN allopregnanolone and exacerbated nerve injury-induced neuropathic pain.
Conclusions
Median nerve injury-induced CN glial ERK activation modulated the development of behavioral hypersensitivity. Allopregnanolone attenuated glial ERK activation and neuropathic pain via γ-aminobutyric acid type A receptors. Reduced endogenous CN allopregnanolone after medroxyprogesterone administration rendered rats more susceptible to CCI-induced neuropathy.
Collapse
|
11
|
Huang CT, Tsai YJ. Docosahexaenoic acid confers analgesic effects after median nerve injury via inhibition of c-Jun N-terminal kinase activation in microglia. J Nutr Biochem 2015; 29:97-106. [PMID: 26895670 DOI: 10.1016/j.jnutbio.2015.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 11/03/2015] [Accepted: 11/20/2015] [Indexed: 12/22/2022]
Abstract
The c-Jun N-terminal kinase (JNK) in the central nervous system plays a critical role in the processing of neuropathic pain. Docosahexaenoic acid (DHA), a predominant omega-3 polyunsaturated fatty acid in the central nervous system, has a neuroprotective efficacy. In this study, we examined the relationships between JNK activation in the cuneate nucleus (CN) and behavioral hypersensitivity after chronic constriction injury (CCI) of the median nerve. We further investigated the effects of DHA administration on JNK activation and development of hypersensitivity. Using immunohistochemistry and immunoblotting, low levels of phosphorylated JNK (p-JNK) were detected in the CN of sham-operated rats. As early as 1 day after CCI, p-JNK levels in the ipsilateral CN were significantly increased and peaked at 7 days. Double-immunofluorescence labeling with cell-specific markers showed that p-JNK immunoreactive cells coexpressed OX-42, a microglia activation marker, suggesting the expression of p-JNK in the microglia. Microinjection of SP600125, a JNK inhibitor, into the CN 1 day after CCI attenuated injury-induced behavioral hypersensitivity in a dose-dependent manner. Furthermore, animals received intravenous injection of DHA at doses of 100, 250 or 500 nmol/kg 30 min after median nerve CCI. DHA treatment decreased p-JNK and OX-42 levels, diminished the release of proinflammatory cytokines and improved behavioral hypersensitivity following CCI. In conclusion, median nerve injury-induced microglial JNK activation in the CN modulated development of behavioral hypersensitivity. DHA has analgesic effects on neuropathic pain, at least in part, by means of suppressing a microglia-mediated inflammatory response through the inhibition of JNK signaling pathway.
Collapse
Affiliation(s)
- Chun-Ta Huang
- Department of Internal Medicine and Traumatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Basic Medicine and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
12
|
Huang CT, Chiang RPY, Chen CL, Tsai YJ. Sleep deprivation aggravates median nerve injury-induced neuropathic pain and enhances microglial activation by suppressing melatonin secretion. Sleep 2014; 37:1513-23. [PMID: 25142572 DOI: 10.5665/sleep.4002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
STUDY OBJECTIVES Sleep deprivation is common in patients with neuropathic pain, but the effect of sleep deprivation on pathological pain remains uncertain. This study investigated whether sleep deprivation aggravates neuropathic symptoms and enhances microglial activation in the cuneate nucleus (CN) in a median nerve chronic constriction injury (CCI) model. Also, we assessed if melatonin supplements during the sleep deprived period attenuates these effects. DESIGN Rats were subjected to sleep deprivation for 3 days by the disc-on-water method either before or after CCI. In the melatonin treatment group, CCI rats received melatonin supplements at doses of 37.5, 75, 150, or 300 mg/kg during sleep deprivation. Melatonin was administered at 23:00 once a day. PARTICIPANTS Male Sprague-Dawley rats, weighing 180-250 g (n = 190), were used. MEASUREMENTS Seven days after CCI, behavioral testing was conducted, and immunohistochemistry, immunoblotting, and enzyme-linked immunosorbent assay were used for qualitative and quantitative analyses of microglial activation and measurements of proinflammatory cytokines. RESULTS In rats who underwent post-CCI sleep deprivation, microglia were more profoundly activated and neuropathic pain was worse than those receiving pre-CCI sleep deprivation. During the sleep deprived period, serum melatonin levels were low over the 24-h period. Administration of melatonin to CCI rats with sleep deprivation significantly attenuated activation of microglia and development of neuropathic pain, and markedly decreased concentrations of proinflammatory cytokines. CONCLUSIONS Sleep deprivation makes rats more vulnerable to nerve injury-induced neuropathic pain, probably because of associated lower melatonin levels. Melatonin supplements to restore a circadian variation in melatonin concentrations during the sleep deprived period could alleviate nerve injury-induced behavioral hypersensitivity.
Collapse
|
13
|
Chiang RPY, Huang CT, Tsai YJ. Melatonin reduces median nerve injury-induced mechanical hypersensitivity via inhibition of microglial p38 mitogen-activated protein kinase activation in rat cuneate nucleus. J Pineal Res 2013; 54:232-44. [PMID: 23237358 DOI: 10.1111/jpi.12029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 11/07/2012] [Indexed: 12/30/2022]
Abstract
In this study, we examined the relationships between p38 mitogen-activated protein kinase (MAPK) activation in the cuneate nucleus (CN) and behavioral hypersensitivity after chronic constriction injury (CCI) of the median nerve. We further investigated effects of melatonin administration and pinealectomy on p38 MAPK activation and development of hypersensitivity. Using immunohistochemistry and immunoblotting, low levels of phosphorylated p38 (p-p38) MAPK were detected in CN of normal rats. As early as 1 day after CCI, p-p38 MAPK levels in the ipsilateral CN were significantly increased (1.4 ± 0.2-fold, P < 0.05), which reached a maximum at 7 days (5.1 ± 0.4-fold, P < 0.001). Double immunofluorescence labeling with cell-specific markers showed that p-p38 MAPK immunoreactive cells co-expressed OX-42, a microglia activation maker, suggesting the expression of p-p38 MAPK in microglia. Microinjection of SB203580, a p38 MAPK inhibitor, into the CN 1 day after CCI attenuated injury-induced behavioral hypersensitivity in a dose-dependent manner. Furthermore, animals received melatonin treatment at daily doses of 37.5, 75, 150, or 300 mg/kg from 30 min before until 3 days after CCI. Melatonin treatment dose-dependently attenuated p-p38 MAPK levels, release of pro-inflammatory cytokines, and behavioral hypersensitivity following CCI; conversely, pinealectomy that resulted in a reduction in endogenous melatonin levels exacerbated these effects. In conclusion, median nerve injury-induced microglial p38 MAPK activation in the CN modulated development of behavioral hypersensitivity. Melatonin supplementation eased neuropathic pain via inhibition of p38 MAPK signaling pathway; contrarily, reducing endogenous blood melatonin levels by pinealectomy promoted phosphorylation of p38 MAPK and made rats more vulnerable to nerve injury-induced neuropathic pain.
Collapse
|
14
|
Stavermann M, Buddrus K, St John JA, Ekberg JA, Nilius B, Deitmer JW, Lohr C. Temperature-dependent calcium-induced calcium release via InsP3 receptors in mouse olfactory ensheathing glial cells. Cell Calcium 2012; 52:113-23. [DOI: 10.1016/j.ceca.2012.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 03/22/2012] [Accepted: 04/24/2012] [Indexed: 02/07/2023]
|
15
|
Development of a glial network in the olfactory nerve: role of calcium and neuronal activity. ACTA ACUST UNITED AC 2011; 6:245-61. [PMID: 21933469 DOI: 10.1017/s1740925x11000081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In adult olfactory nerves of mammals and moths, a network of glial cells ensheathes small bundles of olfactory receptor axons. In the developing antennal nerve (AN) of the moth Manduca sexta, the axons of olfactory receptor neurons (ORNs) migrate from the olfactory sensory epithelium toward the antennal lobe. Here we explore developmental interactions between ORN axons and AN glial cells. During early stages in AN glial-cell migration, glial cells are highly dye coupled, dividing glia are readily found in the nerve and AN glial cells label strongly for glutamine synthetase. By the end of this period, dye-coupling is rare, glial proliferation has ceased, glutamine synthetase labeling is absent, and glial processes have begun to extend to enwrap bundles of axons, a process that continues throughout the remainder of metamorphic development. Whole-cell and perforated-patch recordings in vivo from AN glia at different stages of network formation revealed two potassium currents and an R-like calcium current. Chronic in vivo exposure to the R-type channel blocker SNX-482 halted or greatly reduced AN glial migration. Chronically blocking spontaneous Na-dependent activity by injection of tetrodotoxin reduced the glial calcium current implicating an activity-dependent interaction between ORNs and glial cells in the development of glial calcium currents.
Collapse
|
16
|
Samuels SE, Lipitz JB, Dahl G, Muller KJ. Neuroglial ATP release through innexin channels controls microglial cell movement to a nerve injury. ACTA ACUST UNITED AC 2011; 136:425-42. [PMID: 20876360 PMCID: PMC2947054 DOI: 10.1085/jgp.201010476] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Microglia, the immune cells of the central nervous system, are attracted to sites of injury. The injury releases adenosine triphosphate (ATP) into the extracellular space, activating the microglia, but the full mechanism of release is not known. In glial cells, a family of physiologically regulated unpaired gap junction channels called innexons (invertebrates) or pannexons (vertebrates) located in the cell membrane is permeable to ATP. Innexons, but not pannexons, also pair to make gap junctions. Glial calcium waves, triggered by injury or mechanical stimulation, open pannexon/innexon channels and cause the release of ATP. It has been hypothesized that a glial calcium wave that triggers the release of ATP causes rapid microglial migration to distant lesions. In the present study in the leech, in which a single giant glial cell ensheathes each connective, hydrolysis of ATP with 10 U/ml apyrase or block of innexons with 10 µM carbenoxolone (CBX), which decreased injury-induced ATP release, reduced both movement of microglia and their accumulation at lesions. Directed movement and accumulation were restored in CBX by adding ATP, consistent with separate actions of ATP and nitric oxide, which is required for directed movement but does not activate glia. Injection of glia with innexin2 (Hminx2) RNAi inhibited release of carboxyfluorescein dye and microglial migration, whereas injection of innexin1 (Hminx1) RNAi did not when measured 2 days after injection, indicating that glial cells’ ATP release through innexons was required for microglial migration after nerve injury. Focal stimulation either mechanically or with ATP generated a calcium wave in the glial cell; injury caused a large, persistent intracellular calcium response. Neither the calcium wave nor the persistent response required ATP or its release. Thus, in the leech, innexin membrane channels releasing ATP from glia are required for migration and accumulation of microglia after nerve injury.
Collapse
Affiliation(s)
- Stuart E Samuels
- Neuroscience Program, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
17
|
Verkhratsky A. Physiology of neuronal–glial networking. Neurochem Int 2010; 57:332-43. [DOI: 10.1016/j.neuint.2010.02.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 01/05/2010] [Accepted: 02/01/2010] [Indexed: 10/19/2022]
|
18
|
Araque A, Navarrete M. Glial cells in neuronal network function. Philos Trans R Soc Lond B Biol Sci 2010; 365:2375-81. [PMID: 20603358 DOI: 10.1098/rstb.2009.0313] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Numerous evidence demonstrates that astrocytes, a type of glial cell, are integral functional elements of the synapses, responding to neuronal activity and regulating synaptic transmission and plasticity. Consequently, they are actively involved in the processing, transfer and storage of information by the nervous system, which challenges the accepted paradigm that brain function results exclusively from neuronal network activity, and suggests that nervous system function actually arises from the activity of neuron-glia networks. Most of our knowledge of the properties and physiological consequences of the bidirectional communication between astrocytes and neurons resides at cellular and molecular levels. In contrast, much less is known at higher level of complexity, i.e. networks of cells, and the actual impact of astrocytes in the neuronal network function remains largely unexplored. In the present article, we summarize the current evidence that supports the notion that astrocytes are integral components of nervous system networks and we discuss some functional properties of intercellular signalling in neuron-glia networks.
Collapse
Affiliation(s)
- Alfonso Araque
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain.
| | | |
Collapse
|
19
|
|
20
|
Zhang D, Shooshtarizadeh P, Laventie BJ, Colin DA, Chich JF, Vidic J, de Barry J, Chasserot-Golaz S, Delalande F, Van Dorsselaer A, Schneider F, Helle K, Aunis D, Prévost G, Metz-Boutigue MH. Two chromogranin a-derived peptides induce calcium entry in human neutrophils by calmodulin-regulated calcium independent phospholipase A2. PLoS One 2009; 4:e4501. [PMID: 19225567 PMCID: PMC2639705 DOI: 10.1371/journal.pone.0004501] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 01/15/2009] [Indexed: 12/11/2022] Open
Abstract
Background Antimicrobial peptides derived from the natural processing of chromogranin A (CgA) are co-secreted with catecholamines upon stimulation of chromaffin cells. Since PMNs play a central role in innate immunity, we examine responses by PMNs following stimulation by two antimicrobial CgA-derived peptides. Methodology/Principal Findings PMNs were treated with different concentrations of CgA-derived peptides in presence of several drugs. Calcium mobilization was observed by using flow cytometry and calcium imaging experiments. Immunocytochemistry and confocal microscopy have shown the intracellular localization of the peptides. The calmodulin-binding and iPLA2 activating properties of the peptides were shown by Surface Plasmon Resonance and iPLA2 activity assays. Finally, a proteomic analysis of the material released after PMNs treatment with CgA-derived peptides was performed by using HPLC and Nano-LC MS-MS. By using flow cytometry we first observed that after 15 s, in presence of extracellular calcium, Chromofungin (CHR) or Catestatin (CAT) induce a concentration-dependent transient increase of intracellular calcium. In contrast, in absence of extra cellular calcium the peptides are unable to induce calcium depletion from the stores after 10 minutes exposure. Treatment with 2-APB (2-aminoethoxydiphenyl borate), a store operated channels (SOCs) blocker, inhibits completely the calcium entry, as shown by calcium imaging. We also showed that they activate iPLA2 as the two CaM-binding factors (W7 and CMZ) and that the two sequences can be aligned with the two CaM-binding domains reported for iPLA2. We finally analyzed by HPLC and Nano-LC MS-MS the material released by PMNs following stimulation by CHR and CAT. We characterized several factors important for inflammation and innate immunity. Conclusions/Significance For the first time, we demonstrate that CHR and CAT, penetrate into PMNs, inducing extracellular calcium entry by a CaM-regulated iPLA2 pathway. Our study highlights the role of two CgA-derived peptides in the active communication between neuroendocrine and immune systems.
Collapse
Affiliation(s)
- Dan Zhang
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
- Département de Réanimation Médicale, Hôpital de Hautepierre, Strasbourg, France
- First Hospital, Chongqing University of Medical Sciences, Chongqing, China
| | | | - Benoît-Joseph Laventie
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Didier André Colin
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean-François Chich
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
- INRA, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jasmina Vidic
- INRA, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jean de Barry
- Institut des Neurosciences Cellulaires et Intégratives, UMR 7168 CNRS-Université Louis Pasteur, Strasbourg, France
| | - Sylvette Chasserot-Golaz
- Institut des Neurosciences Cellulaires et Intégratives, UMR 7168 CNRS-Université Louis Pasteur, Strasbourg, France
| | | | - Alain Van Dorsselaer
- Laboratoire de spectrométrie de masse BioOrganique, IPHC-DSA, ULP, CNRS, UMR7178, Strasbourg, France
| | - Francis Schneider
- Département de Réanimation Médicale, Hôpital de Hautepierre, Strasbourg, France
| | - Karen Helle
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Dominique Aunis
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
| | - Gilles Prévost
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | |
Collapse
|
21
|
Verkhratsky A. Neuronismo y reticulismo: neuronal-glial circuits unify the reticular and neuronal theories of brain organization. Acta Physiol (Oxf) 2009; 195:111-22. [PMID: 18983447 DOI: 10.1111/j.1748-1716.2008.01926.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The neuronal doctrine, which shaped the development of neuroscience, was born from a long-lasting struggle between reticularists, who assumed internal continuity of neural networks and neuronists, who defined the brain as a network of physically separated cellular entities, defined as neurones. Modern views regard the brain as a complex of constantly interacting cellular circuits, represented by neuronal networks embedded into internally connected astroglial syncytium. The neuronal-glial circuits endowed with distinct signalling cascades form a 'diffuse nervous net' suggested by Golgi, where millions of synapses belonging to very different neurones are integrated first into neuronal-glial-vascular units and then into more complex structures connected through glial syncytium. These many levels of integration, both morphological and functional, presented by neuronal-glial circuitry ensure the spatial and temporal multiplication of brain cognitive power.
Collapse
Affiliation(s)
- A Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
22
|
Hirth IC, Britz FC, Deitmer JW. G protein activation by uncaging of GTP-gamma-S in the leech giant glial cell. ACTA ACUST UNITED AC 2008; 210:3771-9. [PMID: 17951418 DOI: 10.1242/jeb.008037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glial cells can be activated by neurotransmitters via metabotropic, G protein-coupled receptors. We have studied the effects of 'global' G protein activation by GTP-gamma-S on the membrane potential, membrane conductance, intracellular Ca(2+) and Na(+) of the giant glial cell in isolated ganglia of the leech Hirudo medicinalis. Uncaging GTP-gamma-S (injected into a giant glial cell as caged compound) by moderate UV illumination hyperpolarized the membrane due to an increase in K+ conductance. Uncaging GTP-gamma-S also evoked rises in cytosolic Ca(2+) and Na+, both of which were suppressed after depleting the intracellular Ca(2+) stores with cyclopiazonic acid (20 micromol l(-1)). Uncaging inositol-trisphosphate evoked a transient rise in cytosolic Ca(2+) and Na+ but no change in membrane potential. Injection of the fast Ca(2+) chelator BAPTA or depletion of intracellular Ca(2+) stores did not suppress the membrane hyperpolarization induced by uncaging GTP-gamma-S. Our results suggest that global activation of G proteins in the leech giant glial cell results in a rise of Ca(2+)-independent membrane K+ conductance, a rise of cytosolic Ca(2+), due to release from intracellular stores, and a rise of cytosolic Na+, presumably due to increased Na+/Ca(2+) exchange.
Collapse
Affiliation(s)
- Ingo C Hirth
- Abteilung für Allgemeine Zoologie, FB Biologie, TU Kaiserslautern, Kaiserslautern, Germany
| | | | | |
Collapse
|
23
|
Murai KK, Van Meyel DJ. Neuron glial communication at synapses: insights from vertebrates and invertebrates. Neuroscientist 2007; 13:657-66. [PMID: 17911218 DOI: 10.1177/1073858407304393] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Glial cells are instrumental for many aspects of nervous-system function. Interestingly, complex neuron-glial interactions at synapses are commonly found in both invertebrates and vertebrates. Although these interactions are known to be important for synaptic physiology, the cellular processes and molecular mechanisms involved have not been fully uncovered. Identifying the common and unique features of neuron-glial interactions between invertebrates and vertebrates may provide valuable insights into the relationship of neuron-glial cross-talk to nervous-system function. This review highlights selected studies that have revealed structural and functional insights into neuron-glial interactions at synapses in invertebrate and vertebrate model systems.
Collapse
Affiliation(s)
- Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, McGill University the Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | | |
Collapse
|
24
|
Schuckel J, Siwicki KK, Stengl M. Putative circadian pacemaker cells in the antenna of the hawkmoth Manduca sexta. Cell Tissue Res 2007; 330:271-8. [PMID: 17786482 DOI: 10.1007/s00441-007-0471-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 07/08/2007] [Indexed: 11/25/2022]
Abstract
Antennal sensory neurons in the fruit fly Drosophila melanogaster express circadian rhythms in the clock gene PERIOD (PER) and appear to be sufficient and necessary for circadian rhythms in olfactory responses. Given recent evidence for daily rhythms of pheromone responses in the antenna of the hawkmoth Manduca sexta, we examined whether a peripheral PER-based circadian clock might be present in this species. Several different cell types in the moth antenna were recognized by monoclonal antibodies against Manduca sexta PER. In addition to PER-like staining of pheromone-sensitive olfactory receptor neurons and supporting cells, immunoreactivity was detected in beaded branches contacting the pheromone-sensitive sensilla. The nuclei of apparently all sensory receptor neurons, of sensilla supporting cells, of epithelial cells, and of antennal nerve glial cells were PER-immunoreactive. Expression of per mRNA in antennae was confirmed by the polymerase chain reaction, which showed stronger expression at Zeitgeber-time 15 compared with Zeitgeber-time 3. This evidence for the expression of per gene products suggests that the antenna of the hawkmoth contains endogenous circadian clocks.
Collapse
Affiliation(s)
- Julia Schuckel
- Biology, Animal Physiology, Philipps University of Marburg, Karl von Frisch Strasse, 35043, Marburg, Germany
| | | | | |
Collapse
|
25
|
Heil JE, Oland LA, Lohr C. Acetylcholine-mediated axon-glia signaling in the developing insect olfactory system. Eur J Neurosci 2007; 26:1227-41. [PMID: 17767501 DOI: 10.1111/j.1460-9568.2007.05756.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In the olfactory system of the sphinx moth Manduca sexta, migration of neuropil glial cells is triggered by olfactory receptor axons and depends on intraglial Ca(2+) signaling. It is not known, however, how receptor axons and glial cells communicate and whether Ca(2+) signaling is a consequence of this communication. We studied Ca(2+) increases in glial cells in vivo and in situ, evoked by electrical stimulation of olfactory receptor axons in pupae and by odor stimulation of receptor neurons in adult moths. Axonal activity leads to Ca(2+) increases in neuropil glial cells that are blocked by nicotinic acetylcholine receptor antagonists and can be mimicked by acetylcholine and carbachol application. In addition, Ca(2+) transients were abolished by removal of external Ca(2+) and blockage of voltage-gated Ca(2+) channels. During development, acetylcholine-mediated Ca(2+) signaling could first be elicited at stage 6, the time when neuropil glial cells start to migrate. Glial migration was reduced after injection of nicotinic antagonists into pupae. The results show that Ca(2+) signaling can be induced by acetylcholine release from olfactory receptor axons, which activates nicotinic acetylcholine receptors and leads to voltage-gated Ca(2+) influx. The results further suggest that cholinergic signaling in the olfactory system is required for glial cell migration in Manduca.
Collapse
Affiliation(s)
- Jan E Heil
- Abteilung für Allgemeine Zoologie, TU Kaiserslautern, POB 3049, 67653 Kaiserslautern, Germany
| | | | | |
Collapse
|
26
|
Hartl S, Heil JE, Hirsekorn A, Lohr C. A novel neurotransmitter-independent communication pathway between axons and glial cells. Eur J Neurosci 2007; 25:945-56. [PMID: 17331192 DOI: 10.1111/j.1460-9568.2007.05351.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent studies have provided evidence that transmitters released by neurons can activate glial receptors and stimulate calcium signalling in glial cells. Glial calcium signalling, in turn, may affect neuronal performance such as long-term changes in synaptic efficacy. Olfactory ensheathing cells (OECs) are a special glial cell type in vertebrates and insects and promote axon growth in the developing and mature nervous system. Physiological properties of OECs, however, have not been studied so far in detail. We measured changes in the calcium concentration in OECs of the moth Manduca sexta, in situ and in vivo. Electrical stimulation of olfactory receptor neurons in pupae or odour stimulation of receptor neurons in adults resulted in calcium transients in OECs. Olfactory receptor axons release acetylcholine; however, application of acetylcholine or other transmitters such as glutamate, GABA or nitric oxide did not induce calcium transients in OECs. Upon nerve stimulation, extracellular potassium rose by several millimolar as measured with potassium-sensitive microelectrodes. When potassium in the perfusion saline was increased from 4 to 10 mM or higher, voltage-dependent calcium transients in OECs that resembled stimulation-induced calcium transients were evoked. Blocking neuronal potassium channels with TEA reduced both the stimulation-induced increases in extracellular potassium and the calcium transients in OECs, whereas calcium transients in receptor axons were augmented. Our results show for the first time that accumulation of potassium, released by electrically active axons, is sufficient to evoke voltage-dependent calcium influx into glial cells, whereas neurotransmitters appear not to be involved in this neuron-glia communication in Manduca.
Collapse
Affiliation(s)
- Sandra Hartl
- Abteilung für Allgemeine Zoologie, T. U. Kaiserslautern, PO Box 3049, 67653 Kaiserslautern, Germany
| | | | | | | |
Collapse
|
27
|
Rieger A, Deitmer JW, Lohr C. Axon-glia communication evokes calcium signaling in olfactory ensheathing cells of the developing olfactory bulb. Glia 2007; 55:352-9. [PMID: 17136772 DOI: 10.1002/glia.20460] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Olfactory ensheathing cells (OECs) accompany receptor axons in the olfactory nerve and promote axonal growth into the central nervous system. The mechanisms underlying the communication between axons and OECs, however, have not been studied in detail yet. We investigated the effect of activity-dependent neuronal transmitter release on Ca(2+) signaling of OECs in acute mouse olfactory bulb slices using confocal Ca(2+) imaging. TTX-sensitive axonal activity upon electrical nerve stimulation triggers a rise in cytosolic Ca(2+) in OECs, which can be mimicked by application of DHPG, an agonist of metabotropic glutamate receptors (mGluRs). Both stimulation- and DHPG-induced Ca(2+) transients in OECs were abolished by depletion of intracellular Ca(2+) stores with cyclopiazonic acid (CPA). The mGluR(1)-specific antagonist CPCCOEt completely inhibited DHPG-evoked Ca(2+) transients, but reduced stimulation-induced Ca(2+) transients only partly, suggesting the involvement of another neurotransmitter. Application of ATP evoked CPA-sensitive Ca(2+) transients in OECs, which were inhibited by the P2Y(1)-specific antagonist MRS2179. Co-application of CPCCOEt and MRS2179 almost completely blocked the stimulation-induced Ca(2+) transients, indicating that they were mediated by mGluR(1) and P2Y(1) receptors. Our results show that OECs are able to respond to olfactory nerve activity with an increase in cytosolic Ca(2+) due to glutamate and ATP release.
Collapse
Affiliation(s)
- Anne Rieger
- Abteilung für Allgemeine Zoologie, Technische Universität Kaiserslautern, Postfach 3049, 67653 Kaiserslautern, Germany.
| | | | | |
Collapse
|