1
|
Chen YH, Lin S, Jin SY, Gao TM. Extracellular ATP Is a Homeostatic Messenger That Mediates Cell-Cell Communication in Physiological Processes and Psychiatric Diseases. Biol Psychiatry 2025; 97:41-53. [PMID: 38679359 DOI: 10.1016/j.biopsych.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
Neuronal activity is the basis of information encoding and processing in the brain. During neuronal activation, intracellular ATP (adenosine triphosphate) is generated to meet the high-energy demands. Simultaneously, ATP is secreted, increasing the extracellular ATP concentration and acting as a homeostatic messenger that mediates cell-cell communication to prevent aberrant hyperexcitability of the nervous system. In addition to the confined release and fast synaptic signaling of classic neurotransmitters within synaptic clefts, ATP can be released by all brain cells, diffuses widely, and targets different types of purinergic receptors on neurons and glial cells, making it possible to orchestrate brain neuronal activity and participate in various physiological processes, such as sleep and wakefulness, learning and memory, and feeding. Dysregulation of extracellular ATP leads to a destabilizing effect on the neural network, as found in the etiopathology of many psychiatric diseases, including depression, anxiety, schizophrenia, and autism spectrum disorder. In this review, we summarize advances in the understanding of the mechanisms by which extracellular ATP serves as an intercellular signaling molecule to regulate neural activity, with a focus on how it maintains the homeostasis of neural networks. In particular, we also focus on neural activity issues that result from dysregulation of extracellular ATP and propose that aberrant levels of extracellular ATP may play a role in the etiopathology of some psychiatric diseases, highlighting the potential therapeutic targets of ATP signaling in the treatment of these psychiatric diseases. Finally, we suggest potential avenues to further elucidate the role of extracellular ATP in intercellular communication and psychiatric diseases.
Collapse
Affiliation(s)
- Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Song Lin
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Benitez MJ, Retana D, Ordoñez-Gutiérrez L, Colmena I, Goméz MJ, Álvarez R, Ciorraga M, Dopazo A, Wandosell F, Garrido JJ. Transcriptomic alterations in APP/PS1 mice astrocytes lead to early postnatal axon initial segment structural changes. Cell Mol Life Sci 2024; 81:444. [PMID: 39485512 PMCID: PMC11530419 DOI: 10.1007/s00018-024-05485-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 09/20/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024]
Abstract
Alzheimer´s disease (AD) is characterized by neuronal function loss and degeneration. The integrity of the axon initial segment (AIS) is essential to maintain neuronal function and output. AIS alterations are detected in human post-mortem AD brains and mice models, as well as, neurodevelopmental and mental disorders. However, the mechanisms leading to AIS deregulation in AD and the extrinsic glial origin are elusive. We studied early postnatal differences in AIS cellular/molecular mechanisms in wild-type or APP/PS1 mice and combined neuron-astrocyte co-cultures. We observed AIS integrity alterations, reduced ankyrinG expression and shortening, in APP/PS1 mice from P21 and loss of AIS integrity at 21 DIV in wild-type and APP/PS1 neurons in the presence of APP/PS1 astrocytes. AnkyrinG decrease is due to mRNAs and protein reduction of retinoic acid synthesis enzymes Rdh1 and Aldh1b1, as well as ADNP (Activity-dependent neuroprotective protein) in APP/PS1 astrocytes. This effect was mimicked by wild-type astrocytes expressing ADNP shRNA. In the presence of APP/PS1 astrocytes, wild-type neurons AIS is recovered by inhibition of retinoic acid degradation, and Adnp-derived NAP peptide (NAPVSIPQ) addition or P2X7 receptor inhibition, both regulated by retinoic acid levels. Moreover, P2X7 inhibitor treatment for 2 months impaired AIS disruption in APP/PS1 mice. Our findings extend current knowledge on AIS regulation, providing data to support the role of astrocytes in early postnatal AIS modulation. In conclusion, AD onset may be related to very early glial cell alterations that induce AIS and neuronal function changes, opening new therapeutic approaches to detect and avoid neuronal function loss.
Collapse
Affiliation(s)
- María José Benitez
- Instituto Cajal, CSIC, Madrid, Spain
- Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Lara Ordoñez-Gutiérrez
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Inés Colmena
- Instituto Cajal, CSIC, Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
| | | | - Rebeca Álvarez
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Wandosell
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan José Garrido
- Instituto Cajal, CSIC, Madrid, Spain.
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain.
| |
Collapse
|
3
|
Zhang X, Zhou H, Liu H, Xu P. Role of Oxidative Stress in the Occurrence and Development of Cognitive Dysfunction in Patients with Obstructive Sleep Apnea Syndrome. Mol Neurobiol 2024; 61:5083-5101. [PMID: 38159196 DOI: 10.1007/s12035-023-03899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Obstructive sleep apnea syndrome (OSAS) causes recurrent apnea and intermittent hypoxia at night, leading to several complications such as cognitive dysfunction. However, the molecular mechanisms underlying cognitive dysfunction in OSAS are unclear, and oxidative stress mediated by intermittent hypoxia is an important mechanism. In addition, the improvement of cognitive dysfunction in patients with OSAS varies by different treatment regimens; among them, continuous positive airway pressure therapy (CPAP) is mostly recognized for improving cognitive dysfunction. In this review, we discuss the potential mechanisms of oxidative stress in OSAS, the common factors of affecting oxidative stress and the Links between oxidative stress and inflammation in OSAS, focusing on the potential links between oxidative stress and cognitive dysfunction in OSAS and the potential therapies for neurocognitive dysfunction in patients with OSAS mediated by oxidative stress. Therefore, further analysis on the relationship between oxidative stress and cognitive dysfunction in patients with OSAS will help to clarify the etiology and discover new treatment options, which will be of great significance for early clinical intervention.
Collapse
Affiliation(s)
- XiaoPing Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongyan Zhou
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - HaiJun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
4
|
Uzay B, Donmez-Demir B, Ozcan SY, Kocak EE, Yemisci M, Ozdemir YG, Dalkara T, Karatas H. The effect of P2X7 antagonism on subcortical spread of optogenetically-triggered cortical spreading depression and neuroinflammation. J Headache Pain 2024; 25:120. [PMID: 39044141 PMCID: PMC11267761 DOI: 10.1186/s10194-024-01807-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 06/06/2024] [Indexed: 07/25/2024] Open
Abstract
Migraine is a neurological disorder characterized by episodes of severe headache. Cortical spreading depression (CSD), the electrophysiological equivalent of migraine aura, results in opening of pannexin 1 megachannels that release ATP and triggers parenchymal neuroinflammatory signaling cascade in the cortex. Migraine symptoms suggesting subcortical dysfunction bring subcortical spread of CSD under the light. Here, we investigated the role of purinergic P2X7 receptors on the subcortical spread of CSD and its consequent neuroinflammation using a potent and selective P2X7R antagonist, JNJ-47965567. P2X7R antagonism had no effect on the CSD threshold and characteristics but increased the latency to hypothalamic voltage deflection following CSD suggesting that ATP acts as a mediator in the subcortical spread. P2X7R antagonism also prevented cortical and subcortical neuronal activation following CSD, revealed by bilateral decrease in c-fos positive neuron count, and halted CSD-induced neuroinflammation revealed by decreased neuronal HMGB1 release and decreased nuclear translocation of NF-kappa B-p65 in astrocytes. In conclusion, our data suggest that P2X7R plays a role in CSD-induced neuroinflammation, subcortical spread of CSD and CSD-induced neuronal activation hence can be a potential target.
Collapse
Affiliation(s)
- Burak Uzay
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Buket Donmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
| | - Sinem Yilmaz Ozcan
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
| | - Emine Eren Kocak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
- Department of Psychiatry, Hacettepe University, Ankara, Türkiye
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
- Department of Neurology, Hacettepe University, Ankara, Türkiye
| | - Yasemin Gursoy Ozdemir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
- School of Medicine, Koc University, Istanbul, Türkiye
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
- Department of Neurology, Hacettepe University, Ankara, Türkiye
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye.
| |
Collapse
|
5
|
Chen Q, Li L, Xu L, Yang B, Huang Y, Qiao D, Yue X. Proteomic analysis discovers potential biomarkers of early traumatic axonal injury in the brainstem. Int J Legal Med 2024; 138:207-227. [PMID: 37338605 DOI: 10.1007/s00414-023-03039-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/01/2023] [Indexed: 06/21/2023]
Abstract
OBJECTIVE Application of Tandem Mass Tags (TMT)-based LC-MS/MS analysis to screen for differentially expressed proteins (DEPs) in traumatic axonal injury (TAI) of the brainstem and to predict potential biomarkers and key molecular mechanisms of brainstem TAI. METHODS A modified impact acceleration injury model was used to establish a brainstem TAI model in Sprague-Dawley rats, and the model was evaluated in terms of both functional changes (vital sign measurements) andstructural changes (HE staining, silver-plating staining and β-APP immunohistochemical staining). TMT combined with LC-MS/MS was used to analyse the DEPs in brainstem tissues from TAI and Sham groups. The biological functions of DEPs and potential molecular mechanisms in the hyperacute phase of TAI were analysed by bioinformatics techniques, and candidate biomarkers were validated using western blotting and immunohistochemistry on brainstem tissues from animal models and humans. RESULTS Based on the successful establishment of the brainstem TAI model in rats, TMT-based proteomics identified 65 DEPs, and bioinformatics analysis indicated that the hyperacute phase of TAI involves multiple stages of biological processes including inflammation, oxidative stress, energy metabolism, neuronal excitotoxicity and apoptosis. Three DEPs, CBR1, EPHX2 and CYP2U1, were selected as candidate biomarkers and all three proteins were found to be significantly expressed in brainstem tissue 30 min-7 days after TAI in both animal models and humans. CONCLUSION Using TMT combined with LC-MS/MS analysis for proteomic study of early TAI in rat brainstem, we report for the first time that CBR1, EPHX2 and CYP2U1 can be used as biomarkers of early TAI in brainstem by means of western blotting and immunohistochemical staining, compensating for the limitations of silver-plating staining and β-APP immunohistochemical staining, especially in the case of very short survival time after TAI (shorter than 30 min). A number of other proteins that also have a potential marker role are also presented, providing new insights into the molecular mechanisms, therapeutic targets and forensic identification of early TAI in brainstem.
Collapse
Affiliation(s)
- Qianling Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lingyue Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Luyao Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Bin Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuebing Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dongfang Qiao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
6
|
Fréal A, Jamann N, Ten Bos J, Jansen J, Petersen N, Ligthart T, Hoogenraad CC, Kole MH. Sodium channel endocytosis drives axon initial segment plasticity. SCIENCE ADVANCES 2023; 9:eadf3885. [PMID: 37713493 PMCID: PMC10881073 DOI: 10.1126/sciadv.adf3885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
Activity-dependent plasticity of the axon initial segment (AIS) endows neurons with the ability to adapt action potential output to changes in network activity. Action potential initiation at the AIS highly depends on the clustering of voltage-gated sodium channels, but the molecular mechanisms regulating their plasticity remain largely unknown. Here, we developed genetic tools to label endogenous sodium channels and their scaffolding protein, to reveal their nanoscale organization and longitudinally image AIS plasticity in hippocampal neurons in slices and primary cultures. We find that N-methyl-d-aspartate receptor activation causes both long-term synaptic depression and rapid internalization of AIS sodium channels within minutes. The clathrin-mediated endocytosis of sodium channels at the distal AIS increases the threshold for action potential generation. These data reveal a fundamental mechanism for rapid activity-dependent AIS reorganization and suggests that plasticity of intrinsic excitability shares conserved features with synaptic plasticity.
Collapse
Affiliation(s)
- Amélie Fréal
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Nora Jamann
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jolijn Ten Bos
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jacqueline Jansen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Naomi Petersen
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Thijmen Ligthart
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Casper C. Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Neuroscience, Genentech Inc, South San Francisco, CA, USA
| | - Maarten H. P. Kole
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
7
|
Garrido JJ. Contribution of Axon Initial Segment Structure and Channels to Brain Pathology. Cells 2023; 12:cells12081210. [PMID: 37190119 DOI: 10.3390/cells12081210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Brain channelopathies are a group of neurological disorders that result from genetic mutations affecting ion channels in the brain. Ion channels are specialized proteins that play a crucial role in the electrical activity of nerve cells by controlling the flow of ions such as sodium, potassium, and calcium. When these channels are not functioning properly, they can cause a wide range of neurological symptoms such as seizures, movement disorders, and cognitive impairment. In this context, the axon initial segment (AIS) is the site of action potential initiation in most neurons. This region is characterized by a high density of voltage-gated sodium channels (VGSCs), which are responsible for the rapid depolarization that occurs when the neuron is stimulated. The AIS is also enriched in other ion channels, such as potassium channels, that play a role in shaping the action potential waveform and determining the firing frequency of the neuron. In addition to ion channels, the AIS contains a complex cytoskeletal structure that helps to anchor the channels in place and regulate their function. Therefore, alterations in this complex structure of ion channels, scaffold proteins, and specialized cytoskeleton may also cause brain channelopathies not necessarily associated with ion channel mutations. This review will focus on how the AISs structure, plasticity, and composition alterations may generate changes in action potentials and neuronal dysfunction leading to brain diseases. AIS function alterations may be the consequence of voltage-gated ion channel mutations, but also may be due to ligand-activated channels and receptors and AIS structural and membrane proteins that support the function of voltage-gated ion channels.
Collapse
Affiliation(s)
- Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28002 Madrid, Spain
| |
Collapse
|
8
|
Lezmy J. How astrocytic ATP shapes neuronal activity and brain circuits. Curr Opin Neurobiol 2023; 79:102685. [PMID: 36746109 DOI: 10.1016/j.conb.2023.102685] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 02/06/2023]
Abstract
Astrocytes play a key role in processing information at synapses, by controlling synapse formation, modulating synapse strength and terminating neurotransmitter action. They release ATP to shape brain activity but it is unclear how, as astrocyte processes contact many targets and ATP-mediated effects are diverse and numerous. Here, I review recent studies showing how astrocytic ATP modulates cellular mechanisms in nearby neurons and glia in the grey and white matter, how it affects signal transmission in these areas, and how it modulates behavioural outputs. I attempt to provide a flowchart of astrocytic ATP signalling, showing that it tends to inhibit neural circuits to match energy demands.
Collapse
Affiliation(s)
- Jonathan Lezmy
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
9
|
Huang Y, Omorou M, Gao M, Mu C, Xu W, Xu H. Hydrogen sulfide and its donors for the treatment of cerebral ischaemia-reperfusion injury: A comprehensive review. Biomed Pharmacother 2023; 161:114506. [PMID: 36906977 DOI: 10.1016/j.biopha.2023.114506] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
As an endogenous gas signalling molecule, hydrogen sulfide (H2S) is frequently present in a variety of mammals and plays a significant role in the cardiovascular and nervous systems. Reactive oxygen species (ROS) are produced in large quantities as a result of cerebral ischaemia-reperfusion, which is a very serious class of cerebrovascular diseases. ROS cause oxidative stress and induce specific gene expression that results in apoptosis. H2S reduces cerebral ischaemia-reperfusion-induced secondary injury via anti-oxidative stress injury, suppression of the inflammatory response, inhibition of apoptosis, attenuation of cerebrovascular endothelial cell injury, modulation of autophagy, and antagonism of P2X7 receptors, and it plays an important biological role in other cerebral ischaemic injury events. Despite the many limitations of the hydrogen sulfide therapy delivery strategy and the difficulty in controlling the ideal concentration, relevant experimental evidence demonstrating that H2S plays an excellent neuroprotective role in cerebral ischaemia-reperfusion injury (CIRI). This paper examines the synthesis and metabolism of the gas molecule H2S in the brain as well as the molecular mechanisms of H2S donors in cerebral ischaemia-reperfusion injury and possibly other unknown biological functions. With the active development in this field, it is expected that this review will assist researchers in their search for the potential value of hydrogen sulfide and provide new ideas for preclinical trials of exogenous H2S.
Collapse
Affiliation(s)
- Yiwei Huang
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| | - Moussa Omorou
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Meng Gao
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Chenxi Mu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Weijing Xu
- School of Public Health, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| |
Collapse
|
10
|
Jadhav P, Karande M, Sarkar A, Sahu S, Sarmah D, Datta A, Chaudhary A, Kalia K, Sharma A, Wang X, Bhattacharya P. Glial Cells Response in Stroke. Cell Mol Neurobiol 2023; 43:99-113. [PMID: 35066715 DOI: 10.1007/s10571-021-01183-3] [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: 10/21/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023]
Abstract
As the second-leading cause of death, stroke faces several challenges in terms of treatment because of the limited therapeutic interventions available. Previous studies primarily focused on metabolic and blood flow properties as a target for treating stroke, including recombinant tissue plasminogen activator and mechanical thrombectomy, which are the only USFDA approved therapies. These interventions have the limitation of a narrow therapeutic time window, the possibility of hemorrhagic complications, and the expertise required for performing these interventions. Thus, it is important to identify the contributing factors that exacerbate the ischemic outcome and to develop therapies targeting them for regulating cellular homeostasis, mainly neuronal survival and regeneration. Glial cells, primarily microglia, astrocytes, and oligodendrocytes, have been shown to have a crucial role in the prognosis of ischemic brain injury, contributing to inflammatory responses. They play a dual role in both the onset as well as resolution of the inflammatory responses. Understanding the different mechanisms driving these effects can aid in the development of therapeutic targets and further mitigate the damage caused. In this review, we summarize the functions of various glial cells and their contribution to stroke pathology. The review highlights the therapeutic options currently being explored and developed that primarily target glial cells and can be used as neuroprotective agents for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Poonam Jadhav
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Mayuri Karande
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Abhishek Sarkar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Shubhrakanta Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Antra Chaudhary
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Arvind Sharma
- Department of Neurology, Zydus Hospital, Ahmedabad, 380054, Gujarat, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
11
|
Zhang WJ, Hu DX, Lin SJ, Fang XQ, Ye ZF. Contribution of P2X purinergic receptor in cerebral ischemia injury. Brain Res Bull 2022; 190:42-49. [PMID: 36113681 DOI: 10.1016/j.brainresbull.2022.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/12/2022] [Indexed: 11/02/2022]
Abstract
The development of cerebral ischemia involves brain damage and abnormal changes in brain function, which can cause neurosensory and motor dysfunction, and bring serious consequences to patients. P2X purinergic receptors are expressed in nerve cells and immune cells, and are mainly expressed in microglia. The P2X4 and P2X7 receptors in the P2X purinergic receptors play a significant role in regulating the activity of microglia. Moreover, ATP-P2X purine information transmission is involved in the progression of neurological diseases, including the release of pro-inflammatory factors, driving factors and cytokines after cerebral ischemia injury, inducing inflammation, and aggravating cerebral ischemia injury. P2X receptors activation can mediate the information exchange between microglia and neurons, induce neuronal apoptosis, and aggravate neurological dysfunction after cerebral ischemia. However, inhibiting the activation of P2X receptors, reducing their expression, inhibiting the activation of microglia, and has the effect of protecting nerve function. In this paper, we discussed the relationship between P2X receptors and nervous system function and the role of microglia activation inducing cerebral ischemia injury. Additionally, we explored the potential role of P2X receptors in the progression of cerebral ischemic injury and their potential pharmacological targets for the treatment of cerebral ischemic injury.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Si-Jian Lin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Xiao-Qun Fang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Zhen-Feng Ye
- Department of Urology, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
12
|
Zhang W, Ciorraga M, Mendez P, Retana D, Boumedine-Guignon N, Achón B, Russier M, Debanne D, Garrido JJ. Formin Activity and mDia1 Contribute to Maintain Axon Initial Segment Composition and Structure. Mol Neurobiol 2021; 58:6153-6169. [PMID: 34458961 PMCID: PMC8639558 DOI: 10.1007/s12035-021-02531-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/11/2021] [Indexed: 10/29/2022]
Abstract
The axon initial segment (AIS) is essential for maintaining neuronal polarity, modulating protein transport into the axon, and action potential generation. These functions are supported by a distinctive actin and microtubule cytoskeleton that controls axonal trafficking and maintains a high density of voltage-gated ion channels linked by scaffold proteins to the AIS cytoskeleton. However, our knowledge of the mechanisms and proteins involved in AIS cytoskeleton regulation to maintain or modulate AIS structure is limited. In this context, formins play a significant role in the modulation of actin and microtubules. We show that pharmacological inhibition of formins modifies AIS actin and microtubule characteristics in cultured hippocampal neurons, reducing F-actin density and decreasing microtubule acetylation. Moreover, formin inhibition diminishes sodium channels, ankyrinG and βIV-spectrin AIS density, and AIS length, in cultured neurons and brain slices, accompanied by decreased neuronal excitability. We show that genetic downregulation of the mDia1 formin by interference RNAs also decreases AIS protein density and shortens AIS length. The ankyrinG decrease and AIS shortening observed in pharmacologically inhibited neurons and neuron-expressing mDia1 shRNAs were impaired by HDAC6 downregulation or EB1-GFP expression, known to increase microtubule acetylation or stability. However, actin stabilization only partially prevented AIS shortening without affecting AIS protein density loss. These results suggest that mDia1 maintain AIS composition and length contributing to the stability of AIS microtubules.
Collapse
Affiliation(s)
- Wei Zhang
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Present Address: College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | | | | | | | | | | | - Michaël Russier
- UNIS, INSERM, UMR 1072, Aix-Marseille Université, 13015 Marseille, France
| | - Dominique Debanne
- UNIS, INSERM, UMR 1072, Aix-Marseille Université, 13015 Marseille, France
| | - Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer’s Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
13
|
Hasan D, Shono A, van Kalken CK, van der Spek PJ, Krenning EP, Kotani T. A novel definition and treatment of hyperinflammation in COVID-19 based on purinergic signalling. Purinergic Signal 2021; 18:13-59. [PMID: 34757513 PMCID: PMC8578920 DOI: 10.1007/s11302-021-09814-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/18/2021] [Indexed: 12/15/2022] Open
Abstract
Hyperinflammation plays an important role in severe and critical COVID-19. Using inconsistent criteria, many researchers define hyperinflammation as a form of very severe inflammation with cytokine storm. Therefore, COVID-19 patients are treated with anti-inflammatory drugs. These drugs appear to be less efficacious than expected and are sometimes accompanied by serious adverse effects. SARS-CoV-2 promotes cellular ATP release. Increased levels of extracellular ATP activate the purinergic receptors of the immune cells initiating the physiologic pro-inflammatory immune response. Persisting viral infection drives the ATP release even further leading to the activation of the P2X7 purinergic receptors (P2X7Rs) and a severe yet physiologic inflammation. Disease progression promotes prolonged vigorous activation of the P2X7R causing cell death and uncontrolled ATP release leading to cytokine storm and desensitisation of all other purinergic receptors of the immune cells. This results in immune paralysis with co-infections or secondary infections. We refer to this pathologic condition as hyperinflammation. The readily available and affordable P2X7R antagonist lidocaine can abrogate hyperinflammation and restore the normal immune function. The issue is that the half-maximal effective concentration for P2X7R inhibition of lidocaine is much higher than the maximal tolerable plasma concentration where adverse effects start to develop. To overcome this, we selectively inhibit the P2X7Rs of the immune cells of the lymphatic system inducing clonal expansion of Tregs in local lymph nodes. Subsequently, these Tregs migrate throughout the body exerting anti-inflammatory activities suppressing systemic and (distant) local hyperinflammation. We illustrate this with six critically ill COVID-19 patients treated with lidocaine.
Collapse
Affiliation(s)
| | - Atsuko Shono
- Department of Anaesthesiology and Critical Care Medicine, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | | | - Peter J van der Spek
- Department of Pathology & Clinical Bioinformatics, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE, Rotterdam, The Netherlands
| | | | - Toru Kotani
- Department of Anaesthesiology and Critical Care Medicine, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| |
Collapse
|
14
|
To inhibit or to boost the ATP/P2RX7 pathway to fight cancer-that is the question. Purinergic Signal 2021; 17:619-631. [PMID: 34347213 DOI: 10.1007/s11302-021-09811-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Despite new biological insights and recent therapeutic advances, many tumors remain at baseline during treatments. Therefore, there is an urgent need to find new therapeutic strategies to improve the care of patients with solid tumors. P2RX7 receptor (P2XR7), an ATP-gated ion channel characterized by its ability to form large pore within the cell membrane, is described by most of the investigators as a "chef d'orchestre" of the antitumor immune response. The purpose of this review is to detail the recent information concerning different cellular mechanisms linking P2RX7 to hallmarks of cancer and to discuss different progresses in elucidating how activation of the ATP/P2RX7/NLRP3/IL-18 pathway is a very promising approach to fight cancer progression by increasing antitumor immune responses.
Collapse
|
15
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
16
|
Zhao YF, Tang Y, Illes P. Astrocytic and Oligodendrocytic P2X7 Receptors Determine Neuronal Functions in the CNS. Front Mol Neurosci 2021; 14:641570. [PMID: 33642994 PMCID: PMC7906075 DOI: 10.3389/fnmol.2021.641570] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
P2X7 receptors are members of the ATP-gated cationic channel family with a preferential localization at the microglial cells, the resident macrophages of the brain. However, these receptors are also present at neuroglia (astrocytes, oligodendrocytes) although at a considerably lower density. They mediate necrosis/apoptosis by the release of pro-inflammatory cytokines/chemokines, reactive oxygen species (ROS) as well as the excitotoxic (glio)transmitters glutamate and ATP. Besides mediating cell damage i.e., superimposed upon chronic neurodegenerative processes in Alzheimer’s Disease, Parkinson’s Disease, multiple sclerosis, and amyotrophic lateral sclerosis, they may also participate in neuroglial signaling to neurons under conditions of high ATP concentrations during any other form of neuroinflammation/neurodegeneration. It is a pertinent open question whether P2X7Rs are localized on neurons, or whether only neuroglia/microglia possess this receptor-type causing indirect effects by releasing the above-mentioned signaling molecules. We suggest as based on molecular biology and functional evidence that neurons are devoid of P2X7Rs although the existence of neuronal P2X7Rs cannot be excluded with absolute certainty.
Collapse
Affiliation(s)
- Ya-Fei Zhao
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Tang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,International Collaborative Center on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peter Illes
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,International Collaborative Center on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Villa González M, Vallés-Saiz L, Hernández IH, Avila J, Hernández F, Pérez-Alvarez MJ. Focal cerebral ischemia induces changes in oligodendrocytic tau isoforms in the damaged area. Glia 2020; 68:2471-2485. [PMID: 32515854 DOI: 10.1002/glia.23865] [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: 12/19/2019] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
Abstract
Ischemic stroke is a major cause of death and the first leading cause of long-term disability worldwide. The only therapeutic strategy available to date is reperfusion and not all the patients are suitable for this treatment. Blood flow blockage or reduction leads to considerable brain damage, affecting both gray and white matter. The detrimental effects of ischemia have been studied extensively in the former but not in the latter. Previous reports indicate that preservation of white matter integrity reduces deleterious effect of ischemia on the brain. Oligodendrocytes are sensitive to ischemic damage, however, some reports demonstrate that oligodendrogenesis occurs after ischemia. These glial cells have a complex cytoskeletal network, including tau, that plays a key role to proper myelination. 4R-Tau/3R-Tau, which differ in the presence/absence of Exon 10, are found in oligodendrocytes; but the precise role of each isoform is not understood. Using permanent middle cerebral artery occlusion model and immunofluorescence, we demonstrate that cerebral ischemia induces an increase in 3R-Tau versus 4R-Tau in oligodendrocytes in the damaged area. In addition, cellular distribution of Tau undergoes a change after ischemia, with some oligodendrocytic processes showing positive staining for 3R-Tau. This occurs simultaneously with the amelioration of neurological damage in ischemic rats. We propose that ischemia triggers an endogenous mechanism involving 3R-Tau, that induces colonization of the ischemic damaged area by oligodendrocytes in an attempt to myelinate-injured axons. Understanding the molecular mechanism of this phenomenon could pave the way for the design of therapeutic strategies that exploit glial cells for the treatment of ischemia.
Collapse
Affiliation(s)
- Mario Villa González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Laura Vallés-Saiz
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Ivó H Hernández
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Avila
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Félix Hernández
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
18
|
Abstract
Microglia dynamically interact with neurons influencing the development, structure, and function of neuronal networks. Recent studies suggest microglia may also influence neuronal activity by physically interacting with axonal domains responsible for action potential initiation and propagation. However, the nature of these microglial process interactions is not well understood. Microglial-axonal contacts are present early in development and persist through adulthood, implicating microglial interactions in the regulation of axonal integrity in both the developing and mature central nervous system. Moreover, changes in microglial-axonal contact have been described in disease states such as multiple sclerosis (MS) and traumatic brain injury (TBI). Depending on the disease state, there are increased associations with specific axonal segments. In MS, there is enhanced contact with the axon initial segment and node of Ranvier, while, in TBI, microglia alter interactions with axons at the site of injury, as well as at the axon initial segment. In this article, we review the interactions of microglial processes with axonal segments, analyzing their associations with various axonal domains and how these interactions may differ between MS and TBI. Furthermore, we discuss potential functional consequences and molecular mechanisms of these interactions and how these may differ among various types of microglial-axonal interactions.
Collapse
Affiliation(s)
- Savannah D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Audrey D Lafrenaye
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
19
|
Zhao Y, Wu X, Chen X, Li J, Tian C, Chen J, Xiao C, Zhong G, He S. Calcineurin Signaling Mediates Disruption of the Axon Initial Segment Cytoskeleton after Injury. iScience 2020; 23:100880. [PMID: 32062456 PMCID: PMC7031317 DOI: 10.1016/j.isci.2020.100880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 10/23/2019] [Accepted: 01/29/2020] [Indexed: 11/30/2022] Open
Abstract
The axon initial segment (AIS) cytoskeleton undergoes rapid and irreversible disruption prior to cell death after injury, and loss of AIS integrity can produce profound neurological effects on the nervous system. Here we described a previously unrecognized mechanism for ischemia-induced alterations in AIS integrity. We show that in hippocampal CA1 pyramidal neurons Nav1.6 mostly preserves at the AIS after disruption of the cytoskeleton in a mouse model of middle cerebral artery occlusion. Genetic removal of neurofascin-186 leads to rapid disruption of Nav1.6 following injury, indicating that neurofascin is required for Nav1.6 maintenance at the AIS after cytoskeleton collapse. Importantly, calcineurin inhibition with FK506 fully protects AIS integrity and sufficiently prevents impairments of spatial learning and memory from injury. This study provides evidence that calcineurin activation is primarily involved in initiating disassembly of the AIS cytoskeleton and that maintaining AIS integrity is crucial for therapeutic strategies to facilitate recovery from injury. Ion channels are mostly retained at the AIS after ischemic injury Neurofascin is required for clustering ion channels at the AIS after ischemia Calcineurin inhibition protects AIS structural integrity and function from ischemia Calcineurin inhibition protects cognitive function against impairment by ischemia
Collapse
Affiliation(s)
- Yanan Zhao
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Xuanyuan Wu
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Xin Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Jianan Li
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Cuiping Tian
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Jiangrui Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Xiao
- School of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, KJL-D423, Xuzhou, Jiangsu Province 221004, China
| | - Guisheng Zhong
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China.
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China.
| |
Collapse
|
20
|
Yermakov LM, Griggs RB, Drouet DE, Sugimoto C, Williams MT, Vorhees CV, Susuki K. Impairment of cognitive flexibility in type 2 diabetic db/db mice. Behav Brain Res 2019; 371:111978. [PMID: 31141724 PMCID: PMC6579681 DOI: 10.1016/j.bbr.2019.111978] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/05/2023]
Abstract
Impaired executive function is a major peril for patients with type 2 diabetes, reducing quality of life and ability for diabetes management. Despite the significance of this impairment, few animal models of type 2 diabetes examine domains of executive function such as cognitive flexibility or working memory. Here, we evaluated these executive function domains in db/db mice, an established model of type 2 diabetes, at 10 and 24 weeks of age. The db/db mice showed impaired cognitive flexibility in the Morris water maze reversal phase. However, the db/db mice did not show apparent working memory disturbance in the spatial working memory version of the Morris water maze or in the radial water maze. We also examined axon initial segments (AIS) and nodes of Ranvier, key axonal domains for action potential initiation and propagation. AIS were significantly shortened in medial prefrontal cortex and hippocampus of 26-week-old db/db mice compared with controls, similar to our previous findings in 10-week-old mice. Nodes of Ranvier in corpus callosum, previously shown to be unchanged at 10 weeks, were elongated at 26 weeks, suggesting an important role for this domain in disease progression. Together, the findings help establish db/db mice as a model of impaired cognitive flexibility in type 2 diabetes and advance our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Domenica E Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA.
| |
Collapse
|
21
|
Dynamic Gain Analysis Reveals Encoding Deficiencies in Cortical Neurons That Recover from Hypoxia-Induced Spreading Depolarizations. J Neurosci 2019; 39:7790-7800. [PMID: 31399533 DOI: 10.1523/jneurosci.3147-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 11/21/2022] Open
Abstract
Cortical regions that are damaged by insults, such as ischemia, hypoxia, and trauma, frequently generate spreading depolarization (SD). At the neuronal level, SDs entail complete breakdown of ionic gradients, persisting for seconds to minutes. It is unclear whether these transient events have a more lasting influence on neuronal function. Here, we describe electrophysiological changes in cortical neurons after recovery from hypoxia-induced SD. When examined with standard measures of neuronal excitability several hours after recovery from SD, layer 5 pyramidal neurons in brain slices from mice of either sex appear surprisingly normal. However, we here introduce an additional parameter, dynamic gain, which characterizes the bandwidth of action potential encoding by a neuron, and thereby reflects its potential efficiency in a multineuronal circuit. We find that the ability of neurons that recover from SD to track high-frequency inputs is markedly curtailed; exposure to hypoxia did not have this effect when SD was prevented pharmacologically. Staining for Ankyrin G revealed at least a fourfold decrease in the number of intact axon initial segments in post-SD slices. Since this effect, along with the effect on encoding, was blocked by an inhibitor of the Ca2+-dependent enzyme, calpain, we conclude that both effects were mediated by the SD-induced rise in intracellular Ca2+ Although effects of calpain activation were detected in the axon initial segment, changes in soma-dendritic compartments may also be involved. Whatever the precise molecular mechanism, our findings indicate that in the context of cortical circuit function, effectiveness of neurons that survive SD may be limited.SIGNIFICANCE STATEMENT Spreading depolarization, which commonly accompanies cortical injury, entails transient massive breakdown of neuronal ionic gradients. The function of cortical neurons that recover from hypoxia-induced spreading depolarization is not obviously abnormal when tested for usual measures of neuronal excitability. However, we now demonstrate that they have a reduced bandwidth, reflecting a significant impairment of their ability to precisely encode high-frequency components of their synaptic input in output spike trains. Thus, neurons that recover from spreading depolarizations are less able to function normally as elements in the multineuronal cortical circuitry. These changes are correlated with activation of the calcium-dependent enzyme, calpain.
Collapse
|
22
|
Zhu G, Dai B, Chen Z, He L, Guo J, Dan Y, Liang S, Li G. Effects of chronic lead exposure on the sympathoexcitatory response associated with the P2X7 receptor in rat superior cervical ganglia. Auton Neurosci 2019; 219:33-41. [DOI: 10.1016/j.autneu.2019.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/01/2019] [Accepted: 03/20/2019] [Indexed: 12/23/2022]
|
23
|
Zhang W, Bonadiman A, Ciorraga M, Benitez MJ, Garrido JJ. P2Y1 Purinergic Receptor Modulate Axon Initial Segment Initial Development. Front Cell Neurosci 2019; 13:152. [PMID: 31068791 PMCID: PMC6491782 DOI: 10.3389/fncel.2019.00152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/08/2019] [Indexed: 11/22/2022] Open
Abstract
Morphological and functional polarization of neurons depends on the generation and maintenance of the axon initial segment (AIS). This axonal domain maintains axonal properties but is also the place where the action potential (AP) is generated. All these functions require the AIS, a complex structure that is not fully understood. An integrated structure of voltage-gated ion channels, specific cytoskeleton architecture, as well as, scaffold proteins contributes to these functions. Among them, ankyrinG plays a crucial role to maintain ion channels and membrane proteins. However, it is still elusive how the AIS performs its complex structural and functional regulation. Recent studies reveal that AIS is dynamically regulated in molecular composition, length and location in response to neuronal activity. Some mechanisms acting on AIS plasticity have been uncovered recently, including Ca2+, calpain or calmodulin-mediated modulation, as well as post-translational modifications of cytoskeleton proteins and actin-associated proteins. Neurons are able to respond to different kind of physiological and pathological stimuli from development to maturity by adapting their AIS composition, position and length. This raises the question of which are the neuronal receptors that contribute to the modulation of AIS plasticity. Previous studies have shown that purinergic receptor P2X7 activation is detrimental to AIS maintenance. During initial axonal elongation, P2X7 is coordinated with P2Y1, another purinergic receptor that is essential for proper axon elongation. In this study, we focus on the role of P2Y1 receptor on AIS development and maintenance. Our results show that P2Y1 receptor activity and expression are necessary during AIS initial development, while has no role once AIS maturity is achieved. P2Y1 inhibition or suppression results in a decrease in ankyrinG, βIV-spectrin and voltage-gated sodium channels accumulation that can be rescued by actin stabilization or the modulation of actin-binding proteins at the AIS. Moreover, P2X7 or calpain inhibition also rescues ankyrinG decrease. Hence, a dynamic balance of P2Y1 and P2X7 receptors expression and function during AIS assembly and maturation may represent a fine regulatory mechanism in response to physiological or pathological extracellular purines concentration.
Collapse
Affiliation(s)
- Wei Zhang
- Spanish National Research Council (CSIC), Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Madrid, Spain
| | - Angela Bonadiman
- Spanish National Research Council (CSIC), Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Madrid, Spain
| | - María Ciorraga
- Spanish National Research Council (CSIC), Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Madrid, Spain
| | - María José Benitez
- Spanish National Research Council (CSIC), Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Madrid, Spain.,Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan José Garrido
- Spanish National Research Council (CSIC), Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Madrid, Spain
| |
Collapse
|
24
|
Abstract
Differentiated neurons can undergo cell cycle re-entry during pathological conditions, but it remains largely accepted that M-phase is prohibited in these cells. Here we show that primary neurons at post-synaptogenesis stages of development can enter M-phase. We induced cell cycle re-entry by overexpressing a truncated Cyclin E isoform fused to Cdk2. Cyclin E/Cdk2 expression elicits canonical cell cycle checkpoints, which arrest cell cycle progression and trigger apoptosis. As in mitotic cells, checkpoint abrogation enables cell cycle progression through S and G2-phases into M-phase. Although most neurons enter M-phase, only a small subset undergo cell division. Alternatively, neurons can exit M-phase without cell division and recover the axon initial segment, a structural determinant of neuronal viability. We conclude that neurons and mitotic cells share S, G2 and M-phase regulation.
Collapse
|
25
|
Abstract
The axon initial segment (AIS), the domain responsible for action potential initiation and maintenance of neuronal polarity, is targeted for disruption in a variety of central nervous system pathological insults. Previous work in our laboratory implicates oxidative stress as a potential mediator of structural AIS alterations in two separate mouse models of central nervous system inflammation, as these effects were attenuated following reactive oxygen species scavenging and NADPH oxidase-2 ablation. While these studies suggest a role for oxidative stress in modulation of the AIS, the direct effects of reactive oxygen and nitrogen species (ROS/RNS) on the stability of this domain remain unclear. Here, we demonstrate that oxidative stress, as induced through treatment with 3-morpholinosydnonimine (SIN-1), a spontaneous ROS/RNS generator, drives a reversible loss of AIS protein clustering in primary cortical neurons in vitro. Pharmacological inhibition of both voltage-dependent and intracellular calcium (Ca2+) channels suggests that this mechanism of AIS disruption involves Ca2+ entry specifically through L-type voltage-dependent Ca2+ channels and its release from IP3-gated intracellular stores. Furthermore, ROS/RNS-induced AIS disruption is dependent upon activation of calpain, a Ca2+-activated protease previously shown to drive AIS modulation. Overall, we demonstrate for the first time that oxidative stress, as induced through exogenously applied ROS/RNS, is capable of driving structural alterations in the AIS complex.
Collapse
Affiliation(s)
- Kareem Clark
- 1 Department of Anatomy and Neurobiology, 72054 Virginia Commonwealth University , Richmond, VA, USA.,2 Neuroscience Curriculum, 72054 Virginia Commonwealth University , Richmond, VA, USA
| | - Brooke A Sword
- 3 20125 Hunter Holmes McGuire VA Medical Center , Richmond, VA, USA
| | - Jeffrey L Dupree
- 1 Department of Anatomy and Neurobiology, 72054 Virginia Commonwealth University , Richmond, VA, USA.,3 20125 Hunter Holmes McGuire VA Medical Center , Richmond, VA, USA
| |
Collapse
|
26
|
Schlüter A, Del Turco D, Deller T, Gutzmann A, Schultz C, Engelhardt M. Structural Plasticity of Synaptopodin in the Axon Initial Segment during Visual Cortex Development. Cereb Cortex 2018; 27:4662-4675. [PMID: 28922860 DOI: 10.1093/cercor/bhx208] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
The axon initial segment (AIS) is essential for action potential generation. Recently, the AIS was identified as a site of neuronal plasticity. A subpopulation of AIS in cortical principal neurons contains stacks of endoplasmic reticulum (ER) forming the cisternal organelle (CO). The function of this organelle is poorly understood, but roles in local Ca2+-trafficking and AIS plasticity are discussed. To investigate whether the presence and/or the size of COs are linked to the development and maturation of AIS of cortical neurons, we analyzed the relationship between COs and the AIS during visual cortex development under control and visual deprivation conditions. In wildtype mice, immunolabeling for synaptopodin, ankyrin-G, and ßIV-spectrin were employed to label COs and the AIS, respectively. Dark rearing resulted in an increase in synaptopodin cluster sizes, suggesting a homeostatic function of the CO in this cellular compartment. In line with this observation, synaptopodin-deficient mice lacking the CO showed AIS shortening in the dark. Collectively, these data demonstrate that the CO is an essential part of the AIS machinery required for AIS plasticity during a critical developmental period of the visual cortex.
Collapse
Affiliation(s)
- Annabelle Schlüter
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany.,Department of Neurobiology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Annika Gutzmann
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| | - Christian Schultz
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| |
Collapse
|
27
|
Yermakov LM, Drouet DE, Griggs RB, Elased KM, Susuki K. Type 2 Diabetes Leads to Axon Initial Segment Shortening in db/db Mice. Front Cell Neurosci 2018; 12:146. [PMID: 29937715 PMCID: PMC6002488 DOI: 10.3389/fncel.2018.00146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/14/2018] [Indexed: 01/09/2023] Open
Abstract
Cognitive and mood impairments are common central nervous system complications of type 2 diabetes, although the neuronal mechanism(s) remains elusive. Previous studies focused mainly on neuronal inputs such as altered synaptic plasticity. Axon initial segment (AIS) is a specialized functional domain within neurons that regulates neuronal outputs. Structural changes of AIS have been implicated as a key pathophysiological event in various psychiatric and neurological disorders. Here we evaluated the structural integrity of the AIS in brains of db/db mice, an established animal model of type 2 diabetes associated with cognitive and mood impairments. We assessed the AIS before (5 weeks of age) and after (10 weeks) the development of type 2 diabetes, and after daily exercise treatment of diabetic condition. We found that the development of type 2 diabetes is associated with significant AIS shortening in both medial prefrontal cortex and hippocampus, as evident by immunostaining of the AIS structural protein βIV spectrin. AIS shortening occurs in the absence of altered neuronal and AIS protein levels. We found no change in nodes of Ranvier, another neuronal functional domain sharing a molecular organization similar to the AIS. This is the first study to identify AIS alteration in type 2 diabetes condition. Since AIS shortening is known to lower neuronal excitability, our results may provide a new avenue for understanding and treating cognitive and mood impairments in type 2 diabetes.
Collapse
Affiliation(s)
- Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Domenica E Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Khalid M Elased
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
28
|
The Axon Initial Segment: An Updated Viewpoint. J Neurosci 2018; 38:2135-2145. [PMID: 29378864 DOI: 10.1523/jneurosci.1922-17.2018] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/20/2018] [Accepted: 01/25/2018] [Indexed: 12/20/2022] Open
Abstract
At the base of axons sits a unique compartment called the axon initial segment (AIS). The AIS generates and shapes the action potential before it is propagated along the axon. Neuronal excitability thus depends crucially on the AIS composition and position, and these adapt to developmental and physiological conditions. The AIS also demarcates the boundary between the somatodendritic and axonal compartments. Recent studies have brought insights into the molecular architecture of the AIS and how it regulates protein trafficking. This Viewpoints article summarizes current knowledge about the AIS and highlights future challenges in understanding this key actor of neuronal physiology.
Collapse
|
29
|
Beamer E, Kovács G, Sperlágh B. ATP released from astrocytes modulates action potential threshold and spontaneous excitatory postsynaptic currents in the neonatal rat prefrontal cortex. Brain Res Bull 2017; 135:129-142. [PMID: 29030320 DOI: 10.1016/j.brainresbull.2017.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 01/12/2023]
Abstract
Maternal immune activation during pregnancy is a risk factor for neurodevelopmental disorders, such as schizophrenia; however, a full mechanistic understanding has yet to be established. The activity of a transient cell population, the subplate neurons, is critical for the development of cortical inhibition and functional thalamocortical connections. Sensitivity of these cells to factors released during inflammation, therefore, may offer a link between maternal immune activation and the aberrant cortical development underlying some neuropsychiatric disorders. An elevated extracellular ATP concentration is associated with inflammation and has been shown to have an effect on neuronal activity. Here, we investigated the effect of ATP on the electrophysiological properties of subplate neurons. Exogenous ATP increased the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) at micromolar concentrations. Further, ATP released by astrocytes activated by the PAR-1 agonist, TFLLR-NH2, also increased the amplitude and frequency of sEPSCs in subplate neurons. The electrophysiological properties of subplate neurons recorded from prefrontal cortical (PFC) slices from neonatal rats were also disrupted in a maternal immune activation rat model of schizophrenia, with a suramin-sensitive increase in frequency and amplitude of sEPSCs. An alternative neurodevelopmental rat model of schizophrenia, MAM-E17, which did not rely on maternal immune activation, however, showed no change in subplate neuron activity. Both models were validated with behavioral assays, showing schizophrenia-like endophenotypes in young adulthood. The purinergic modulation of subplate neuron activity offers a potential explanatory link between maternal immune activation and disruptions in cortical development that lead to the emergence of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Edward Beamer
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Kovács
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Beata Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
30
|
Neuronal P2X7 Receptor: Involvement in Neuronal Physiology and Pathology. J Neurosci 2017; 37:7063-7072. [PMID: 28747389 DOI: 10.1523/jneurosci.3104-16.2017] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/28/2022] Open
Abstract
The proposed presence of P2X7 receptor (P2X7R) in neurons has been the source of some contention. Initial studies suggested an absence of P2X7R mRNA in neurons, and the apparent nonspecificity of the antibodies used to identify P2X7R raised further doubts. However, subsequent studies using new pharmacological and biomolecular tools provided conclusive evidence supporting the existence of functional P2X7Rs in neurons. The P2X7 receptor has since been shown to play a leading role in multiple aspects of neuronal physiology, including axonal elongation and branching and neurotransmitter release. P2X7R has also been implicated in neuronal pathologies, in which it may influence neuronal survival. Together, this body of research suggests that P2X7R may constitute an important therapeutic target for a variety of neurological disorders.
Collapse
|
31
|
Reassembly of Excitable Domains after CNS Axon Regeneration. J Neurosci 2017; 36:9148-60. [PMID: 27581456 DOI: 10.1523/jneurosci.1747-16.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/15/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Action potential initiation and propagation in myelinated axons require ion channel clustering at axon initial segments (AIS) and nodes of Ranvier. Disruption of these domains after injury impairs nervous system function. Traditionally, injured CNS axons are considered refractory to regeneration, but some recent approaches challenge this view by showing robust long-distance regeneration. However, whether these approaches allow remyelination and promote the reestablishment of AIS and nodes of Ranvier is unknown. Using mouse optic nerve crush as a model for CNS traumatic injury, we performed a detailed analysis of AIS and node disruption after nerve crush. We found significant disruption of AIS and loss of nodes within days of the crush, and complete loss of nodes 1 week after injury. Genetic deletion of the tumor suppressor phosphatase and tensin homolog (Pten) in retinal ganglion cells (RGCs), coupled with stimulation of RGCs by inflammation and cAMP, dramatically enhanced regeneration. With this treatment, we found significant reestablishment of RGC AIS, remyelination, and even reassembly of nodes in regions proximal, within, and distal to the crush site. Remyelination began near the retina, progressed distally, and was confirmed by electron microscopy. Although axons grew rapidly, remyelination and nodal ion channel clustering was much slower. Finally, genetic deletion of ankyrinG from RGCs to block AIS reassembly did not affect axon regeneration, indicating that preservation of neuronal polarity is not required for axon regeneration. Together, our results demonstrate, for the first time, that regenerating CNS axons can be remyelinated and reassemble new AIS and nodes of Ranvier. SIGNIFICANCE STATEMENT We show, for the first time, that regenerated CNS axons have the capacity to both remyelinate and reassemble the axon initial segments and nodes of Ranvier necessary for rapid and efficient action potential propagation.
Collapse
|
32
|
Jamann N, Jordan M, Engelhardt M. Activity-dependent axonal plasticity in sensory systems. Neuroscience 2017; 368:268-282. [PMID: 28739523 DOI: 10.1016/j.neuroscience.2017.07.035] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/23/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022]
Abstract
The rodent whisker-to-barrel cortex pathway is a classic model to study the effects of sensory experience and deprivation on neuronal circuit formation, not only during development but also in the adult. Decades of research have produced a vast body of evidence highlighting the fundamental role of neuronal activity (spontaneous and/or sensory-evoked) for circuit formation and function. In this context, it has become clear that neuronal adaptation and plasticity is not just a function of the neonatal brain, but persists into adulthood, especially after experience-driven modulation of network status. Mechanisms for structural remodeling of the somatodendritic or axonal domain include microscale alterations of neurites or synapses. At the same time, functional alterations at the nanoscale such as expression or activation changes of channels and receptors contribute to the modulation of intrinsic excitability or input-output relationships. However, it remains elusive how these forms of structural and functional plasticity come together to shape neuronal network formation and function. While specifically somatodendritic plasticity has been studied in great detail, the role of axonal plasticity, (e.g. at presynaptic boutons, branches or axonal microdomains), is rather poorly understood. Therefore, this review will only briefly highlight somatodendritic plasticity and instead focus on axonal plasticity. We discuss (i) the role of spontaneous and sensory-evoked plasticity during critical periods, (ii) the assembly of axonal presynaptic sites, (iii) axonal plasticity in the mature brain under baseline and sensory manipulation conditions, and finally (iv) plasticity of electrogenic axonal microdomains, namely the axon initial segment, during development and in the mature CNS.
Collapse
Affiliation(s)
- Nora Jamann
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany
| | - Merryn Jordan
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany.
| |
Collapse
|
33
|
Benusa SD, George NM, Sword BA, DeVries GH, Dupree JL. Acute neuroinflammation induces AIS structural plasticity in a NOX2-dependent manner. J Neuroinflammation 2017; 14:116. [PMID: 28595650 PMCID: PMC5465457 DOI: 10.1186/s12974-017-0889-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/25/2017] [Indexed: 02/08/2023] Open
Abstract
Background Chronic microglia-mediated inflammation and oxidative stress are well-characterized underlying factors in neurodegenerative disease, whereby reactive inflammatory microglia enhance ROS production and impact neuronal integrity. Recently, it has been shown that during chronic inflammation, neuronal integrity is compromised through targeted disruption of the axon initial segment (AIS), the axonal domain critical for action potential initiation. AIS disruption was associated with contact by reactive inflammatory microglia which wrap around the AIS, increasing association with disease progression. While it is clear that chronic microglial inflammation and enhanced ROS production impact neuronal integrity, little is known about how acute microglial inflammation influences AIS stability. Here, we demonstrate that acute neuroinflammation induces AIS structural plasticity in a ROS-mediated and calpain-dependent manner. Methods C57BL/6J and NOX2−/− mice were given a single injection of lipopolysaccharide (LPS; 5 mg/kg) or vehicle (0.9% saline, 10 mL/kg) and analyzed at 6 h–2 weeks post-injection. Anti-inflammatory Didox (250 mg/kg) or vehicle (0.9% saline, 10 mL/kg) was administered beginning 24 h post-LPS injection and continued for 5 days; animals were analyzed 1 week post-injection. Microglial inflammation was assessed using immunohistochemistry (IHC) and RT-qPCR, and AIS integrity was quantitatively analyzed using ankyrinG immunolabeling. Data were statistically compared by one-way or two-way ANOVA where mean differences were significant as assessed using Tukey’s post hoc analysis. Results LPS-induced neuroinflammation, characterized by enhanced microglial inflammation and increased expression of ROS-producing enzymes, altered AIS protein clustering. Importantly, inflammation-induced AIS changes were reversed following resolution of microglial inflammation. Modulation of the inflammatory response using anti-inflammatory Didox, even after significant AIS disruption occurred, increased the rate of AIS recovery. qPCR and IHC analysis revealed that expression of microglial NOX2, a ROS-producing enzyme, was significantly increased correlating with AIS disruption. Furthermore, ablation of NOX2 prevented inflammation-induced AIS plasticity, suggesting that ROS drive AIS structural plasticity. Conclusions In the presence of acute microglial inflammation, the AIS undergoes an adaptive change that is capable of spontaneous recovery. Moreover, recovery can be therapeutically accelerated. Together, these findings underscore the dynamic capabilities of this domain in the presence of a pathological insult and provide evidence that the AIS is a viable therapeutic target.
Collapse
Affiliation(s)
- S D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Curriculum, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - N M George
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Graduate Program, University of Colorado, Denver, CO, 80204, USA
| | - B A Sword
- Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - G H DeVries
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - J L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA. .,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA.
| |
Collapse
|
34
|
Guo Y, Su ZJ, Chen YK, Chai Z. Brain-derived neurotrophic factor/neurotrophin 3 regulate axon initial segment location and affect neuronal excitability in cultured hippocampal neurons. J Neurochem 2017; 142:260-271. [DOI: 10.1111/jnc.14050] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/07/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Yu Guo
- State Key Laboratory of Membrane Biology; College of Life Sciences; Peking University; Beijing China
| | - Zi-jun Su
- State Key Laboratory of Membrane Biology; College of Life Sciences; Peking University; Beijing China
| | - Yi-kun Chen
- State Key Laboratory of Membrane Biology; College of Life Sciences; Peking University; Beijing China
| | - Zhen Chai
- State Key Laboratory of Membrane Biology; College of Life Sciences; Peking University; Beijing China
| |
Collapse
|
35
|
The role of axonal Kv1 channels in CA3 pyramidal cell excitability. Sci Rep 2017; 7:315. [PMID: 28331203 PMCID: PMC5428268 DOI: 10.1038/s41598-017-00388-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 02/22/2017] [Indexed: 01/24/2023] Open
Abstract
Axonal ion channels control spike initiation and propagation along the axon and determine action potential waveform. We show here that functional suppression of axonal Kv1 channels with local puff of dendrotoxin (DTx), laser or mechanical axotomy significantly increased excitability measured in the cell body. Importantly, the functional effect of DTx puffing or axotomy was not limited to the axon initial segment but was also seen on axon collaterals. In contrast, no effects were observed when DTx was puffed on single apical dendrites or after single dendrotomy. A simple model with Kv1 located in the axon reproduced the experimental observations and showed that the distance at which the effects of axon collateral cuts are seen depends on the axon space constant. In conclusion, Kv1 channels located in the axon proper greatly participate in intrinsic excitability of CA3 pyramidal neurons. This finding stresses the importance of the axonal compartment in the regulation of intrinsic neuronal excitability.
Collapse
|
36
|
NF-κB regulates neuronal ankyrin-G via a negative feedback loop. Sci Rep 2017; 7:42006. [PMID: 28181483 PMCID: PMC5299403 DOI: 10.1038/srep42006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
The axon initial segment (AIS) is a neuronal compartment defined by ankyrin-G expression. We here demonstrate that the IKK-complex co-localizes and interacts with the cytoskeletal anchor protein ankyrin-G in immunoprecipitation and proximity-ligation experiments in cortical neurons. Overexpression of the 270 kDa variant of ankyrin-G suppressed, while gene-silencing of ankyrin-G expression increased nuclear factor-κB (NF-κB) activity in primary neurons, suggesting that ankyrin-G sequesters the transcription factor in the AIS. We also found that p65 bound to the ank3 (ankyrin-G) promoter sequence in chromatin immunoprecipitation analyses thereby increasing ank3 expression and ankyrin-G levels at the AIS. Gene-silencing of p65 or ankyrin-G overexpression suppressed ank3 reporter activity. Collectively these data demonstrate that p65/NF-κB controls ankyrin-G levels via a negative feedback loop, thereby linking NF-κB signaling with neuronal polarity and axonal plasticity.
Collapse
|
37
|
Beamer E, Fischer W, Engel T. The ATP-Gated P2X7 Receptor As a Target for the Treatment of Drug-Resistant Epilepsy. Front Neurosci 2017; 11:21. [PMID: 28210205 PMCID: PMC5288361 DOI: 10.3389/fnins.2017.00021] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
Despite the progress made in the development of new antiepileptic drugs (AEDs), the biggest challenges that epilepsy presents to drug development have remained unchanged for the last 80 years: finding a treatment with potential for modifying disease progression and reducing the percentage of patients resistant to all pharmacological interventions. The mechanism of action of the majority of AEDs is based on blocking Na+ and/or Ca2+ channels, promotion of GABA or inhibition of glutamate signaling. In order for further progress to be made, however, a fuller picture of epilepsy will need to be considered, including changes to blood–brain barrier permeability, synaptic plasticity, network reorganization, and gliosis. In particular, brain inflammation has attracted much attention over recent years. Emerging evidence demonstrates a causal role for brain inflammation in lowering seizure thresholds and driving epileptogenesis. Consistent with this, intervening in pro-inflammatory cascades has shown promise in animal models of epilepsy, with clinical trials of anti-inflammatory agents already underway. The ATP-gated purinergic P2X7 receptor (P2X7) has been proposed as a novel drug target for a host of neurological conditions, including epilepsy. Constitutive expression of P2X7 in the CNS is mainly on microglia, but neuronal and astroglial expression has also been suggested. Its function as a gatekeeper of inflammation is most clearly understood, however, it also plays a number of other important roles pertinent to icto- and epileptogenesis: depolarization of the cell membrane, release of macromolecules, induction of apoptosis and synaptic reorganization. Changes in P2X7 expression have been reported following prolonged seizures (status epilepticus) and during chronic epilepsy in both experimental models and patients. While much of the early work focused on the study of P2X7 during status epilepticus, there is now mounting data showing involvement of this receptor during epilepsy. The present short review will discuss the most recent findings concerning P2X7 expression and function during epilepsy and the clinical potential for P2X7 antagonists as novel AEDs.
Collapse
Affiliation(s)
- Edward Beamer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland Dublin, Ireland
| | - Wolfgang Fischer
- Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig Leipzig, Germany
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland Dublin, Ireland
| |
Collapse
|
38
|
Tapia M, Dominguez A, Zhang W, Del Puerto A, Ciorraga M, Benitez MJ, Guaza C, Garrido JJ. Cannabinoid Receptors Modulate Neuronal Morphology and AnkyrinG Density at the Axon Initial Segment. Front Cell Neurosci 2017; 11:5. [PMID: 28179879 PMCID: PMC5263140 DOI: 10.3389/fncel.2017.00005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/10/2017] [Indexed: 12/27/2022] Open
Abstract
Neuronal polarization underlies the ability of neurons to integrate and transmit information. This process begins early in development with axon outgrowth, followed by dendritic growth and subsequent maturation. In between these two steps, the axon initial segment (AIS), a subcellular domain crucial for generating action potentials (APs) and maintaining the morphological and functional polarization, starts to develop. However, the cellular/molecular mechanisms and receptors involved in AIS initial development and maturation are mostly unknown. In this study, we have focused on the role of the type-1 cannabinoid receptor (CB1R), a highly abundant G-protein coupled receptor (GPCR) in the nervous system largely involved in different phases of neuronal development and differentiation. Although CB1R activity modulation has been related to changes in axons or dendrites, its possible role as a modulator of AIS development has not been yet explored. Here we analyzed the potential role of CB1R on neuronal morphology and AIS development using pharmacological and RNA interference approaches in cultured hippocampal neurons. CB1R inhibition, at a very early developmental stage, has no effect on axonal growth, yet CB1R activation can promote it. By contrast, subsequent dendritic growth is impaired by CB1R inhibition, which also reduces ankyrinG density at the AIS. Moreover, our data show a significant correlation between early dendritic growth and ankyrinG density. However, CB1R inhibition in later developmental stages after dendrites are formed only reduces ankyrinG accumulation at the AIS. In conclusion, our data suggest that neuronal CB1R basal activity plays a role in initial development of dendrites and indirectly in AIS proteins accumulation. Based on the lack of CB1R expression at the AIS, we hypothesize that CB1R mediated modulation of dendritic arbor size during early development indirectly determines the accumulation of ankyrinG and AIS development. Further studies will be necessary to determine which CB1R-dependent mechanisms can coordinate these two domains, and what may be the impact of these early developmental changes once neurons mature and are embedded in a functional brain network.
Collapse
Affiliation(s)
- Mónica Tapia
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - Ana Dominguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - Wei Zhang
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - Ana Del Puerto
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - María Ciorraga
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - María José Benitez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)Madrid, Spain; Department of Quimica Fisica Aplicada, Universidad Autónoma de MadridMadrid, Spain
| | - Carmen Guaza
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| | - Juan José Garrido
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) Madrid, Spain
| |
Collapse
|
39
|
Dumitrescu AS, Evans MD, Grubb MS. Evaluating Tools for Live Imaging of Structural Plasticity at the Axon Initial Segment. Front Cell Neurosci 2016; 10:268. [PMID: 27932952 PMCID: PMC5120105 DOI: 10.3389/fncel.2016.00268] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/04/2016] [Indexed: 11/20/2022] Open
Abstract
The axon initial segment (AIS) is a specialized neuronal compartment involved in the maintenance of axo-dendritic polarity and in the generation of action potentials. It is also a site of significant structural plasticity—manipulations of neuronal activity in vitro and in vivo can produce changes in AIS position and/or size that are associated with alterations in intrinsic excitability. However, to date all activity-dependent AIS changes have been observed in experiments carried out on fixed samples, offering only a snapshot, population-wide view of this form of plasticity. To extend these findings by following morphological changes at the AIS of individual neurons requires reliable means of labeling the structure in live preparations. Here, we assessed five different immunofluorescence-based and genetically-encoded tools for live-labeling the AIS of dentate granule cells (DGCs) in dissociated hippocampal cultures. We found that an antibody targeting the extracellular domain of neurofascin provided accurate live label of AIS structure at baseline, but could not follow rapid activity-dependent changes in AIS length. Three different fusion constructs of GFP with full-length AIS proteins also proved unsuitable: while neurofascin-186-GFP and NaVβ4-GFP did not localize to the AIS in our experimental conditions, overexpressing 270kDa-AnkyrinG-GFP produced abnormally elongated AISs in mature neurons. In contrast, a genetically-encoded construct consisting of a voltage-gated sodium channel intracellular domain fused to yellow fluorescent protein (YFP-NaVII–III) fulfilled all of our criteria for successful live AIS label: this construct specifically localized to the AIS, accurately revealed plastic changes at the structure within hours, and, crucially, did not alter normal cell firing properties. We therefore recommend this probe for future studies of live AIS plasticity in vitro and in vivo.
Collapse
Affiliation(s)
- Adna S Dumitrescu
- Centre for Developmental Neurobiology, King's College London London, UK
| | - Mark D Evans
- Centre for Developmental Neurobiology, King's College London London, UK
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, King's College London London, UK
| |
Collapse
|
40
|
Abstract
Myelinated axons are divided into polarized subdomains including axon initial segments and nodes of Ranvier. These domains initiate and propagate action potentials and regulate the trafficking and localization of somatodendritic and axonal proteins. Formation of axon initial segments and nodes of Ranvier depends on intrinsic (neuronal) and extrinsic (glial) interactions. Several levels of redundancy in both mechanisms and molecules also exist to ensure efficient node formation. Furthermore, the establishment of polarized domains at and near nodes of Ranvier reflects the intrinsic polarity of the myelinating glia responsible for node assembly. Here, we discuss the various polarized domains of myelinated axons, how they are established by both intrinsic and extrinsic interactions, and the polarity of myelinating glia.
Collapse
Affiliation(s)
- Daniel R Zollinger
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| | - Kelli L Baalman
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| |
Collapse
|
41
|
Petersen AV, Cotel F, Perrier JF. Plasticity of the Axon Initial Segment: Fast and Slow Processes with Multiple Functional Roles. Neuroscientist 2016; 23:364-373. [PMID: 27143656 DOI: 10.1177/1073858416648311] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The axon initial segment (AIS) is a key neuronal compartment because it is responsible for action potential initiation. The local density of Na+ channels, the biophysical properties of K+ channels, as well as the length and diameter of the AIS determine the spiking of neurons. These parameters undergo important modifications during development. The development of the AIS is governed by intrinsic mechanisms. In addition, surrounding neuronal networks modify its maturation. As a result, neurons get tuned to particular physiological functions. Neuronal activity also influences the morphology of the mature AIS. When excitatory neurons are hyperactive, their AIS undergo structural changes that decrease their excitability and thereby maintain the activity within a given range. These slow homeostatic regulatory mechanisms occur on a time scale of hours or days. In contrast, the activation of metabotropic receptors modulates the properties of ion channels expressed at the AIS within seconds and consequently produces fast adjustments of neuronal excitability. Recent results suggest that this plasticity plays important roles in physiological functions as diverse as memory formation, hearing, and motor control.
Collapse
Affiliation(s)
- Anders Victor Petersen
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Florence Cotel
- 2 Queensland Brain Institute, University of Queensland, St Lucia, Australia
| | - Jean-François Perrier
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
|
43
|
Stoler O, Fleidervish IA. Functional implications of axon initial segment cytoskeletal disruption in stroke. Acta Pharmacol Sin 2016; 37:75-81. [PMID: 26687934 DOI: 10.1038/aps.2015.107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Axon initial segment (AIS) is the proximal part of the axon, which is not covered with a myelin sheath and possesses a distinctive, specialized assembly of voltage-gated ion channels and associated proteins. AIS plays critical roles in synaptic integration and action potential generation in central neurons. Recent evidence shows that stroke causes rapid, irreversible calpain-mediated proteolysis of the AIS cytoskeleton of neurons surrounding the ischemic necrotic core. A better understanding of the molecular mechanisms underlying this "non-lethal" neuronal damage might provide new therapeutic strategies for improving stroke outcome. Here, we present a brief overview of the structure and function of the AIS. We then discuss possible mechanisms underlying stroke-induced AIS damage, including the roles of calpains and possible sources of Ca(2+) ions, which are necessary for the activation of calpains. Finally, we discuss the potential functional implications of the loss of the AIS cytoskeleton and ion channel clusters for neuronal excitability.
Collapse
|
44
|
Mittal R, Chan B, Grati M, Mittal J, Patel K, Debs LH, Patel AP, Yan D, Chapagain P, Liu XZ. Molecular Structure and Regulation of P2X Receptors With a Special Emphasis on the Role of P2X2 in the Auditory System. J Cell Physiol 2015; 231:1656-70. [PMID: 26627116 DOI: 10.1002/jcp.25274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/01/2015] [Indexed: 12/23/2022]
Abstract
The P2X purinergic receptors are cation-selective channels gated by extracellular adenosine 5'-triphosphate (ATP). These purinergic receptors are found in virtually all mammalian cell types and facilitate a number of important physiological processes. Within the past few years, the characterization of crystal structures of the zebrafish P2X4 receptor in its closed and open states has provided critical insights into the mechanisms of ligand binding and channel activation. Understanding of this gating mechanism has facilitated to design and interpret new modeling and structure-function experiments to better elucidate how different agonists and antagonists can affect the receptor with differing levels of potency. This review summarizes the current knowledge on the structure, activation, allosteric modulators, function, and location of the different P2X receptors. Moreover, an emphasis on the P2X2 receptors has been placed in respect to its role in the auditory system. In particular, the discovery of three missense mutations in P2X2 receptors could become important areas of study in the field of gene therapy to treat progressive and noise-induced hearing loss. J. Cell. Physiol. 231: 1656-1670, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Brandon Chan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Kunal Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Luca H Debs
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Amit P Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, Florida.,Biomolecular Science Institute, Florida International University, Miami, Florida
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biochemistry, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
45
|
Post-translational allosteric activation of the P2X7 receptor through glycosaminoglycan chains of CD44 proteoglycans. Cell Death Discov 2015; 1:15005. [PMID: 27551441 PMCID: PMC4979527 DOI: 10.1038/cddiscovery.2015.5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/21/2022] Open
Abstract
Here, we present evidence for the positive allosteric modulation of the P2X7 receptor through glycosaminoglycans (GAGs) in CHO (cell line derived from the ovary of the Chinese hamster) cells. The marked potentiation of P2X7 activity through GAGs in the presence of non-saturating agonists concentrations was evident with the endogenous expression of the receptor in CHO cells. The presence of GAGs on the surface of CHO cells greatly increased the sensitivity to adenosine 5'-triphosphate and changed the main P2X7 receptor kinetic parameters EC50, Hill coefficient and E max. GAGs decreased the allosteric inhibition of P2X7 receptor through Mg(2+). GAGs activated P2X7 receptor-mediated cytoplasmic Ca(2+) influx and pore formation. Consequently, wild-type CHO-K1 cells were 2.5-fold more sensitive to cell death induced through P2X7 agonists than mutant CHO-745 cells defective in GAGs biosynthesis. In the present study, we provide the first evidence that the P2X7 receptor interacts with CD44 on the CHO-K1 cell surface. Thus, these data demonstrated that GAGs positively modulate the P2X7 receptor, and sCD44 is a part of a regulatory positive feedback loop linking P2X7 receptor activation for the intracellular response mediated through P2X7 receptor stimulation.
Collapse
|
46
|
Miras-Portugal MT, Gomez-Villafuertes R, Gualix J, Diaz-Hernandez JI, Artalejo AR, Ortega F, Delicado EG, Perez-Sen R. Nucleotides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:243-54. [PMID: 26359530 DOI: 10.1016/j.neuropharm.2015.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 02/03/2023]
Abstract
Brain injury generates the release of a multitude of factors including extracellular nucleotides, which exhibit bi-functional properties and contribute to both detrimental actions in the acute phase and also protective and reparative actions in the later recovery phase to allow neuroregeneration. A promising strategy toward restoration of neuronal function is based on activation of endogenous adult neural stem/progenitor cells. The implication of purinergic signaling in stem cell biology, including regulation of proliferation, differentiation, and cell death has become evident in the last decade. In this regard, current strategies of acute transplantation of ependymal stem/progenitor cells after spinal cord injury restore altered expression of P2X4 and P2X7 receptors and improve functional locomotor recovery. The expression of both receptors is transcriptionally regulated by Sp1 factor, which plays a key role in the startup of the transcription machinery to induce regeneration-associated genes expression. Finally, general signaling pathways triggered by nucleotide receptors in neuronal populations converge on several intracellular kinases, such as PI3K/Akt, GSK3 and ERK1,2, as well as the Nrf-2/heme oxigenase-1 axis, which specifically link them to neuroprotection. In this regard, regulation of dual specificity protein phosphatases can become novel mechanism of actions for nucleotide receptors that associate them to cell homeostasis regulation. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- M Teresa Miras-Portugal
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Rosa Gomez-Villafuertes
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain.
| | - Javier Gualix
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Juan Ignacio Diaz-Hernandez
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Antonio R Artalejo
- Department of Toxicology and Pharmacology, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Felipe Ortega
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Esmerilda G Delicado
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Raquel Perez-Sen
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| |
Collapse
|
47
|
Sperlágh B, Illes P. P2X7 receptor: an emerging target in central nervous system diseases. Trends Pharmacol Sci 2014; 35:537-47. [PMID: 25223574 DOI: 10.1016/j.tips.2014.08.002] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/07/2014] [Accepted: 08/14/2014] [Indexed: 12/19/2022]
Abstract
The ATP-sensitive homomeric P2X7 receptor (P2X7R) has received particular attention as a potential drug target because of its widespread involvement in inflammatory diseases as a key regulatory element of the inflammasome complex. However, it has only recently become evident that P2X7Rs also play a pivotal role in central nervous system (CNS) pathology. There is an explosion of data indicating that genetic deletion and pharmacological blockade of P2X7Rs alter responsiveness in animal models of neurological disorders, such as stroke, neurotrauma, epilepsy, neuropathic pain, multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, and Huntington's disease. Moreover, recent studies suggest that P2X7Rs regulate the pathophysiology of psychiatric disorders, including mood disorders, implicating P2X7Rs as drug targets in a variety of CNS pathology.
Collapse
Affiliation(s)
- Beáta Sperlágh
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1450 Budapest, Hungary.
| | - Peter Illes
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, D-04107 Leipzig, Germany
| |
Collapse
|