1
|
Hadadi M, Farazi MM, Mehrabani M, Tashakori-Miyanroudi M, Behroozi Z. Curcumin reduces pain after spinal cord injury in rats by decreasing oxidative stress and increasing GABAA receptor and GAD65 levels. Sci Rep 2025; 15:12910. [PMID: 40234536 PMCID: PMC12000590 DOI: 10.1038/s41598-025-93726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
About 70% of spinal cord injury (SCI) patients experience neuropathic pain (NP), posing an important medical challenge. Painkillers are used to manage pain today and often have undesirable side effects. Curcumin's antioxidant properties may help alleviate NP following SCI (NP-SCI). We decided to study curcumin's effects on NP-SCI for the first time. Male Wistar rats were divided into five groups (n = 8): Control (no injury/no treatment), Sham (laminectomy), SCI (spinal cord compression at T11-T12 using a clip), Curcumin100 and Curcumin200 (Curcumin at 100 and 200 mg/kg administered 30 min after SCI for 10days). Motor function, allodynia, and hyperalgesia were assessed using the BBB scale, acetone, and tail-flick until six weeks after SCI. H&E staining for assaying cavity, western blot for measuring GAD65 and GABA-A receptors, and biochemical kits for assaying SOD, catalase, total antioxidant capacity, and MDA were used. PRISM software analyzed data. Results showed significant improvements in motion, allodynia, hyperalgesia, cavity, urinary retention (P < 0.0001), and weight in curcumin treatments. There was also a reduction in MDA, with increasing GABA-A receptors, GAD65, and antioxidants in them. Findings suggest curcumin may provide good analgesic effects through its antioxidants, and extensive studies are needed to confirm it as a treatment for NP-SCI in the clinic. Keyboards: spinal cord injury, curcumin, antioxidant, pain, GABA, GAD65 enzyme.
Collapse
Affiliation(s)
- Maryam Hadadi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Mojtaba Farazi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahsa Tashakori-Miyanroudi
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Schmitt-Ulms G, Wang X, Watts J, Booth S, Wille H, Zhao W. A unified model for the origins of spongiform degeneration and other neuropathological features in prion diseases. ARXIV 2025:arXiv:2412.16678v2. [PMID: 39876936 PMCID: PMC11774453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Decades after their initial observation in prion-infected brain tissues, the identities of virus-like dense particles, varicose tubules, and oval bodies containing parallel bands and fibrils have remained elusive. Our recent work revealed that a phenotype of dilation of the endoplasmic reticulum (ER), most notable for the perinuclear space (PNS), contributes to spongiform degeneration. To assess the significance of this phenotype for the etiology of prion diseases, we explored whether it can be functionally linked to other neuropathological hallmarks observed in these diseases, as this would indicate it to be a central event. Having surveyed the neuropathological record and other distant literature niches, we propose a model in which pathogenic forms of the prion protein poison raft domains, including essential Na+, K+-ATPases (NKAs) embedded within them, thereby triggering an ER-centered cellular rescue program coordinated by the unfolded protein response (UPR). The execution of this program stalls general protein synthesis, causing the deterioration of synaptic spines. As the disease progresses, cells selectively increase sterol biosynthesis, along with ribosome and ER biogenesis. These adaptive rescue attempts cause morphological changes to the ER which manifest as ER dilation or ER hypertrophy in a manner that is influenced by Ca2+ influx into the cell. The nuclear-to-cytoplasmic transport of mRNAs and tRNAs interrupts in late stage disease, thereby depriving ribosomes of supplies and inducing them to aggregate into a paracrystalline form. In support of this model, we share previously reported data, whose features are consistent with the interpretation that 1) the phenotype of ER dilation is observed in major prion diseases, 2) varicose tubules and oval bodies represent ER hypertrophy, and 3) virus-like dense particles are paracrystalline aggregates of inactive ribosomes.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Xinzhu Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Joel Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Stephanie Booth
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Holger Wille
- Department of Biochemistry, University of Alberta, Edmonton, Canada
- Centre for Prions and Protein Folding Diseases, University of Edmonton, Edmonton, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
3
|
Duarte JMN. Challenges of Investigating Compartmentalized Brain Energy Metabolism Using Nuclear Magnetic Resonance Spectroscopy in vivo. Neurochem Res 2025; 50:73. [PMID: 39754627 PMCID: PMC11700056 DOI: 10.1007/s11064-024-04324-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Brain function requires continuous energy supply. Thus, unraveling brain metabolic regulation is critical not only for our basic understanding of overall brain function, but also for the cellular basis of functional neuroimaging techniques. While it is known that brain energy metabolism is exquisitely compartmentalized between astrocytes and neurons, the metabolic and neuro-energetic basis of brain activity is far from fully understood. 1H nuclear magnetic resonance (NMR) spectroscopy has been widely used to detect variations in metabolite levels, including glutamate and GABA, while 13C NMR spectroscopy has been employed to study metabolic compartmentation and to determine metabolic rates coupled brain activity, focusing mainly on the component corresponding to excitatory glutamatergic neurotransmission. The rates of oxidative metabolism in neurons and astrocytes are both associated with the rate of the glutamate-glutamine cycle between neurons and astrocytes. However, any possible correlation between energy metabolism pathways and the inhibitory GABAergic neurotransmission rate in the living brain remains to be experimentally demonstrated. That is due to low GABA levels, and the consequent challenge of determining GABAergic rates in a non-invasive manner. This brief review surveys the state-of-the-art analyses of energy metabolism in neurons and astrocytes contributing to glutamate and GABA synthesis using 13C NMR spectroscopy in vivo, and identifies limitations that need to be overcome in future studies.
Collapse
Affiliation(s)
- João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
4
|
Liu XX, Ke XY, Jiang C, Bo LW, Sun N, Li LL, Qin SQ, He JC, Ren JL, Wu QQ, Li SZ, Yang JL, Yu LL, Lu QY, Liu LZ, Li WY, Xian XH, Zhang LN. Na +-K +-ATPase/GLT-1 interaction participates in EGCG protection against cerebral ischemia-reperfusion injury in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156349. [PMID: 39765036 DOI: 10.1016/j.phymed.2024.156349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/21/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND In China, stroke is the primary cause of adult death and disability. Because of the increased rate of blood vessel reperfusion, it is important to prevent cerebral ischemia-reperfusion injury, in which glutamate (Glu) excitotoxicity plays a critical role. The most important Glu transporter, GLT-1, is essential for the regulation of Glu, which is dependent on Na+-K+-ATPase (NKA)-induced ion concentration gradient differences. EGCG, a substance found in tea polyphenols, can reduce infarct areas in ischemia-reperfusion models, reduce stroke incidence, and prolong life in which NKA is involved. PURPOSE In this study, we investigated the potential of EGCG in protecting against cerebral ischemia-reperfusion injury by regulating the interaction between NKA and GLT-1. STUDY DESIGN This study was designed to investigate the protective effects of EGCG against cerebral ischemia-reperfusion injury by modulating the interaction between NKA and GLT-1, utilizing both the rat middle cerebral artery occlusion/reperfusion (MCAO/R) model and the oxygen-glucose deprivation/reoxygenation (OGD/R) model in co-cultures of rat hippocampal neurons and astrocytes. METHODS The neuronal survival rate was assessed using CCK8, and the cerebral infarction area and neurological function were determined by TTC staining and neurological deficit scores. NKA activity was measured using an inorganic phosphorous detection method, and NKA and GLT-1 expression was detected using western blotting. The interaction between NKAα2 and GLT-1 was identified by co-immunoprecipitation (CoIP) assay, laser confocal microscopy, and Imaris 3D confocal rendering technology. An adenovirus vector with overexpression of NKAα2 was constructed, packaged, and injected into the rat lateral ventricle. Neurological function and the cerebral infarction area were identified, and the interaction between NKAα2 and GLT-1 was identified using CoIP assay. RESULTS EGCG reduced the infarction area and neurological deficit scores, restored NKA activity, alleviated the decrease in membrane NKAα2 and GLT-1 expression, and relieved the uncoupling of NKAα2 and GLT-1 in the hippocampal CA1 after rat MCAO/R injury. By promoting the coupling of NKAα2 and GLT-1 in rat MCAO/R models, overexpression of NKAα2 reduced the cerebral infarction area and neurological impairment scores. CONCLUSION EGCG improved cerebral ischemia-reperfusion injury by restoring NKA activity and increasing membrane GLT-1 expression due to NKA-GLT-1 interaction. For the first time, our findings demonstrate the critical role that NKA and GLT-1 colocalization plays in cerebral ischemia-reperfusion damage. Our findings provide new strategic directions for the pathogenesis and prevention of thrombolytic injury in the clinical treatment of stroke, while also serving as a basis for further development and utilization of EGCG.
Collapse
Affiliation(s)
- Xin-Xin Liu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Department of Science and Education, Xingtai People's Hospital, 818 Xiangdu North Road, Xingtai 054001, China
| | - Xue-Ying Ke
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Chen Jiang
- Forensic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Ling-Wei Bo
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Nan Sun
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Lin-Lin Li
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Shi-Qi Qin
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Jin-Chen He
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Jia-Lin Ren
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Qian-Qian Wu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Shuai-Zhen Li
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Jia-Lei Yang
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Lan-Ling Yu
- Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Qi-Yong Lu
- Department of Neurosurgery, Hengshui Fifth People's Hospital, 1638 Shengli West Road, Hengshui 053010, China
| | - Li-Zhe Liu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Wen-Ya Li
- Department of Physiology, Hebei University of Chinese Medicine, 3 Xingyuan Road, Shijiazhuang 050200, China.
| | - Xiao-Hui Xian
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China.
| | - Li-Nan Zhang
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China.
| |
Collapse
|
5
|
Zalaquett NG, Salameh E, Kim JM, Ghanbarian E, Tawk K, Abouzari M. The Dawn and Advancement of the Knowledge of the Genetics of Migraine. J Clin Med 2024; 13:2701. [PMID: 38731230 PMCID: PMC11084801 DOI: 10.3390/jcm13092701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Migraine is a prevalent episodic brain disorder known for recurrent attacks of unilateral headaches, accompanied by complaints of photophobia, phonophobia, nausea, and vomiting. Two main categories of migraine are migraine with aura (MA) and migraine without aura (MO). Main body: Early twin and population studies have shown a genetic basis for these disorders, and efforts have been invested since to discern the genes involved. Many techniques, including candidate-gene association studies, loci linkage studies, genome-wide association, and transcription studies, have been used for this goal. As a result, several genes were pinned with concurrent and conflicting data among studies. It is important to understand the evolution of techniques and their findings. Conclusions: This review provides a chronological understanding of the different techniques used from the dawn of migraine genetic investigations and the genes linked with the migraine subtypes.
Collapse
Affiliation(s)
- Nader G. Zalaquett
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Elio Salameh
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Jonathan M. Kim
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Karen Tawk
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Mehdi Abouzari
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| |
Collapse
|
6
|
Takahashi K, Sato K. The Conventional and Breakthrough Tool for the Study of L-Glutamate Transporters. MEMBRANES 2024; 14:77. [PMID: 38668105 PMCID: PMC11052088 DOI: 10.3390/membranes14040077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/26/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024]
Abstract
In our recent report, we clarified the direct interaction between the excitatory amino acid transporter (EAAT) 1/2 and polyunsaturated fatty acids (PUFAs) by applying electrophysiological and molecular biological techniques to Xenopus oocytes. Xenopus oocytes have a long history of use in the scientific field, but they are still attractive experimental systems for neuropharmacological studies. We will therefore summarize the pharmacological significance, advantages (especially in the study of EAAT2), and experimental techniques that can be applied to Xenopus oocytes; our new findings concerning L-glutamate (L-Glu) transporters and PUFAs; and the significant outcomes of our data. The data obtained from electrophysiological and molecular biological studies of Xenopus oocytes have provided us with further important questions, such as whether or not some PUFAs can modulate EAATs as allosteric modulators and to what extent docosahexaenoic acid (DHA) affects neurotransmission and thereby affects brain functions. Xenopus oocytes have great advantages in the studies about the interactions between molecules and functional proteins, especially in the case when the expression levels of the proteins are small in cell culture systems without transfections. These are also proper to study the mechanisms underlying the interactions. Based on the data collected in Xenopus oocyte experiments, we can proceed to the next step, i.e., the physiological roles of the compounds and their significances. In the case of EAAT2, the effects on the neurotransmission should be examined by electrophysiological approach using acute brain slices. For new drug development, pharmacokinetics pharmacodynamics (PKPD) data and blood brain barrier (BBB) penetration data are also necessary. In order not to miss the promising candidate compounds at the primary stages of drug development, we should reconsider using Xenopus oocytes in the early phase of drug development.
Collapse
Grants
- a Research Grant on Regulatory Harmonization and Evaluation of Pharmaceuticals, Medical Devices, Regenerative and Cellular Therapy Products, Gene Therapy Products, and Cosmetics from AMED, Japan Japan Agency for Medical Research and Development
- KAKENHI 18700373, 21700422, 17K08330 Ministry of Education, Culture, Sports, Science and Technology
- a Grant for the Program for Promotion of Fundamental Studies in Health Sciences of NIBIO National Institute of Biomedical Innovation, Health and Nutrition
- a grant for Research on Risks of Chemicals, a Labor Science Research Grant for Research on New Drug Development MHLW
- a Grant-in-Aid from Hoansha Foundation Hoansha Foundation
Collapse
Affiliation(s)
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan;
| |
Collapse
|
7
|
Pietrobon D, Conti F. Astrocytic Na +, K + ATPases in physiology and pathophysiology. Cell Calcium 2024; 118:102851. [PMID: 38308916 DOI: 10.1016/j.ceca.2024.102851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
The Na+, K+ ATPases play a fundamental role in the homeostatic functions of astrocytes. After a brief historic prologue and discussion of the subunit composition and localization of the astrocytic Na+, K+ ATPases, the review focuses on the role of the astrocytic Na+, K+ pumps in extracellular K+ and glutamate homeostasis, intracellular Na+ and Ca2+ homeostasis and signaling, regulation of synaptic transmission and neurometabolic coupling between astrocytes and neurons. Loss-of-function mutations in the gene encoding the astrocytic α2 Na+, K+ ATPase cause a rare monogenic form of migraine with aura (familial hemiplegic migraine type 2). On the other hand, the α2 Na+, K+ ATPase is upregulated in spinal cord and brain samples from amyotrophic lateral sclerosis and Alzheimer disease patients, respectively. In the last part, the review focuses on i) the migraine relevant phenotypes shown by familial hemiplegic migraine type 2 knock-in mice with 50 % reduced expression of the astrocytic α2 Na+, K+ ATPase and the insights into the pathophysiology of migraine obtained from these genetic mouse models, and ii) the evidence that upregulation of the astrocytic α2 Na+, K+ ATPase in mouse models of amyotrophic lateral sclerosis and Alzheimer disease promotes neuroinflammation and contributes to progressive neurodegeneration.
Collapse
Affiliation(s)
- Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, Padova 35131, Italy.
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
8
|
Staehr C, Aalkjaer C, Matchkov V. The vascular Na,K-ATPase: clinical implications in stroke, migraine, and hypertension. Clin Sci (Lond) 2023; 137:1595-1618. [PMID: 37877226 PMCID: PMC10600256 DOI: 10.1042/cs20220796] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
In the vascular wall, the Na,K-ATPase plays an important role in the control of arterial tone. Through cSrc signaling, it contributes to the modulation of Ca2+ sensitivity in vascular smooth muscle cells. This review focuses on the potential implication of Na,K-ATPase-dependent intracellular signaling pathways in severe vascular disorders; ischemic stroke, familial migraine, and arterial hypertension. We propose similarity in the detrimental Na,K-ATPase-dependent signaling seen in these pathological conditions. The review includes a retrospective proteomics analysis investigating temporal changes after ischemic stroke. The analysis revealed that the expression of Na,K-ATPase α isoforms is down-regulated in the days and weeks following reperfusion, while downstream Na,K-ATPase-dependent cSrc kinase is up-regulated. These results are important since previous studies have linked the Na,K-ATPase-dependent cSrc signaling to futile recanalization and vasospasm after stroke. The review also explores a link between the Na,K-ATPase and migraine with aura, as reduced expression or pharmacological inhibition of the Na,K-ATPase leads to cSrc kinase signaling up-regulation and cerebral hypoperfusion. The review discusses the role of an endogenous cardiotonic steroid-like compound, ouabain, which binds to the Na,K-ATPase and initiates the intracellular cSrc signaling, in the pathophysiology of arterial hypertension. Currently, our understanding of the precise control mechanisms governing the Na,K-ATPase/cSrc kinase regulation in the vascular wall is limited. Understanding the role of vascular Na,K-ATPase signaling is essential for developing targeted treatments for cerebrovascular disorders and hypertension, as the Na,K-ATPase is implicated in the pathogenesis of these conditions and may contribute to their comorbidity.
Collapse
Affiliation(s)
- Christian Staehr
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Aarhus, Denmark
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Danish Cardiovascular Academy, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| |
Collapse
|
9
|
de Ruiter Swain J, Michalopoulou E, Noch EK, Lukey MJ, Van Aelst L. Metabolic partitioning in the brain and its hijacking by glioblastoma. Genes Dev 2023; 37:681-702. [PMID: 37648371 PMCID: PMC10546978 DOI: 10.1101/gad.350693.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The different cell types in the brain have highly specialized roles with unique metabolic requirements. Normal brain function requires the coordinated partitioning of metabolic pathways between these cells, such as in the neuron-astrocyte glutamate-glutamine cycle. An emerging theme in glioblastoma (GBM) biology is that malignant cells integrate into or "hijack" brain metabolism, co-opting neurons and glia for the supply of nutrients and recycling of waste products. Moreover, GBM cells communicate via signaling metabolites in the tumor microenvironment to promote tumor growth and induce immune suppression. Recent findings in this field point toward new therapeutic strategies to target the metabolic exchange processes that fuel tumorigenesis and suppress the anticancer immune response in GBM. Here, we provide an overview of the intercellular division of metabolic labor that occurs in both the normal brain and the GBM tumor microenvironment and then discuss the implications of these interactions for GBM therapy.
Collapse
Affiliation(s)
- Jed de Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Cold Spring Harbor Laboratory School of Biological Sciences, Cold Spring Harbor, New York 11724, USA
| | | | - Evan K Noch
- Department of Neurology, Division of Neuro-oncology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Michael J Lukey
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| |
Collapse
|
10
|
de Melo AD, Freire VAF, Diogo ÍL, Santos HDL, Barbosa LA, de Carvalho LED. Antioxidant Therapy Reduces Oxidative Stress, Restores Na,K-ATPase Function and Induces Neuroprotection in Rodent Models of Seizure and Epilepsy: A Systematic Review and Meta-Analysis. Antioxidants (Basel) 2023; 12:1397. [PMID: 37507936 PMCID: PMC10376594 DOI: 10.3390/antiox12071397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/30/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by epileptic seizures resulting from neuronal hyperexcitability, which may be related to failures in Na,K-ATPase activity and oxidative stress participation. We conducted this study to investigate the impact of antioxidant therapy on oxidative stress, Na,K-ATPase activity, seizure factors, and mortality in rodent seizure/epilepsy models induced by pentylenetetrazol (PTZ), pilocarpine (PILO), and kainic acid (KA). After screening 561 records in the MEDLINE, EMBASE, Web of Science, Science Direct, and Scopus databases, 22 were included in the systematic review following the PRISMA guidelines. The meta-analysis included 14 studies and showed that in epileptic animals there was an increase in the oxidizing agents nitric oxide (NO) and malondialdehyde (MDA), with a reduction in endogenous antioxidants reduced glutathione (GSH) and superoxide dismutase (SO). The Na,K-ATPase activity was reduced in all areas evaluated. Antioxidant therapy reversed all of these parameters altered by seizure or epilepsy induction. In addition, there was a percentage decrease in the number of seizures and mortality, and a meta-analysis showed a longer seizure latency in animals using antioxidant therapy. Thus, this study suggests that the use of antioxidants promotes neuroprotective effects and mitigates the effects of epilepsy. The protocol was registered in the Prospective Register of Systematic Reviews (PROSPERO) CRD42022356960.
Collapse
Affiliation(s)
- Anderson Dutra de Melo
- Departamento de Ciências e Linguagens, Instituto Federal de Minas Gerais, Bambui 38900-000, Minas Gerais, Brazil
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Divinopolis 35501-296, Minas Gerais, Brazil
| | - Victor Antonio Ferreira Freire
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Divinopolis 35501-296, Minas Gerais, Brazil
| | - Ítalo Leonardo Diogo
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Divinopolis 35501-296, Minas Gerais, Brazil
| | - Hérica de Lima Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Divinopolis 35501-296, Minas Gerais, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Divinopolis 35501-296, Minas Gerais, Brazil
| | | |
Collapse
|
11
|
Murlanova K, Jouroukhin Y, Novototskaya-Vlasova K, Huseynov S, Pletnikova O, Morales MJ, Guan Y, Kamiya A, Bergles DE, Dietz DM, Pletnikov MV. Loss of Astrocytic µ Opioid Receptors Exacerbates Aversion Associated with Morphine Withdrawal in Mice: Role of Mitochondrial Respiration. Cells 2023; 12:1412. [PMID: 37408246 PMCID: PMC10216734 DOI: 10.3390/cells12101412] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/19/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Astrocytes express mu/µ opioid receptors, but the function of these receptors remains poorly understood. We evaluated the effects of astrocyte-restricted knockout of µ opioid receptors on reward- and aversion-associated behaviors in mice chronically exposed to morphine. Specifically, one of the floxed alleles of the Oprm1 gene encoding µ opioid receptor 1 was selectively deleted from brain astrocytes in Oprm1 inducible conditional knockout (icKO) mice. These mice did not exhibit changes in locomotor activity, anxiety, or novel object recognition, or in their responses to the acute analgesic effects of morphine. Oprm1 icKO mice displayed increased locomotor activity in response to acute morphine administration but unaltered locomotor sensitization. Oprm1 icKO mice showed normal morphine-induced conditioned place preference but exhibited stronger conditioned place aversion associated with naloxone-precipitated morphine withdrawal. Notably, elevated conditioned place aversion lasted up to 6 weeks in Oprm1 icKO mice. Astrocytes isolated from the brains of Oprm1 icKO mice had unchanged levels of glycolysis but had elevated oxidative phosphorylation. The basal augmentation of oxidative phosphorylation in Oprm1 icKO mice was further exacerbated by naloxone-precipitated withdrawal from morphine and, similar to that for conditioned place aversion, was still present 6 weeks later. Our findings suggest that µ opioid receptors in astrocytes are linked to oxidative phosphorylation and they contribute to long-term changes associated with opioid withdrawal.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yan Jouroukhin
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ksenia Novototskaya-Vlasova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Shovgi Huseynov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Olga Pletnikova
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J. Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurological Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dwight E. Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David M. Dietz
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V. Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Costa RT, Santos MB, Alberto-Silva C, Carrettiero DC, Ribeiro CAJ. Methylmalonic Acid Impairs Cell Respiration and Glutamate Uptake in C6 Rat Glioma Cells: Implications for Methylmalonic Acidemia. Cell Mol Neurobiol 2023; 43:1163-1180. [PMID: 35674974 PMCID: PMC11414442 DOI: 10.1007/s10571-022-01236-1] [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: 01/07/2022] [Accepted: 05/20/2022] [Indexed: 11/28/2022]
Abstract
Methylmalonic acidemia is an organic acidemia caused by deficient activity of L-methylmalonyl-CoA mutase or its cofactor cyanocobalamin and it is biochemically characterized by an accumulation of methylmalonic acid (MMA) in tissue and body fluids of patients. The main clinical manifestations of this disease are neurological and observable symptoms during metabolic decompensation are encephalopathy, cerebral atrophy, coma, and seizures, which commonly appear in newborns. This study aimed to investigate the toxic effects of MMA in a glial cell line presenting astrocytic features. Astroglial C6 cells were exposed to MMA (0.1-10 mM) for 24 or 48 h and cell metabolic viability, glucose consumption, and oxygen consumption rate, as well as glutamate uptake and ATP content were analyzed. The possible preventive effects of bezafibrate were also evaluated. MMA significantly reduced cell metabolic viability after 48-h period and increased glucose consumption during the same period of incubation. Regarding the energy homeostasis, MMA significantly reduced respiratory parameters of cells after 48-h exposure, indicating that cell metabolism is compromised at resting and reserve capacity state, which might influence the cell capacity to meet energetic demands. Glutamate uptake and ATP content were also compromised after exposure to MMA, which can be influenced energy metabolism impairment, affecting the functionality of the astroglial cells. Our findings suggest that these effects could be involved in the pathophysiology of neurological dysfunction of this disease. Methylmalonic acid compromises mitochondrial functioning leading to reduced ATP production and reduces glutamate uptake by C6 astroglial cells.
Collapse
Affiliation(s)
- Renata T Costa
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Marcella B Santos
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Carlos Alberto-Silva
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Daniel C Carrettiero
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - César A J Ribeiro
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil.
| |
Collapse
|
13
|
Conti F, Pietrobon D. Astrocytic Glutamate Transporters and Migraine. Neurochem Res 2023; 48:1167-1179. [PMID: 36583835 DOI: 10.1007/s11064-022-03849-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022]
Abstract
Glutamate levels and lifetime in the brain extracellular space are dinamically regulated by a family of Na+- and K+-dependent glutamate transporters, which thereby control numerous brain functions and play a role in numerous neurological and psychiatric diseases. Migraine is a neurological disorder characterized by recurrent attacks of typically throbbing and unilateral headache and by a global dysfunction in multisensory processing. Familial hemiplegic migraine type 2 (FHM2) is a rare monogenic form of migraine with aura caused by loss-of-function mutations in the α2 Na/K ATPase (α2NKA). In the adult brain, this pump is expressed almost exclusively in astrocytes where it is colocalized with glutamate transporters. Knockin mouse models of FHM2 (FHM2 mice) show a reduced density of glutamate transporters in perisynaptic astrocytic processes (mirroring the reduced expression of α2NKA) and a reduced rate of glutamate clearance at cortical synapses during neuronal activity and sensory stimulation. Here we review the migraine-relevant alterations produced by the astrocytic glutamate transport dysfunction in FHM2 mice and their underlying mechanisms, in particular regarding the enhanced brain susceptibility to cortical spreading depression (the phenomenon that underlies migraine aura and can also initiate the headache mechanisms) and the enhanced algesic response to a migraine trigger.
Collapse
Affiliation(s)
- Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.
| | - Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, 35131, Padua, Italy.
- CNR Institute of Neuroscience, 35131, Padua, Italy.
| |
Collapse
|
14
|
Garcia IJP, Kinoshita PF, Valadares JMDM, de Carvalho LED, Cortes VF, Barbosa LA, Scavone C, Santos HDL. Effect of Ouabain on Glutamate Transport in the Hippocampus of Rats with LPS-Induced Neuroinflammation. Biomedicines 2023; 11:biomedicines11030920. [PMID: 36979899 PMCID: PMC10045517 DOI: 10.3390/biomedicines11030920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
A lipopolysaccharide (LPS)-induced neuroinflammation rat model was used to study the effects of ouabain (OUA) at low concentrations, which can interact with the Na,K-ATPase, causing the modulation of intracellular signalling pathways in the Central Nervous System. Our study aimed to analyse the effects of OUA on glutamate transport in the hippocampus of rats with LPS-induced neuroinflammation. Adult male Wistar rats were divided into four groups: OUA (1.8 µg/kg), saline (CTR), LPS (200 µg/kg), and OUA + LPS (OUA 20 min before LPS). The animals were sacrificed after 2 h, and the hippocampus was collected for analysis. After treatment, we determined the activities of Na,K-ATPase and glutamine synthetase (GS). In addition, expression of the α1, α2, and α3 isoforms of Na,K-ATPase and the glutamate transporters, EAAT1 and EAAT2, were also analysed. Treatment with OUA caused a specific increase in the α2 isoform expression (~20%), whereas LPS decreased its expression (~22%), and treatment with OUA before LPS prevented the effects of LPS. Moreover, LPS caused a decrease of approximately 50% in GS activity compared with that in the CTR group; however, OUA pre-treatment attenuated this effect of LPS. Notably, it was found that treatment with OUA caused an increase in the expression of EAAT1 (~30%) and EAAT2 (~25%), whereas LPS caused a decrease in the expression of EAAT1 (~23%) and EAAT2 (~25%) compared with that in the CTR group. When treated with OUA, the effects of LPS were abrogated. In conclusion, the OUA pre-treatment abolished the effect caused by LPS, suggesting that this finding may be related to the restoration of the interaction between FXYD2 and the studied membrane proteins.
Collapse
Affiliation(s)
- Israel José Pereira Garcia
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Paula Fernanda Kinoshita
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jéssica Martins de Moura Valadares
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Luciana Estefani Drumond de Carvalho
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Vanessa Faria Cortes
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Leandro Augusto Barbosa
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Cristoforo Scavone
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| | - Hérica de Lima Santos
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| |
Collapse
|
15
|
EPO has multiple positive effects on astrocytes in an experimental model of ischemia. Brain Res 2023; 1802:148207. [PMID: 36549360 DOI: 10.1016/j.brainres.2022.148207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Erythropoietin (EPO) has neuroprotective effects in central nervous system injury models. In clinical trials EPO has shown beneficial effects in traumatic brain injury (TBI) as well as in ischemic stroke. We have previously shown that EPO has short-term effects on astrocyte glutamatergic signaling in vitro and that administration of EPO after experimental TBI decreases early cytotoxic brain edema and preserves structural and functional properties of the blood-brain barrier. These effects have been attributed to preserved or restored astrocyte function. Here we explored the effects of EPO on astrocytes undergoing oxygen-glucose-deprivation, an in vitro model of ischemia. Measurements of glutamate uptake, intracellular pH, intrinsic NADH fluorescence, Na,K-ATPase activity, and lactate release were performed. We found that EPO within minutes caused a Na,K-ATPase-dependent increase in glutamate uptake, restored intracellular acidification caused by glutamate and increased lactate release. The effects on intracellular pH were dependent on the sodium/hydrogen exchanger NHE. In neuron-astrocyte co-cultures, EPO increased NADH production both in astrocytes and neurons, however the increase was greater in astrocytes. We suggest that EPO preserves astrocyte function under ischemic conditions and thus may contribute to neuroprotection in ischemic stroke and brain ischemia secondary to TBI.
Collapse
|
16
|
Silva-Parra J, Sandu C, Felder-Schmittbuhl MP, Hernández-Kelly LC, Ortega A. Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. Neurotox Res 2023; 41:103-117. [PMID: 36607593 DOI: 10.1007/s12640-022-00623-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janisse Silva-Parra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México.
| |
Collapse
|
17
|
Kang EJ, Prager O, Lublinsky S, Oliveira-Ferreira AI, Reiffurth C, Major S, Müller DN, Friedman A, Dreier JP. Stroke-prone salt-sensitive spontaneously hypertensive rats show higher susceptibility to spreading depolarization (SD) and altered hemodynamic responses to SD. J Cereb Blood Flow Metab 2023; 43:210-230. [PMID: 36329390 PMCID: PMC9903222 DOI: 10.1177/0271678x221135085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Spreading depolarization (SD) occurs in a plethora of clinical conditions including migraine aura, delayed ischemia after subarachnoid hemorrhage and malignant hemispheric stroke. It describes waves of near-breakdown of ion homeostasis, particularly Na+ homeostasis in brain gray matter. SD induces tone alterations in resistance vessels, causing either hyperperfusion in healthy tissue; or hypoperfusion (inverse hemodynamic response = spreading ischemia) in tissue at risk. Observations from mice with genetic dysfunction of the ATP1A2-encoded α2-isoform of Na+/K+-ATPase (α2NaKA) suggest a mechanistic link between (1) SD, (2) vascular dysfunction, and (3) salt-sensitive hypertension via α2NaKA. Thus, α2NaKA-dysfunctional mice are more susceptible to SD and show a shift toward more inverse hemodynamic responses. α2NaKA-dysfunctional patients suffer from familial hemiplegic migraine type 2, a Mendelian model disease of SD. α2NaKA-dysfunctional mice are also a genetic model of salt-sensitive hypertension. To determine whether SD thresholds and hemodynamic responses are also altered in other genetic models of salt-sensitive hypertension, we examined these variables in stroke-prone spontaneously hypertensive rats (SHRsp). Compared with Wistar Kyoto control rats, we found in SHRsp that electrical SD threshold was significantly reduced, propagation speed was increased, and inverse hemodynamic responses were prolonged. These results may have relevance to both migraine with aura and stroke.
Collapse
Affiliation(s)
- Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ofer Prager
- Department of Physiology & Cell Biology, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Svetlana Lublinsky
- Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ana I Oliveira-Ferreira
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Clemens Reiffurth
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alon Friedman
- Department of Physiology & Cell Biology, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
18
|
Martinez NP, Pinch M, Kandel Y, Hansen IA. Knockdown of the Sodium/Potassium ATPase Subunit Beta 2 Reduces Egg Production in the Dengue Vector, Aedes aegypti. INSECTS 2023; 14:50. [PMID: 36661978 PMCID: PMC9862990 DOI: 10.3390/insects14010050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/26/2022] [Accepted: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The Na+/K+ ATPase (NKA) is present in the cellular membrane of most eukaryotic cells. It utilizes energy released by ATP hydrolysis to pump sodium ions out of the cell and potassium ions into the cell, which establishes and controls ion gradients. Functional NKA pumps consist of three subunits, alpha, beta, and FXYD. The alpha subunit serves as the catalytic subunit while the beta and FXYD subunits regulate the proper folding and localization, and ion affinity of the alpha subunit, respectively. Here we demonstrate that knockdown of NKA beta subunit 2 mRNA (nkaβ2) reduces fecundity in female Ae. aegypti. We determined the expression pattern of nkaβ2 in several adult mosquito organs using qRT-PCR. We performed RNAi-mediated knockdown of nkaβ2 and assayed for lethality, and effects on female fecundity. Tissue expression levels of nkaβ2 mRNA were highest in the ovaries with the fat body, midgut and thorax having similar expression levels, while Malpighian tubules had significantly lower expression. Survival curves recorded post dsRNA injection showed a non-significant decrease in survival of nkaβ2 dsRNA-injected mosquitoes compared to GFP dsRNA-injected mosquitoes. We observed a significant reduction in the number of eggs laid by nkaβ2 dsRNA-injected mosquitoes compared to control mosquitoes. These results, coupled with the tissue expression profile of nkaβ2, indicate that this subunit plays a role in normal female Ae. aegypti fecundity. Additional research needs to be conducted to determine the exact role played by NKAβ2 in mosquito post-blood meal nutrient sensing, transport, yolk precursor protein (YPP) synthesis and yolk deposition.
Collapse
Affiliation(s)
- Nathan P. Martinez
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA
| | | | | | | |
Collapse
|
19
|
Functional investigation of SLC1A2 variants associated with epilepsy. Cell Death Dis 2022; 13:1063. [PMID: 36543780 PMCID: PMC9772344 DOI: 10.1038/s41419-022-05457-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022]
Abstract
Epilepsy is a common neurological disorder and glutamate excitotoxicity plays a key role in epileptic pathogenesis. Astrocytic glutamate transporter GLT-1 is responsible for preventing excitotoxicity via clearing extracellular accumulated glutamate. Previously, three variants (G82R, L85P, and P289R) in SLC1A2 (encoding GLT-1) have been clinically reported to be associated with epilepsy. However, the functional validation and underlying mechanism of these GLT-1 variants in epilepsy remain undetermined. In this study, we reported that these disease-linked mutants significantly decrease glutamate uptake, cell membrane expression of the glutamate transporter, and glutamate-elicited current. Additionally, we found that these variants may disturbed stromal-interacting molecule 1 (STIM1)/Orai1-mediated store-operated Ca2+ entry (SOCE) machinery in the endoplasmic reticulum (ER), in which GLT-1 may be a new partner of SOCE. Furthermore, knock-in mice with disease-associated variants showed a hyperactive phenotype accompanied by reduced glutamate transporter expression. Therefore, GLT-1 is a promising and reliable therapeutic target for epilepsy interventions.
Collapse
|
20
|
Asah S, Alganem K, McCullumsmith RE, O'Donovan SM. A bioinformatic inquiry of the EAAT2 interactome in postmortem and neuropsychiatric datasets. Schizophr Res 2022; 249:38-46. [PMID: 32197935 PMCID: PMC7494586 DOI: 10.1016/j.schres.2020.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Altered expression and localization of the glutamate transporter EAAT2 is found in schizophrenia and other neuropsychiatric (major depression, MDD) and neurological disorders (amyotrophic lateral sclerosis, ALS). However, the EAAT2 interactome, the network of proteins that physically or functionally interact with EAAT2 to support its activity, has yet to be characterized in severe mental illness. We compiled a list of "core" EAAT2 interacting proteins. Using Kaleidoscope, an R-shiny application, we data mined publically available postmortem transcriptome datasets to determine whether components of the EAAT2 interactome are differentially expressed in schizophrenia and, using Reactome, identify which interactome-associated biological pathways are altered. Overall, these "look up" studies highlight region-specific, primarily frontal cortex (dorsolateral prefrontal cortex and anterior cingulate cortex), changes in the EAAT2 interactome and implicate altered metabolism pathways in schizophrenia. Pathway analyses also suggest that perturbation of components of the EAAT2 interactome in animal models of antipsychotic administration impact metabolism. Similar changes in metabolism pathways are seen in ALS, in addition to altered expression of many components of the EAAT2 interactome. However, although EAAT2 expression is altered in a postmortem MDD dataset, few other components of the EAAT2 interactome are changed. Thus, "look up" studies suggest region- and disease-relevant biological pathways related to the EAAT2 interactome that implicate glutamate reuptake perturbations in schizophrenia, while providing a useful tool to exploit "omics" datasets.
Collapse
Affiliation(s)
- Sophie Asah
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | | | | |
Collapse
|
21
|
Suñol M, Alemany S, Bustamante M, Diez I, Contreras-Rodríguez O, Laudo B, Macià D, Martínez-Vilavella G, Martínez-Zalacaín I, Menchón JM, Pujol J, Sunyer J, Sepulcre J, Soriano-Mas C. Neurogenetics of Dynamic Connectivity Patterns Associated With Obsessive-Compulsive Symptoms in Healthy Children. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:411-420. [PMID: 36324658 PMCID: PMC9616269 DOI: 10.1016/j.bpsgos.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/28/2021] [Accepted: 11/14/2021] [Indexed: 01/31/2023] Open
Abstract
Background Obsessive-compulsive symptoms (OCSs) during childhood predispose to obsessive-compulsive disorder and have been associated with changes in brain circuits altered in obsessive-compulsive disorder samples. OCSs may arise from disturbed glutamatergic neurotransmission, impairing cognitive oscillations and promoting overstable functional states. Methods A total of 227 healthy children completed the Obsessive Compulsive Inventory-Child Version and underwent a resting-state functional magnetic resonance imaging examination. Genome-wide data were obtained from 149 of them. We used a graph theory-based approach and characterized associations between OCSs and dynamic functional connectivity (dFC). dFC evaluates fluctuations over time in FC between brain regions, which allows characterizing regions with stable connectivity patterns (attractors). We then compared the spatial similarity between OCS-dFC correlation maps and mappings of genetic expression across brain regions to identify genes potentially associated with connectivity changes. In post hoc analyses, we investigated which specific single nucleotide polymorphisms of these genes moderated the association between OCSs and patterns of dFC. Results OCSs correlated with decreased attractor properties in the left ventral putamen and increased attractor properties in (pre)motor areas and the left hippocampus. At the specific symptom level, increased attractor properties in the right superior parietal cortex correlated with ordering symptoms. In the hippocampus, we identified two single nucleotide polymorphisms in glutamatergic neurotransmission genes (GRM7, GNAQ) that moderated the association between OCSs and attractor features. Conclusions We provide evidence that in healthy children, the association between dFC changes and OCSs may be mapped onto brain circuits predicted by prevailing neurobiological models of obsessive-compulsive disorder. Moreover, our findings support the involvement of glutamatergic neurotransmission in such brain network changes.
Collapse
Affiliation(s)
- Maria Suñol
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
- Mental Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- Gordon Center for Medical Imaging, Department of Radiology and Nuclear Medicine, Harvard Medical School, Boston
| | - Silvia Alemany
- Epidemiology and Public Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- Barcelona Institute for Global Health, Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Mariona Bustamante
- Epidemiology and Public Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- Barcelona Institute for Global Health, Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology and Nuclear Medicine, Harvard Medical School, Boston
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts
| | - Oren Contreras-Rodríguez
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
- Mental Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
| | - Berta Laudo
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Dídac Macià
- Barcelona Institute for Global Health, Barcelona, Spain
- MRI Research Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
| | | | - Ignacio Martínez-Zalacaín
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - José Manuel Menchón
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
- Mental Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
| | - Jesús Pujol
- Mental Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- MRI Research Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
| | - Jordi Sunyer
- Epidemiology and Public Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- Barcelona Institute for Global Health, Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Medical Research Institute, Hospital del Mar, Barcelona, Spain
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology and Nuclear Medicine, Harvard Medical School, Boston
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts
| | - Carles Soriano-Mas
- Department of Psychiatry, Bellvitge University Hospital and Bellvitge Biomedical Research Institute, Barcelona, Spain
- Mental Health Networking Biomedical Research Centre, Carlos III Health Institute, Barcelona, Spain
- Department of Psychobiology and Methodology in Health Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Rabinowicz S, Schreiber T, Heimer G, Bar-Yosef O, Nissenkorn A, E ZD, Arkush L, Hamed N, Ben-Zeev B, Tzadok M. Felbamate for pediatric epilepsy—should we keep on using it as the last resort? Front Neurol 2022; 13:979725. [PMID: 36203978 PMCID: PMC9530252 DOI: 10.3389/fneur.2022.979725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionConcerns regarding felbamate adverse effects restrict its widespread use in children with drug-resistant epilepsy. We aimed to examine the efficacy and safety of felbamate in those children and identify the ones who may benefit most from its use.MethodsWe retrospectively reviewed the medical files of all patients who were treated with felbamate in a tertiary pediatric epilepsy clinic between 2009–2021. Drug efficacy was determined at the first 3 months of treatment and thereafter. Therapeutic response and adverse reactions were monitored throughout the course of treatment.ResultsOur study included 75 children (age 8.9 ± 3.7 years), of whom 53 were treated with felbamate for seizures, 16 for electrical status epilepticus during sleep and 6 for both. The median follow-up time was 16 months (range 1–129 months). The most common cause for epilepsy was genetic (29%). The median number of previous anti-seizure medications was six [4–8]. A therapeutic response ≥50% was documented in 37 (51%) patients, and a complete response in 9 (12%). Nineteen patients (25%) sustained adverse reactions, including three cases of elevated liver enzymes and one case of neutropenia with normal bone marrow aspiration. In all cases, treatment could be continued. All children with intractable epilepsy following herpes encephalitis showed a response to felbamate.ConclusionFelbamate is an efficacious and safe anti-seizure medication in the pediatric population.
Collapse
Affiliation(s)
- Shira Rabinowicz
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- *Correspondence: Shira Rabinowicz
| | - Tal Schreiber
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gali Heimer
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Omer Bar-Yosef
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Andreea Nissenkorn
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Zohar-Dayan E
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Leo Arkush
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nasrin Hamed
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Bruria Ben-Zeev
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michal Tzadok
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
23
|
Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol 2022; 217:102331. [PMID: 35872221 DOI: 10.1016/j.pneurobio.2022.102331] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes contribute to the complex cellular pathology of Alzheimer's disease (AD). Neurons and astrocytes function in close collaboration through neurotransmitter recycling, collectively known as the glutamate/GABA-glutamine cycle, which is essential to sustain neurotransmission. Neurotransmitter recycling is intimately linked to astrocyte energy metabolism. In the course of AD, astrocytes undergo extensive metabolic remodeling, which may profoundly affect the glutamate/GABA-glutamine cycle. The consequences of altered astrocyte function and metabolism in relation to neurotransmitter recycling are yet to be comprehended. Metabolic alterations of astrocytes in AD deprive neurons of metabolic support, thereby contributing to synaptic dysfunction and neurodegeneration. In addition, several astrocyte-specific components of the glutamate/GABA-glutamine cycle, including glutamine synthesis and synaptic neurotransmitter uptake, are perturbed in AD. Integration of the complex astrocyte biology within the context of AD is essential for understanding the fundamental mechanisms of the disease, while restoring astrocyte metabolism may serve as an approach to arrest or even revert clinical progression of AD.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
24
|
Rodrigues T, Piccirillo S, Magi S, Preziuso A, Dos Santos Ramos V, Serfilippi T, Orciani M, Maciel Palacio Alvarez M, Luis Dos Santos Tersariol I, Amoroso S, Lariccia V. Control of Ca 2+ and metabolic homeostasis by the Na +/Ca 2+ exchangers (NCXs) in health and disease. Biochem Pharmacol 2022; 203:115163. [PMID: 35803319 DOI: 10.1016/j.bcp.2022.115163] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
Abstract
Spatial and temporal control of calcium (Ca2+) levels is essential for the background rhythms and responses of living cells to environmental stimuli. Whatever other regulators a given cellular activity may have, localized and wider scale Ca2+ events (sparks, transients, and waves) are hierarchical determinants of fundamental processes such as cell contraction, excitability, growth, metabolism and survival. Different cell types express specific channels, pumps and exchangers to efficiently generate and adapt Ca2+ patterns to cell requirements. The Na+/Ca2+ exchangers (NCXs) in particular contribute to Ca2+ homeostasis by buffering intracellular Ca2+ loads according to the electrochemical gradients of substrate ions - i.e., Ca2+ and sodium (Na+) - and under a dynamic control of redundant regulatory processes. An interesting feature of NCX emerges from the strict relationship that connects transporter activity with cell metabolism: on the one hand NCX operates under constant control of ATP-dependent regulatory processes, on the other hand the ion fluxes generated through NCX provide mechanistic support for the Na+-driven uptake of glutamate and Ca2+ influx to fuel mitochondrial respiration. Proof of concept evidence highlights therapeutic potential of preserving a timed and balanced NCX activity in a growing rate of diseases (including excitability, neurodegenerative, and proliferative disorders) because of an improved ability of stressed cells to safely maintain ion gradients and mitochondrial bioenergetics. Here, we will summarize and review recent works that have focused on the pathophysiological roles of NCXs in balancing the two-way relationship between Ca2+ signals and metabolism.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vyctória Dos Santos Ramos
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Histology, University "Politecnica delle Marche", Ancona, Italy.
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, São Paulo School of Medicine, Federal University of São Paulo (Unifesp) São Paulo, SP, Brazil
| | | | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| |
Collapse
|
25
|
Abulseoud OA, Alasmari F, Hussein AM, Sari Y. Ceftriaxone as a Novel Therapeutic Agent for Hyperglutamatergic States: Bridging the Gap Between Preclinical Results and Clinical Translation. Front Neurosci 2022; 16:841036. [PMID: 35864981 PMCID: PMC9294323 DOI: 10.3389/fnins.2022.841036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of glutamate homeostasis is a well-established core feature of neuropsychiatric disorders. Extracellular glutamate concentration is regulated by glutamate transporter 1 (GLT-1). The discovery of a beta-lactam antibiotic, ceftriaxone (CEF), as a safe compound with unique ability to upregulate GLT-1 sparked the interest in testing its efficacy as a novel therapeutic agent in animal models of neuropsychiatric disorders with hyperglutamatergic states. Indeed, more than 100 preclinical studies have shown the efficacy of CEF in attenuating the behavioral manifestations of various hyperglutamatergic brain disorders such as ischemic stroke, amyotrophic lateral sclerosis (ALS), seizure, Huntington’s disease, and various aspects of drug use disorders. However, despite rich and promising preclinical data, only one large-scale clinical trial testing the efficacy of CEF in patients with ALS is reported. Unfortunately, in that study, there was no significant difference in survival between placebo- and CEF-treated patients. In this review, we discussed the translational potential of preclinical efficacy of CEF based on four different parameters: (1) initiation of CEF treatment in relation to induction of the hyperglutamatergic state, (2) onset of response in preclinical models in relation to onset of GLT-1 upregulation, (3) mechanisms of action of CEF on GLT-1 expression and function, and (4) non-GLT-1-mediated mechanisms for CEF. Our detailed review of the literature brings new insights into underlying molecular mechanisms correlating the preclinical efficacy of CEF. We concluded here that CEF may be clinically effective in selected cases in acute and transient hyperglutamatergic states such as early drug withdrawal conditions.
Collapse
Affiliation(s)
- Osama A. Abulseoud
- Department of Psychiatry and Psychology, Alex School of Medicine at Mayo Clinic, Phoenix, AZ, United States
- *Correspondence: Osama A. Abulseoud,
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Youssef Sari,
| |
Collapse
|
26
|
Murlanova K, Jouroukhin Y, Huseynov S, Pletnikova O, Morales MJ, Guan Y, Baraban JM, Bergles DE, Pletnikov MV. Deficient mitochondrial respiration in astrocytes impairs trace fear conditioning and increases naloxone-precipitated aversion in morphine-dependent mice. Glia 2022; 70:1289-1300. [PMID: 35275429 PMCID: PMC9773362 DOI: 10.1002/glia.24169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
Abstract
Mitochondria are abundant in the fine processes of astrocytes, however, potential roles for astrocyte mitochondria remain poorly understood. In the present study, we performed a systematic examination of the effects of abnormal oxidative phosphorylation in astrocytes on several mouse behaviors. Impaired astrocyte oxidative phosphorylation was produced by astrocyte-specific deletion of the nuclear mitochondrial gene, Cox10, that encodes an accessory protein of complex IV, the protoheme:heme-O-farnesyl transferase. As expected, conditional deletion of the Cox10 gene in mice (cKO mice) significantly reduced expression of COX10 and Cytochrome c oxidase subunit I (MTCO1) of Complex IV, resulting in decreased oxidative phosphorylation without significantly affecting glycolysis. No effects of the deletion were observed on locomotor activity, anxiety-like behavior, nociception, or spontaneous alternation. Cox10 cKO female mice exhibited mildly impaired novel object recognition, while Cox10 cKO male mice were moderately deficient in trace fear conditioning. No group-related changes were observed in conditional place preference (CPP) that assessed effects of morphine on reward. In contrast to CPP, Cox10 cKO mice demonstrated significantly increased aversive behaviors produced by naloxone-precipitated withdrawal following chronic exposure to morphine, that is, jumping and avoidance behavior as assessed by conditional place aversion (CPA). Our study suggests that astrocyte oxidative phosphorylation may contribute to behaviors associated with greater cognitive load and/or aversive and stressful conditions.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine, State University of New York at Buffalo, Buffalo, New York, USA
| | - Yan Jouroukhin
- Department of Physiology and Biophysics, Jacobs School of Medicine, State University of New York at Buffalo, Buffalo, New York, USA
| | - Shovgi Huseynov
- Department of Physiology and Biophysics, Jacobs School of Medicine, State University of New York at Buffalo, Buffalo, New York, USA,Molecular Basis of Integrative Activity, Academician Abdulla Garayev Institute of Physiology, National Academy of Sciences of Azerbaijan, Baku, Azerbaijan
| | - Olga Pletnikova
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine; State University of New York at Buffalo, Buffalo, New York, USA,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael J. Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine, State University of New York at Buffalo, Buffalo, New York, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Neurological Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jay M. Baraban
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dwight E. Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mikhail V. Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine, State University of New York at Buffalo, Buffalo, New York, USA,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Xiong Y, Xu G, Chen M, Ma H. Intestinal Uptake and Tolerance to Food Antigens. Front Immunol 2022; 13:906122. [PMID: 35757706 PMCID: PMC9226482 DOI: 10.3389/fimmu.2022.906122] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Food allergy is a growing concern due to its increasing world-wide incidence. Strict avoidance of allergens is a passive treatment strategy. Since the mechanisms responsible for the occurrence and development of food allergy have not yet been fully elucidated, effective individualized treatment options are lacking. In this review, we summarize the pathways through which food antigens enter the intestine and review the proposed mechanisms describing how the intestine acquires and tolerates food antigens. When oral tolerance is not established, food allergy occurs. In addition, we also discuss the contribution of commensal bacteria of the gut in shaping tolerance to food antigens in the intestinal tract. Finally, we propose that elucidating the mechanisms of intestinal uptake and tolerance of food antigens will provide additional clues for potential treatment options for food allergy.
Collapse
Affiliation(s)
- Yuhong Xiong
- Department of Pediatrics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Guifeng Xu
- Department of Pediatrics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mingwu Chen
- Department of Pediatrics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongdi Ma
- Department of Pediatrics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
28
|
Sijben HJ, Dall’ Acqua L, Liu R, Jarret A, Christodoulaki E, Onstein S, Wolf G, Verburgt SJ, Le Dévédec SE, Wiedmer T, Superti-Furga G, IJzerman AP, Heitman LH. Impedance-Based Phenotypic Readout of Transporter Function: A Case for Glutamate Transporters. Front Pharmacol 2022; 13:872335. [PMID: 35677430 PMCID: PMC9169222 DOI: 10.3389/fphar.2022.872335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
Excitatory amino acid transporters (EAAT/SLC1) mediate Na+-dependent uptake of extracellular glutamate and are potential drug targets for neurological disorders. Conventional methods to assess glutamate transport in vitro are based on radiolabels, fluorescent dyes or electrophysiology, which potentially compromise the cell’s physiology and are generally less suited for primary drug screens. Here, we describe a novel label-free method to assess human EAAT function in living cells, i.e., without the use of chemical modifications to the substrate or cellular environment. In adherent HEK293 cells overexpressing EAAT1, stimulation with glutamate or aspartate induced cell spreading, which was detected in real-time using an impedance-based biosensor. This change in cell morphology was prevented in the presence of the Na+/K+-ATPase inhibitor ouabain and EAAT inhibitors, which suggests the substrate-induced response was ion-dependent and transporter-specific. A mechanistic explanation for the phenotypic response was substantiated by actin cytoskeleton remodeling and changes in the intracellular levels of the osmolyte taurine, which suggests that the response involves cell swelling. In addition, substrate-induced cellular responses were observed for cells expressing other EAAT subtypes, as well as in a breast cancer cell line (MDA-MB-468) with endogenous EAAT1 expression. These findings allowed the development of a label-free high-throughput screening assay, which could be beneficial in early drug discovery for EAATs and holds potential for the study of other transport proteins that modulate cell shape.
Collapse
Affiliation(s)
- Hubert J. Sijben
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Laura Dall’ Acqua
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Rongfang Liu
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Abigail Jarret
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Svenja Onstein
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Simone J. Verburgt
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Laura H. Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
- Oncode Institute, Leiden, Netherlands
- *Correspondence: Laura H. Heitman,
| |
Collapse
|
29
|
Iovino L, Giusti V, Pischedda F, Giusto E, Plotegher N, Marte A, Battisti I, Di Iacovo A, Marku A, Piccoli G, Bandopadhyay R, Perego C, Bonifacino T, Bonanno G, Roseti C, Bossi E, Arrigoni G, Bubacco L, Greggio E, Hilfiker S, Civiero L. Trafficking of the glutamate transporter is impaired in LRRK2-related Parkinson's disease. Acta Neuropathol 2022; 144:81-106. [PMID: 35596783 PMCID: PMC9217889 DOI: 10.1007/s00401-022-02437-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022]
Abstract
The Excitatory Amino Acid Transporter 2 (EAAT2) accounts for 80% of brain glutamate clearance and is mainly expressed in astrocytic perisynaptic processes. EAAT2 function is finely regulated by endocytic events, recycling to the plasma membrane and degradation. Noteworthy, deficits in EAAT2 have been associated with neuronal excitotoxicity and neurodegeneration. In this study, we show that EAAT2 trafficking is impaired by the leucine-rich repeat kinase 2 (LRRK2) pathogenic variant G2019S, a common cause of late-onset familial Parkinson’s disease (PD). In LRRK2 G2019S human brains and experimental animal models, EAAT2 protein levels are significantly decreased, which is associated with elevated gliosis. The decreased expression of the transporter correlates with its reduced functionality in mouse LRRK2 G2019S purified astrocytic terminals and in Xenopus laevis oocytes expressing human LRRK2 G2019S. In LRRK2 G2019S knock-in mouse brain, the correct surface localization of the endogenous transporter is impaired, resulting in its interaction with a plethora of endo-vesicular proteins. Mechanistically, we report that pathogenic LRRK2 kinase activity delays the recycling of the transporter to the plasma membrane via Rabs inactivation, causing its intracellular re-localization and degradation. Taken together, our results demonstrate that pathogenic LRRK2 interferes with the physiology of EAAT2, pointing to extracellular glutamate overload as a possible contributor to neurodegeneration in PD.
Collapse
|
30
|
Nagaoka K, Kurauchi Y, Asano D, Morita A, Sakamoto K, Nakahara T. Pharmacological inhibition of Na +/K +-ATPase induces neurovascular degeneration and glial cell alteration in the rat retina. Exp Eye Res 2022; 220:109107. [PMID: 35568201 DOI: 10.1016/j.exer.2022.109107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/03/2022] [Accepted: 05/06/2022] [Indexed: 11/04/2022]
Abstract
Na+/K+-ATPase (NKA) plays an important role in ion homeostasis and neurotransmitter uptake. In the retina, multidirectional communications among neurons, glia, and blood vessels (that is, neuro-glio-vascular interaction) are crucial for maintaining tissue homeostasis. We investigated the role of NKA in the elements of neuro-glio-vascular unit in neonatal and adult rat retinas. Male Sprague-Dawley rats (1- and 8-week-old) were injected intravitreally with ouabain (20 nmol/eye), an inhibitor of NKA. Morphological changes in retinal neurons, glia, and blood vessels were examined. The intravitreal injection of ouabain decreased the number of cells in the ganglion cell layer, as well as the thicknesses of the inner plexiform and inner nuclear layers in neonatal and adult rats compared to age-matched controls. The ouabain-induced neuronal cell damage was partially prevented by D-(-)-2-amino-5-phosphonopentanoic acid, an antagonist of N-methyl-D-aspartic acid receptors. In the deep retinal vascular plexus of the ouabain-injected eyes, angiogenesis was delayed in neonatal rats, whereas capillary degeneration occurred in adult rats. The immunoreactivity of glutamine synthetase and vascular endothelial growth factor (VEGF) decreased in the retinas of neonatal and adult rats injected intravitreally with ouabain. The immunoreactivity of glial fibrillary acidic protein was enhanced in the retinas of ouabain-injected adult eyes. After the ouabain injection, CD45-positive leukocytes and Iba1-positive microglia increased in the inner retinal layer of neonatal rats, whereas they increased in the middle retinal layer of adult rats. These results suggest that the inhibition of NKA induces the degeneration of neuronal and vascular cells and alteration of glial cells in both neonatal and adult retinas. In addition to the direct effects of NKA inhibition, the disturbance of retinal glutamate metabolism and decreased VEGF expression may contribute to neurovascular degeneration. The activity of NKA is crucial for maintaining elements of neuro-glio-vascular unit in the retina.
Collapse
Affiliation(s)
- Koki Nagaoka
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oehonmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Daiki Asano
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
31
|
Ge Y, Zhen F, Liu Z, Feng Z, Wang G, Zhang C, Wang X, Sun Y, Zheng X, Bai Y, Yao R. Alpha-Asaronol Alleviates Dysmyelination by Enhancing Glutamate Transport Through the Activation of PPARγ-GLT-1 Signaling in Hypoxia-Ischemia Neonatal Rats. Front Pharmacol 2022; 13:766744. [PMID: 35401225 PMCID: PMC8984140 DOI: 10.3389/fphar.2022.766744] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/21/2022] [Indexed: 11/15/2022] Open
Abstract
Preterm white matter injury (PWMI) is the most common form of brain damage in premature infants caused by hypoxia-ischemia (HI), inflammation, or excitotoxicity. It is characterized by oligodendrocyte precursor cell (OPC) differentiation disorder and dysmyelination. Our previous study confirmed that alpha-asarone (α-asaronol), a major compound isolated from the Chinese medicinal herb Acorus gramineus by our lab, could alleviate neuronal overexcitation and improve the cognitive function of aged rats. In the present study, we investigated the effect and mechanism of α-asaronol on myelination in a rat model of PWMI induced by HI. Notably, α-asaronol promoted OPC differentiation and myelination in the corpus callosum of PWMI rats. Meanwhile, the concentration of glutamate was significantly decreased, and the levels of PPARγ and glutamate transporter 1 (GLT-1) were increased by α-asaronol treatment. In vitro, it was also confirmed that α-asaronol increased GLT-1 expression and recruitment of the PPARγ coactivator PCG-1a in astrocytes under oxygen and glucose deprivation (OGD) conditions. The PPARγ inhibitor GW9662 significantly reversed the effect of α-asaronol on GLT-1 expression and PCG-1a recruitment. Interestingly, the conditioned medium from α-asaronol-treated astrocytes decreased the number of OPCs and increased the number of mature oligodendrocytes. These results suggest that α-asaronol can promote OPC differentiation and relieve dysmyelination by regulating glutamate levels via astrocyte PPARγ-GLT-1 signaling. Although whether α-asaronol binds to PPARγ directly or indirectly is not investigated here, this study still indicates that α-asaronol may be a promising small molecular drug for the treatment of myelin-related diseases.
Collapse
Affiliation(s)
- Yuhang Ge
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Department of Human Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Fei Zhen
- Hongze Huaian District People's Hospital, Hongze, China
| | - Ziqi Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Zhaowei Feng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Gui Wang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Chu Zhang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Xingqi Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Ying Sun
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, China
| | - Xiaohui Zheng
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, China
| | - Yajun Bai
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
32
|
Gupta S, Bazargani N, Drew J, Howden JH, Modi S, Al Awabdh S, Marie H, Attwell D, Kittler JT. The non-adrenergic imidazoline-1 receptor protein nischarin is a key regulator of astrocyte glutamate uptake. iScience 2022; 25:104127. [PMID: 35434559 PMCID: PMC9010640 DOI: 10.1016/j.isci.2022.104127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 12/24/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Astrocytic GLT-1 is the main glutamate transporter involved in glutamate buffering in the brain, pivotal for glutamate removal at excitatory synapses to terminate neurotransmission and for preventing excitotoxicity. We show here that the surface expression and function of GLT-1 can be rapidly modulated through the interaction of its N-terminus with the nonadrenergic imidazoline-1 receptor protein, Nischarin. The phox domain of Nischarin is critical for interaction and internalization of surface GLT-1. Using live super-resolution imaging, we found that glutamate accelerated Nischarin-GLT-1 internalization into endosomal structures. The surface GLT-1 level increased in Nischarin knockout astrocytes, and this correlated with a significant increase in transporter uptake current. In addition, Nischarin knockout in astrocytes is neuroprotective against glutamate excitotoxicity. These data provide new molecular insights into regulation of GLT-1 surface level and function and suggest new drug targets for the treatment of neurological disorders. Nischarin phox domain interacts with the N-terminus of the glutamate transporter, GLT-1 Nischarin promotes internalization of GLT-1 to endosomes Glutamate modulates GLT-1 surface levels by regulating the Nischarin-GLT-1 interaction Nischarin loss enhances GLT-1 surface levels, transport currents, and neuroprotection
Collapse
Affiliation(s)
- Swati Gupta
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Narges Bazargani
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - James Drew
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Jack H. Howden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Souvik Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Sana Al Awabdh
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Hélène Marie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Josef T. Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
- Corresponding author
| |
Collapse
|
33
|
Suryavanshi P, Reinhart KM, Shuttleworth CW, Brennan KC. Action Potentials Are Critical for the Propagation of Focally Elicited Spreading Depolarizations. J Neurosci 2022; 42:2371-2383. [PMID: 34857650 PMCID: PMC8936615 DOI: 10.1523/jneurosci.2930-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/11/2023] Open
Abstract
Spreading depolarizations (SDs) of gray matter occur in the brain in different pathologic conditions, and cause varying degrees of tissue damage depending on the extent of metabolic burden on the tissue. As might be expected for such large depolarizations, neurons exhibit bursts of action potentials (APs) as the wave propagates. However, the specific role of APs in SD propagation is unclear. This is potentially consequential, since sodium channel modulation has not been considered as a therapeutic target for SD-associated disorders, because of ambiguous experimental evidence. Using whole-cell electrophysiology and single-photon imaging in acute cortical slices from male C57Bl6 mice, we tested the effects of AP blockade on SDs generated by two widely used induction paradigms. We found that AP blockade using tetrodotoxin (TTX) restricted propagation of focally induced SDs, and significantly reduced the amplitude of neuronal depolarization, as well as its Ca2+ load. TTX also abolished the suppression of spontaneous synaptic activity that is a hallmark of focally induced SD. In contrast, TTX did not affect the propagation of SD induced by global superfusion of high [K+]e containing artificial CSF (ACSF). Thus, we show that voltage-gated sodium channel (Nav)-mediated neuronal AP bursts are critical for the propagation and downstream effects of focally induced SD but are less important when the ionic balance of the extracellular space is already compromised. In doing so we corroborate the notion that two different SD induction paradigms, each relevant to different clinical situations, vary significantly in their characteristics and potentially their response to treatment.SIGNIFICANCE STATEMENT Our findings suggest that voltage-gated sodium channel (Nav) channels have a critical role in the propagation and downstream neural effects of focally induced spreading depolarization (SD). As SDs are likely induced focally in many disease conditions, these studies support sodium channel modulation, a previously underappreciated therapeutic option in SD-associated disorders, as a viable approach.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah 84108
- Interdepartmental Neuroscience Program, University of Utah School of Medicine, Salt Lake City, Utah 84108
| | - Katelyn M Reinhart
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah 84108
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131
| | - K C Brennan
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah 84108
| |
Collapse
|
34
|
Jakkamsetti V, Ma Q, Pascual JM. A subset of synaptic transmission events is coupled to acetyl coenzyme A production. J Neurophysiol 2022; 127:623-636. [PMID: 35080429 PMCID: PMC8897004 DOI: 10.1152/jn.00200.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Biological principles sustain the inference that synaptic function is coupled to neural metabolism, but the precise relationship between these two activities is not known. For example, it is unclear whether all synaptic transmission events are uniformly dependent on metabolic flux. Most synapses use glutamate, and the principal metabolic function of the brain is glucose oxidation, which starts with glycolysis. Thus, we asked how glutamatergic synaptic currents are modified by partial deficiency of the main glycolytic enzyme pyruvate dehydrogenase (PDH), which generates the intermediary metabolism product acetyl coenzyme A (acetyl-CoA). Using brain slices obtained from mice that were genetically modified to harbor a behaviorally relevant degree of PDH suppression, we also asked whether such impact is indeed metabolic via the bypassing of PDH with a glycolysis-independent acetyl-CoA substrate. We analyzed spontaneous synaptic currents under recording conditions that minimize artificial metabolic augmentation. Principal component analysis identified synaptic charge transfer as the major difference between a subset of wild-type and PDH-deficiency (PDHD) postsynaptic currents. This was due to reduced charge transfer as well as diminished current rise and decay times. The alternate acetyl-CoA source acetate rapidly restored these features but only for events of large amplitude as revealed by correlational and kernel density analyses. Application of tetrodotoxin to block large-amplitude events evoked by action potentials removed synaptic event charge transfer and decay-time differences between wild-type and PDHD neurons. These results suggest that glucose metabolic flux and excitatory transmission are intimately coupled for synaptic events characterized by large current amplitude.NEW & NOTEWORTHY In all tissues, metabolism and excitation are coupled but the details of this relationship remain elusive. Using a brain-targeted genetic approach in mice, reduction of pyruvate dehydrogenase, a major gateway in glucose metabolism, leads to changes that affect the synaptic event charge associated primarily with large excitatory (i.e., glutamate mediated) synaptic potentials. This can be modified in the direction of normal using the alternative fuel acetate, indicating that this phenomenon depends on rapid metabolic flux.
Collapse
Affiliation(s)
- Vikram Jakkamsetti
- 1Rare Brain Disorders Program, Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Qian Ma
- 1Rare Brain Disorders Program, Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Juan M. Pascual
- 1Rare Brain Disorders Program, Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, Texas,2Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas,3Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas,4Eugene McDermott Center for Human Growth & Development/Center for Human Genetics, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
35
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
36
|
Role of Na +/K +-ATPase in ischemic stroke: in-depth perspectives from physiology to pharmacology. J Mol Med (Berl) 2021; 100:395-410. [PMID: 34839371 DOI: 10.1007/s00109-021-02143-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022]
Abstract
Na+/K+-ATPase (NKA) is a large transmembrane protein expressed in all cells. It is well studied for its ion exchanging function, which is indispensable for the maintenance of electrochemical gradients across the plasma membrane and herein neuronal excitability. The widely recognized pump function of NKA closely depends on its unique structure features and conformational changes upon binding of specific ions. Various Na+-dependent secondary transport systems are rigorously controlled by the ionic gradients generated by NKA and are essential for multiple physiological processes. In addition, roles of NKA as a signal transducer have also been unveiled nowadays. Plethora of signaling cascades are defined including Src-Ras-MAPK signaling, IP3R-mediated calcium oscillation, inflammation, and autophagy though most underlying mechanisms remain elusive. Ischemic stroke occurs when the blood flow carrying nutrients and oxygen into the brain is disrupted by blood clots, which is manifested by excitotoxicity, oxidative stress, inflammation, etc. The protective effect of NKA against ischemic stress is emerging gradually with the application of specific NKA inhibitor. However, NKA-related research is limited due to the opposite effects caused by NKA inhibitor at lower doses. The present review focuses on the recent progression involving different aspects about NKA in cellular homeostasis to present an in-depth understanding of this unique protein. Moreover, essential roles of NKA in ischemic pathology are discussed to provide a platform and bright future for the improvement in clinical research on ischemic stroke.
Collapse
|
37
|
Vizuete AFK, Mussulini BH, Zenki KC, Baggio S, Pasqualotto A, Rosemberg DB, Bogo MR, de Oliveira DL, Rico EP. Prolonged ethanol exposure alters glutamate uptake leading to astrogliosis and neuroinflammation in adult zebrafish brain. Neurotoxicology 2021; 88:57-64. [PMID: 34728274 DOI: 10.1016/j.neuro.2021.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/04/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
High ethanol (EtOH) consumption is a serious condition that induces tremors, alcoholic psychosis, and delirium, being considered a public health problem worldwide. Prolonged EtOH exposure promotes neurodegeneration, affecting several neurotransmitter systems and transduction signaling pathways. Glutamate is the major excitatory amino acid in the central nervous system (CNS) and the extracellular glutamatergic tonus is controlled by glutamate transporters mostly located in astrocytes. Here, we explore the effects of prolonged EtOH exposure on the glutamatergic uptake system and its relationship with astroglial markers (GFAP and S100B), neuroinflammation (IL-1β and TNF-α), and brain derived neurotrophic factor (BDNF) levels in the CNS of adult zebrafish. Animals were exposed to 0.5% EtOH for 7, 14, and 28 days continuously. Glutamate uptake was significantly decreased after 7 and 14 days of EtOH exposure, returning to baseline levels after 28 days of exposure. No alterations were observed in crucial enzymatic activities linked to glutamate uptake, like Na,K-ATPase or glutamine synthetase. Prolonged EtOH exposure increased GFAP, S100B, and TNF-α levels after 14 days. Additionally, increased BDNF mRNA levels were observed after 14 and 28 days of EtOH exposure, while BDNF protein levels increased only after 28 days. Collectively, our data show markedly brain astroglial, neuroinflammatory and neurotrofic responses after an initial impairment of glutamate uptake following prolonged EtOH exposure. This neuroplasticity event could play a key role in the modulatory effect of EtOH on glutamate uptake after 28 days of continuous exposure.
Collapse
Affiliation(s)
- Adriana Fernanda Kuckartz Vizuete
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Ben Hur Mussulini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Kamila Cagliari Zenki
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Suelen Baggio
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Amanda Pasqualotto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Denis Broock Rosemberg
- Programa de Pós-Graduação em Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, RS, 97105-900, Santa Maria, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA
| | - Maurício Reis Bogo
- Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Pontifícia Universidade Católica do Rio Grande do Sul, Brazil
| | - Diogo Lösch de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Extreme Southern Catarinense (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
38
|
Extracellular Glutamate Concentration Increases Linearly in Proportion to Decreases in Residual Cerebral Blood Flow After the Loss of Membrane Potential in a Rat Model of Ischemia. J Neurosurg Anesthesiol 2021; 33:356-362. [PMID: 31834249 DOI: 10.1097/ana.0000000000000666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/26/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Brain ischemia due to disruption of cerebral blood flow (CBF) results in increases in extracellular glutamate concentration and neuronal cell damage. However, the impact of CBF on glutamate dynamics after the loss of the membrane potential remains unknown. MATERIALS AND METHODS To determine this impact, we measured extracellular potential, CBF, and extracellular glutamate concentration in the parietal cortex in male Sprague-Dawley rats (n=21). CBF was reduced by bilateral occlusion of the common carotid arteries and exsanguination until loss of extracellular membrane potential was observed (low-flow group), or until CBF was further reduced by 5% to 10% of preischemia levels (severe-low-flow group). CBF was promptly restored 10 minutes after the loss of membrane potential. Histologic outcomes were evaluated 5 days later. RESULTS Extracellular glutamate concentration in the low-flow group was significantly lower than that in the severe-low-flow group. Moreover, increases in extracellular glutamate concentration exhibited a linear relationship with decreases in CBF after the loss of membrane potential in the severe-low-flow group, and the percentage of damaged neurons exhibited a dose-response relationship with the extracellular glutamate concentration. The extracellular glutamate concentration required to cause 50% neuronal damage was estimated to be 387 μmol/L, at 8.7% of preischemia CBF. Regression analyses revealed that extracellular glutamate concentration increased by 21 μmol/L with each 1% decrease in residual CBF and that the percentage of damaged neurons increased by 2.6%. CONCLUSION Our results indicate that residual CBF is an important factor that determines the extracellular glutamate concentration after the loss of membrane potential, and residual CBF would be one of the important determinants of neuronal cell prognosis.
Collapse
|
39
|
Ma K, Zhang A, She X, Yang H, Wang K, Zhu Y, Gao X, Cui B. Disruption of Glutamate Release and Uptake-Related Protein Expression After Noise-Induced Synaptopathy in the Cochlea. Front Cell Dev Biol 2021; 9:720902. [PMID: 34422838 PMCID: PMC8373299 DOI: 10.3389/fcell.2021.720902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/14/2021] [Indexed: 02/03/2023] Open
Abstract
High-intensity noise can cause permanent hearing loss; however, short-duration medium-intensity noise only induces a temporary threshold shift (TTS) and damages synapses formed by inner hair cells (IHCs) and spiral ganglion nerves. Synaptopathy is generally thought to be caused by glutamate excitotoxicity. In this study, we investigated the expression levels of vesicle transporter protein 3 (Vglut3), responsible for the release of glutamate; glutamate/aspartate transporter protein (GLAST), responsible for the uptake of glutamate; and Na+/K+-ATPase α1 coupled with GLAST, in the process of synaptopathy in the cochlea. The results of the auditory brainstem response (ABR) and CtBP2 immunofluorescence revealed that synaptopathy was induced on day 30 after 100 dB SPL noise exposure in C57BL/6J mice. We found that GLAST and Na+/K+-ATPase α1 were co-localized in the cochlea, mainly in the stria vascularis, spiral ligament, and spiral ganglion cells. Furthermore, Vglut3, GLAST, and Na+/K+-ATPase α1 expression were disrupted after noise exposure. These results indicate that disruption of glutamate release and uptake-related protein expression may exacerbate the occurrence of synaptopathy.
Collapse
Affiliation(s)
- Kefeng Ma
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Anran Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.,Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yingwen Zhu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.,Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
40
|
Belo do Nascimento I, Damblon J, Ingelbrecht C, Goursaud S, Massart M, Dumont A, Desmet N, Hermans E. Pharmacological evidence for the concept of spare glutamate transporters. Neurochem Int 2021; 149:105142. [PMID: 34314789 DOI: 10.1016/j.neuint.2021.105142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 01/30/2023]
Abstract
Through the efficient clearance of extracellular glutamate, high affinity astrocytic glutamate transporters constantly shape excitatory neurotransmission in terms of duration and spreading. Even though the glutamate transporter GLT-1 (also known as EAAT2/SLC1A2) is amongst the most abundant proteins in the mammalian brain, its density and activity are tightly regulated. In order to study the influence of changes in the expression of GLT-1 on glutamate uptake capacity, we have developed a model in HEK cells where the density of the transporter can be manipulated thanks to a tetracycline-inducible promoter. Exposing the cells to doxycycline concentration-dependently increased GLT-1 expression and substrate uptake velocity. However, beyond a certain level of induction, increasing the density of transporters at the cell surface failed to increase the maximal uptake. This suggested the progressive generation of a pool of spare transporters, a hypothesis that was further validated using the selective GLT-1 blocker WAY-213613 of which potency was influenced by the density of the transporters. The curve showing inhibition of uptake by increasing concentrations of WAY-213613 was indeed progressively rightward shifted when tested in cells where the transporter density was robustly induced. As largely documented in the context of cell-surface receptors, the existence of 'spare' glutamate transporters in the nervous tissue and particularly in astrocytes could impact on the consequences of physiological or pathological regulation of these transporters.
Collapse
Affiliation(s)
- Inês Belo do Nascimento
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Jonathan Damblon
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Caroline Ingelbrecht
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Stéphanie Goursaud
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Marion Massart
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Amélie Dumont
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Nathalie Desmet
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium.
| |
Collapse
|
41
|
Parker PD, Suryavanshi P, Melone M, Sawant-Pokam PA, Reinhart KM, Kaufmann D, Theriot JJ, Pugliese A, Conti F, Shuttleworth CW, Pietrobon D, Brennan KC. Non-canonical glutamate signaling in a genetic model of migraine with aura. Neuron 2021; 109:611-628.e8. [PMID: 33321071 PMCID: PMC7889497 DOI: 10.1016/j.neuron.2020.11.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 10/09/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022]
Abstract
Migraine with aura is a common but poorly understood sensory circuit disorder. Monogenic models allow an opportunity to investigate its mechanisms, including spreading depolarization (SD), the phenomenon underlying migraine aura. Using fluorescent glutamate imaging, we show that awake mice carrying a familial hemiplegic migraine type 2 (FHM2) mutation have slower clearance during sensory processing, as well as previously undescribed spontaneous "plumes" of glutamate. Glutamatergic plumes overlapped anatomically with a reduced density of GLT-1a-positive astrocyte processes and were mimicked in wild-type animals by inhibiting glutamate clearance. Plume pharmacology and plume-like neural Ca2+ events were consistent with action-potential-independent spontaneous glutamate release, suggesting plumes are a consequence of inefficient clearance following synaptic release. Importantly, a rise in basal glutamate and plume frequency predicted the onset of SD in both FHM2 and wild-type mice, providing a novel mechanism in migraine with aura and, by extension, the other neurological disorders where SD occurs.
Collapse
Affiliation(s)
- Patrick D Parker
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Pratyush Suryavanshi
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy; Center for Neurobiology of Aging, IRCCS INRCA, Ancona 60020, Italy
| | - Punam A Sawant-Pokam
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Katelyn M Reinhart
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Dan Kaufmann
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Jeremy J Theriot
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Arianna Pugliese
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy; Center for Neurobiology of Aging, IRCCS INRCA, Ancona 60020, Italy; Foundation for Molecular Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, 35131 Padova, Italy; CNR Institute of Neuroscience, 35131 Padova, Italy.
| | - K C Brennan
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84108, USA.
| |
Collapse
|
42
|
Lopachev AV, Lagarkova MA, Lebedeva OS, Ezhova MA, Kazanskaya RB, Timoshina YA, Khutorova AV, Akkuratov EE, Fedorova TN, Gainetdinov RR. Ouabain-Induced Gene Expression Changes in Human iPSC-Derived Neuron Culture Expressing Dopamine and cAMP-Regulated Phosphoprotein 32 and GABA Receptors. Brain Sci 2021; 11:brainsci11020203. [PMID: 33562186 PMCID: PMC7915459 DOI: 10.3390/brainsci11020203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiotonic steroids (CTS) are specific inhibitors and endogenous ligands of a key enzyme in the CNS-the Na+, K+-ATPase, which maintains and creates an ion gradient on the plasma membrane of neurons. CTS cause the activation of various signaling cascades and changes in gene expression in neurons and other cell types. It is known that intracerebroventricular injection of cardiotonic steroid ouabain causes mania-like behavior in rodents, in part due to activation of dopamine-related signaling cascades in the dopamine and cAMP-regulated phosphoprotein 32 (DARPP-32) expressing medium spiny neurons in the striatum. Dopaminergic projections in the striatum innervate these GABAergic medium spiny neurons. The objective of this study was to assess changes in the expression of all genes in human iPSC-derived expressing DARPP-32 and GABA receptors neurons under the influence of ouabain. We noted a large number of statistically significant upregulated and downregulated genes after a 16-h incubation with non-toxic concentration (30 nM) of ouabain. These changes in the transcriptional activity were accomplished with activation of MAP-kinase ERK1/2 and transcriptional factor cAMP response element-binding protein (CREB). Thus, it can be concluded that 30 nM ouabain incubated for 16 h with human iPSC-derived expressing DARPP-32 and GABA receptors neurons activates genes associated with neuronal maturation and synapse formation, by increasing the expression of genes associated with translation, vesicular transport, and increased electron transport chain function. At the same time, the expression of genes associated with proliferation, migration, and early development of neurons decreases. These data indicate that non-toxic concentrations of ouabain may induce neuronal maturation, neurite growth, and increased synaptogenesis in dopamine-receptive GABAergic neurons, suggesting formation of plasticity and the establishment of new neuronal junctions.
Collapse
Affiliation(s)
- Alexander V. Lopachev
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Correspondence:
| | - Maria A. Lagarkova
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine Federal Medical Biological Agency, 119435 Moscow, Russia; (M.A.L.); (O.S.L.)
| | - Olga S. Lebedeva
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine Federal Medical Biological Agency, 119435 Moscow, Russia; (M.A.L.); (O.S.L.)
| | - Margarita A. Ezhova
- Laboratory of Plant Genomics, Institute for Information Transmission Problems of the Russian Academy of Sciences, 127051 Moscow, Russia;
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Rogneda B. Kazanskaya
- Biological Department, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| | - Yulia A. Timoshina
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Biological Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anastasiya V. Khutorova
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Biological Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Evgeny E. Akkuratov
- Department of Applied Physics, Royal Institute of Technology, Science for Life Laboratory, 171 65 Stockholm, Sweden;
| | - Tatiana N. Fedorova
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine and Saint Petersburg University Hospital, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| |
Collapse
|
43
|
Decreased content of ascorbic acid (vitamin C) in the brain of knockout mouse models of Na+,K+-ATPase-related neurologic disorders. PLoS One 2021; 16:e0246678. [PMID: 33544780 PMCID: PMC7864419 DOI: 10.1371/journal.pone.0246678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/23/2021] [Indexed: 12/21/2022] Open
Abstract
Na+,K+-ATPase is a crucial protein responsible for maintaining the electrochemical gradients across the cell membrane. The Na+,K+-ATPase is comprised of catalytic α, β, and γ subunits. In adult brains, the α3 subunit, encoded by ATP1A3, is predominantly expressed in neurons, whereas the α2 subunit, encoded by ATP1A2, is expressed in glial cells. In foetal brains, the α2 is expressed in neurons as well. Mutations in α subunits cause a variety of neurologic disorders. Notably, the onset of symptoms in ATP1A2- and ATP1A3-related neurologic disorders is usually triggered by physiological or psychological stressors. To gain insight into the distinct roles of the α2 and α3 subunits in the developing foetal brain, whose developmental dysfunction may be a predisposing factor of neurologic disorders, we compared the phenotypes of mouse foetuses with double homozygous knockout of Atp1a2 and Atp1a3 (α2α3-dKO) to those with single knockout. The brain haemorrhage phenotype of α2α3-dKO was similar to that of homozygous knockout of the gene encoding ascorbic acid (ASC or vitamin C) transporter, SVCT2. The α2α3-dKO brain showed significantly decreased level of ASC compared with the wild-type (WT) and single knockout. We found that the ASC content in the basal ganglia and cerebellum was significantly lower in the adult Atp1a3 heterozygous knockout mouse (α3-HT) than in the WT. Interestingly, we observed a significant decrease in the ASC level in the basal ganglia and cerebellum of α3-HT in the peripartum period, during which mice are under physiological stress. These observations indicate that the α2 and α3 subunits independently contribute to the ASC level in the foetal brain and that the α3 subunit contributes to ASC transport in the adult basal ganglia and cerebellum. We propose that decreases in ASC levels may affect neural network development and are linked to the pathophysiology of ATP1A2- and ATP1A3-related neurologic disorders.
Collapse
|
44
|
Li LL, Ke XY, Jiang C, Qin SQ, Liu YY, Xian XH, Liu LZ, He JC, Chen YM, An HF, Sun N, Hu YH, Wang Y, Zhang LN, Lu QY. Na + , K + -ATPase participates in the protective mechanism of rat cerebral ischemia-reperfusion through the interaction with glutamate transporter-1. Fundam Clin Pharmacol 2021; 35:870-881. [PMID: 33481320 DOI: 10.1111/fcp.12652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/19/2021] [Indexed: 11/30/2022]
Abstract
Glutamate excitotoxicity in cerebral ischemia/reperfusion is an important cause of neurological damage. The aim of this study was to investigate the mechanism of Na+, K+-ATPase (NKA) involved in l ow concentration of ouabain (Oua, activating NKA)-induced protection of rat cerebral ischemia-reperfusion injury. The 2,3,5-triphenyltetrazolium chloride (TTC) staining and neurological deficit scores (NDS) were performed to evaluate rat cerebral injury degree respectively at 2 h, 6 h, 1 d and 3 d after reperfusion of middle cerebral artery occlusion (MCAO) 2 h in rats. NKA α1/α2 subunits and glutamate transporter-1 (GLT-1) protein expression were investigated by Western blotting. The cerebral infarct volume ratio were evidently decreased in Oua group vs MCAO/R group at 1 d and 3 d after reperfusion of 2 h MCAO in rats (*p < 0.05 ). Moreover, NDS were not significantly different (p > 0.05 ). NKA α1 was decreased at 6 h and 1 d after reperfusion of 2 h MCAO in rats, and was improved in Oua group. However, NKA α1 and α2 were increased at 3 d after reperfusion of 2 h MCAO in rats, and was decreased in Oua group. GLT-1 was decreased at 6 h, 1 d and 3 d after reperfusion of 2 h MCAO in rats, and was improved in Oua group. These data indicated that l ow concentration of Oua could improve MCAO/R injury through probably changing NKA α1/α2 and GLT-1 protein expression, then increasing GLT-1 function and promoting Glu transport and absorption, which could be useful to determine potential therapeutic strategies for patients with stroke. Low concentration of Oua improved rat MCAO/R injury via NKA α1/α2 and GLT-1.
Collapse
Affiliation(s)
- Lin-Lin Li
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Xue-Ying Ke
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Chen Jiang
- Forensic Medical College, Hebei Medical University, Hebei, China
| | - Shi-Qi Qin
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yang-Yang Liu
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Li-Zhe Liu
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Jin-Chen He
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Ya-Meng Chen
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Hong-Fei An
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Nan Sun
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yue-Hua Hu
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yan Wang
- North China University of Science and Technology Affiliated Hospital, Hebei, China
| | - Li-Nan Zhang
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Qi-Yong Lu
- Department of Neurosurgery, Hengshui Fifth People's Hospital, Hebei, China
| |
Collapse
|
45
|
Mirsaeed-Ghazi F, Sharifzadeh M, Ashrafi-Kooshk MR, Karima S, Meknatkhah S, Riazi G, Mokhtari F. Astaxanthin Decreases Spatial Memory and Glutamate Transport Impairment Induced by Fluoride. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:238-254. [PMID: 35194443 PMCID: PMC8842617 DOI: 10.22037/ijpr.2021.114919.15107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Excessive exposure to the sources of fluoride in drinking water, oral care products, and food is a widespread problem. Fluoride is associated with impairment in child intelligence development. It causes DNA damage, oxidative stress, and mitochondrial dysfunction, mainly due to the production of reactive oxygen species (ROS). It has been postulated that the use of antioxidants such as astaxanthin, may alleviate fluoride's adverse effects. This study assessed the effects of fluoride on cellular ROS content and rat's learning and memory ability and investigated the protective potency of astaxanthin with emphasis on the role of glutamate using the Morris Water Maze test, glutamate concentration determination, and western blot techniques. The fluoride treatment of cells results in an increment of cellular ROS, whereas astaxanthin inhibits lipid peroxidation. Fluoride significantly decreases the cellular glutamate uptake and glutamate transporter, protein level, possibly due to the disruption of mitochondrial energy metabolism and defect of the transporter recycle, respectively. The in-vivo study indicated that the treatment of rats with fluoride led to a loss of learning, while astaxanthin improved memory dysfunction. Measurement of ROS and glutamate levels of rat brain hippocampus showed that fluoride increased the ROS but decreased the glutamate. On the other hand, the utilization of astaxanthin decreased the brain ROS content and increased the glutamate level. It seems that fluoride disrupts the normal function of neurons via increment of ROS production and decrement of glutamate level, whereas astaxanthin has neuroprotective potency due to the ROS scavenging ability.
Collapse
Affiliation(s)
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sogol Meknatkhah
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Gholamhossein Riazi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.,Corresponding author: E-mail:
| | | |
Collapse
|
46
|
Smith SE, Chen X, Brier LM, Bumstead JR, Rensing NR, Ringel AE, Shin H, Oldenborg A, Crowley JR, Bice AR, Dikranian K, Ippolito JE, Haigis MC, Papouin T, Zhao G, Wong M, Culver JP, Bonni A. Astrocyte deletion of α2-Na/K ATPase triggers episodic motor paralysis in mice via a metabolic pathway. Nat Commun 2020; 11:6164. [PMID: 33268780 PMCID: PMC7710756 DOI: 10.1038/s41467-020-19915-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Familial hemiplegic migraine is an episodic neurological disorder characterized by transient sensory and motor symptoms and signs. Mutations of the ion pump α2-Na/K ATPase cause familial hemiplegic migraine, but the mechanisms by which α2-Na/K ATPase mutations lead to the migraine phenotype remain incompletely understood. Here, we show that mice in which α2-Na/K ATPase is conditionally deleted in astrocytes display episodic paralysis. Functional neuroimaging reveals that conditional α2-Na/K ATPase knockout triggers spontaneous cortical spreading depression events that are associated with EEG low voltage activity events, which correlate with transient motor impairment in these mice. Transcriptomic and metabolomic analyses show that α2-Na/K ATPase loss alters metabolic gene expression with consequent serine and glycine elevation in the brain. A serine- and glycine-free diet rescues the transient motor impairment in conditional α2-Na/K ATPase knockout mice. Together, our findings define a metabolic mechanism regulated by astrocytic α2-Na/K ATPase that triggers episodic motor paralysis in mice.
Collapse
Affiliation(s)
- Sarah E Smith
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
- MD-PhD Program, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiaoying Chen
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lindsey M Brier
- MD-PhD Program, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jonathan R Bumstead
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
| | - Nicholas R Rensing
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alison E Ringel
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Haewon Shin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anna Oldenborg
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jan R Crowley
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Annie R Bice
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Krikor Dikranian
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph E Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph P Culver
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
- Department of Physics, Washington University in St. Louis, St. Louis, MO, 63105, USA
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
47
|
Early Postnatal Exposure to a Low Dose of Nanoparticulate Silver Induces Alterations in Glutamate Transporters in Brain of Immature Rats. Int J Mol Sci 2020; 21:ijms21238977. [PMID: 33256007 PMCID: PMC7730297 DOI: 10.3390/ijms21238977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Due to strong antimicrobial properties, silver nanoparticles (AgNPs) are used in a wide range of medical and consumer products, including those dedicated for infants and children. While AgNPs are known to exert neurotoxic effects, current knowledge concerning their impact on the developing brain is scarce. During investigations of mechanisms of neurotoxicity in immature rats, we studied the influence of AgNPs on glutamate transporter systems which are involved in regulation of extracellular concentration of glutamate, an excitotoxic amino acid, and compared it with positive control—Ag citrate. We identified significant deposition of AgNPs in brain tissue of exposed rats over the post-exposure time. Ultrastructural alterations in endoplasmic reticulum (ER) and Golgi complexes were observed in neurons of AgNP-exposed rats, which are characteristics of ER stress. These changes presumably underlie substantial long-lasting downregulation of neuronal glutamate transporter EAAC1, which was noted in AgNP-exposed rats. Conversely, the expression of astroglial glutamate transporters GLT-1 and GLAST was not affected by exposure to AgNPs, but the activity of the transporters was diminished. These results indicate that even low doses of AgNPs administered during an early stage of life create a substantial risk for health of immature organisms. Hence, the safety of AgNP-containing products for infants and children should be carefully considered.
Collapse
|
48
|
Shao LR, Janicot R, Stafstrom CE. Na +-K +-ATPase functions in the developing hippocampus: regional differences in CA1 and CA3 neuronal excitability and role in epileptiform network bursting. J Neurophysiol 2020; 125:1-11. [PMID: 33206576 DOI: 10.1152/jn.00453.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Na+-K+-ATPase (Na+-K+ pump) is essential for setting resting membrane potential and restoring transmembrane Na+ and K+ gradients after neuronal firing, yet its roles in developing neurons are not well understood. This study examined the contribution of the Na+-K+ pump to resting membrane potential and membrane excitability of developing CA1 and CA3 neurons and its role in maintaining synchronous network bursting. Experiments were conducted in postnatal day (P)9 to P13 rat hippocampal slices using whole cell patch-clamp and extracellular field-potential recordings. Blockade of the Na+-K+ pump with strophanthidin caused marked depolarization (23.1 mV) in CA3 neurons but only a modest depolarization (3.3 mV) in CA1 neurons. Regarding other membrane properties, strophanthidin differentially altered the voltage-current responses, input resistance, action-potential threshold and amplitude, rheobase, and input-output relationship in CA3 vs. CA1 neurons. At the network level, strophanthidin stopped synchronous epileptiform bursting in CA3 induced by 0 Mg2+ and 4-aminopyridine. Furthermore, dual whole cell recordings revealed that strophanthidin disrupted the synchrony of CA3 neuronal firing. Finally, strophanthidin reduced spontaneous excitatory postsynaptic current (sEPSC) bursts (i.e., synchronous transmitter release) and transformed them into individual sEPSC events (i.e., nonsynchronous transmitter release). These data suggest that the Na+-K+ pump plays a more profound role in membrane excitability in developing CA3 neurons than in CA1 neurons and that the pump is essential for the maintenance of synchronous network bursting in CA3. Compromised Na+-K+ pump function leads to cessation of ongoing synchronous network activity, by desynchronizing neuronal firing and neurotransmitter release in the CA3 synaptic network. These findings have implications for the regulation of network excitability and seizure generation in the developing brain.NEW & NOTEWORTHY Despite the extensive literature showing the importance of the Na+-K+ pump in various neuronal functions, its roles in the developing brain are not well understood. This study reveals that the Na+-K+ pump differentially regulates the excitability of CA3 and CA1 neurons in the developing hippocampus, and the pump activity is crucial for maintaining network activity. Compromised Na+-K+ pump activity desynchronizes neuronal firing and transmitter release, leading to cessation of ongoing epileptiform network bursting.
Collapse
Affiliation(s)
- Li-Rong Shao
- Division of Pediatric Neurology, Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Remi Janicot
- Division of Pediatric Neurology, Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
49
|
Zeng H, Zhang X, Wang W, Shen Z, Dai Z, Yu Z, Xu S, Yan G, Huang Q, Wu R, Chen X, Xu H. Maternal separation with early weaning impairs neuron-glia integrity: non-invasive evaluation and substructure demonstration. Sci Rep 2020; 10:19440. [PMID: 33173142 PMCID: PMC7656452 DOI: 10.1038/s41598-020-76640-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/29/2020] [Indexed: 02/05/2023] Open
Abstract
Astrocytes and oligodendrocytes play essential roles in regulating neural signal transduction along neural circuits in CNS. The perfect coordination of neuron/astrocyte and neuron/oligodendrocyte entities was termed as neuron-glia integrity recently. Here we monitored the status of neuron-glia integrity via non-invasive neuroimaging methods and demonstrated the substructures of it using other approaches in an animal model of maternal separation with early weaning (MSEW), which mimics early life neglect and abuse in humans. Compared to controls, MSEW rats showed higher glutamate level, but lower GABA in prefrontal cortex (PFC) detected by chemical exchange saturation transfer and 1H-MRS methods, lower levels of glial glutamate transporter-1 and ATP-α, but increased levels of glutamate decarboxylase-65 and glutamine synthetase in PFC; reduced fractional anisotropy in various brain regions revealed by diffusion tensor imaging, along with increased levels of N-acetyl-aspartate measured by 1H-MRS; and hypomyelination in PFC as evidenced by relevant cellular and molecular changes.
Collapse
Affiliation(s)
- Haiyan Zeng
- The Mental Health Center, Shantou University Medical College, Shantou, China
- Xianyue Hospital/Xiamen Mental Health Center, Xiamen, China
| | - Xiaolei Zhang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Wenqiang Wang
- Xianyue Hospital/Xiamen Mental Health Center, Xiamen, China
| | - Zhiwei Shen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhuozhi Dai
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhijia Yu
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Shuqin Xu
- Department of Anatomy, Shantou University Medical College, Shantou, China
| | - Gen Yan
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Qingjun Huang
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Renhua Wu
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xi Chen
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, USA
| | - Haiyun Xu
- The Mental Health Center, Shantou University Medical College, Shantou, China.
- Department of Anatomy, Shantou University Medical College, Shantou, China.
- The School of Psychiatry, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
50
|
Rose J, Brian C, Pappa A, Panayiotidis MI, Franco R. Mitochondrial Metabolism in Astrocytes Regulates Brain Bioenergetics, Neurotransmission and Redox Balance. Front Neurosci 2020; 14:536682. [PMID: 33224019 PMCID: PMC7674659 DOI: 10.3389/fnins.2020.536682] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/14/2020] [Indexed: 01/17/2023] Open
Abstract
In the brain, mitochondrial metabolism has been largely associated with energy production, and its dysfunction is linked to neuronal cell loss. However, the functional role of mitochondria in glial cells has been poorly studied. Recent reports have demonstrated unequivocally that astrocytes do not require mitochondria to meet their bioenergetics demands. Then, the question remaining is, what is the functional role of mitochondria in astrocytes? In this work, we review current evidence demonstrating that mitochondrial central carbon metabolism in astrocytes regulates overall brain bioenergetics, neurotransmitter homeostasis and redox balance. Emphasis is placed in detailing carbon source utilization (glucose and fatty acids), anaplerotic inputs and cataplerotic outputs, as well as carbon shuttles to neurons, which highlight the metabolic specialization of astrocytic mitochondria and its relevance to brain function.
Collapse
Affiliation(s)
- Jordan Rose
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Christian Brian
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Electron Microscopy & Molecular Pathology, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|