1
|
Chauhan R, Mohan M, Mannan A, Devi S, Singh TG. Unravelling the role of Interleukin-12 in Neuroinflammatory mechanisms: Pathogenic pathways linking Neuroinflammation to neuropsychiatric disorders. Int Immunopharmacol 2025; 156:114654. [PMID: 40294470 DOI: 10.1016/j.intimp.2025.114654] [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: 12/12/2024] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Neuropsychiatric disorders are clinically characterized conditions involving both neurology and psychiatry, arising from dysfunctioning of cerebral function, or indirect effects of extra cerebral disease. Neuropsychiatric disorders tend to influence emotions, mood, and brain functioning. Growing evidence indicates that the etiology of these disorders is not confined to neuronal abnormalities but extends to include inflammation. While the underlying mechanism of these disorders is still in its infancy, recent data highlights the significant role of neuroinflammation in their pathophysiology. Neuroinflammation concerns the inflammation within the neural tissue characterized by alteration in astrocytes, cytokines, microglia, and chemokines within the central nervous system. The cytokines include IFN-γ, IL-1β, IL-2, IL4, IL-6, IL-8, IL-10, and IL-12. This review focuses on interleukin-12 (IL-12), a key mediator of neuroinflammation, and its potential involvement in neuropsychiatric disorders. IL-12 promotes neuroinflammation and influences neurotransmitter systems. Additionally, it also affects the HPA axis, impairs neuroplasticity, and activates microglia by interacting with TLR, JAK-STAT, PI3K/Akt, GSK-3, NMDA, MAPK, PKC, VEGFR, ROCK, and Wnt signaling pathways and elicit its role in ND. In this review, we dwell on the current evidence supporting IL-12's pathogenic role and explore the possible mechanisms by which it contributes to the development and progression of these conditions. This review aims to provide insights that may aid in future therapeutic strategies by illuminating the interplay between neuroinflammation and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rupali Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
2
|
Escamilla S, Sáez-Valero J, Cuchillo-Ibáñez I. NMDARs in Alzheimer's Disease: Between Synaptic and Extrasynaptic Membranes. Int J Mol Sci 2024; 25:10220. [PMID: 39337704 PMCID: PMC11431980 DOI: 10.3390/ijms251810220] [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: 09/02/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate receptors with key roles in synaptic communication and plasticity. The activation of synaptic NMDARs initiates plasticity and stimulates cell survival. In contrast, the activation of extrasynaptic NMDARs can promote cell death underlying a potential mechanism of neurodegeneration occurring in Alzheimer's disease (AD). The distribution of synaptic versus extrasynaptic NMDARs has emerged as an important parameter contributing to neuronal dysfunction in neurodegenerative diseases including AD. Here, we review the concept of extrasynaptic NMDARs, as this population is present in numerous neuronal cell membranes but also in the membranes of various non-neuronal cells. Previous evidence regarding the membranal distribution of synaptic versus extrasynaptic NMDRs in relation to AD mice models and in the brains of AD patients will also be reviewed.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Inmaculada Cuchillo-Ibáñez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
3
|
Mollinari C, Cardinale A, Lupacchini L, Martire A, Chiodi V, Martinelli A, Rinaldi AM, Fini M, Pazzaglia S, Domenici MR, Garaci E, Merlo D. The DNA repair protein DNA-PKcs modulates synaptic plasticity via PSD-95 phosphorylation and stability. EMBO Rep 2024; 25:3707-3737. [PMID: 39085642 PMCID: PMC11315936 DOI: 10.1038/s44319-024-00198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The key DNA repair enzyme DNA-PKcs has several and important cellular functions. Loss of DNA-PKcs activity in mice has revealed essential roles in immune and nervous systems. In humans, DNA-PKcs is a critical factor for brain development and function since mutation of the prkdc gene causes severe neurological deficits such as microcephaly and seizures, predicting yet unknown roles of DNA-PKcs in neurons. Here we show that DNA-PKcs modulates synaptic plasticity. We demonstrate that DNA-PKcs localizes at synapses and phosphorylates PSD-95 at newly identified residues controlling PSD-95 protein stability. DNA-PKcs -/- mice are characterized by impaired Long-Term Potentiation (LTP), changes in neuronal morphology, and reduced levels of postsynaptic proteins. A PSD-95 mutant that is constitutively phosphorylated rescues LTP impairment when over-expressed in DNA-PKcs -/- mice. Our study identifies an emergent physiological function of DNA-PKcs in regulating neuronal plasticity, beyond genome stability.
Collapse
Affiliation(s)
- Cristiana Mollinari
- Istituto Superiore di Sanita', Department of Neuroscience, 00161, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, 00133, Rome, Italy
| | | | | | - Alberto Martire
- Istituto Superiore di Sanita', National Centre for Drug Research and Evaluation, 00161, Rome, Italy
| | - Valentina Chiodi
- Istituto Superiore di Sanita', National Centre for Drug Research and Evaluation, 00161, Rome, Italy
| | - Andrea Martinelli
- Istituto Superiore di Sanita', Experimental Animal Welfare Sector, 00161, Rome, Italy
| | - Anna Maria Rinaldi
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133, Rome, Italy
| | | | - Simonetta Pazzaglia
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00123, Rome, Italy
| | - Maria Rosaria Domenici
- Istituto Superiore di Sanita', National Centre for Drug Research and Evaluation, 00161, Rome, Italy
| | - Enrico Garaci
- IRCCS San Raffaele Roma, 00163, Rome, Italy
- MEBIC Consortium, 00166, Rome, Italy
| | - Daniela Merlo
- Istituto Superiore di Sanita', Department of Neuroscience, 00161, Rome, Italy.
| |
Collapse
|
4
|
Iacono D, Murphy EK, Stimpson CD, Perl DP, Day RM. Low-dose brain radiation: lowering hyperphosphorylated-tau without increasing DNA damage or oncogenic activation. Sci Rep 2023; 13:21142. [PMID: 38036591 PMCID: PMC10689500 DOI: 10.1038/s41598-023-48146-w] [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: 05/05/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023] Open
Abstract
Brain radiation has been medically used to alter the metabolism of cancerous cells and induce their elimination. Rarely, though, brain radiation has been used to interfere with the pathomechanisms of non-cancerous brain disorders, especially neurodegenerative disorders. Data from low-dose radiation (LDR) on swine brains demonstrated reduced levels of phosphorylated-tau (CP13) and amyloid precursor protein (APP) in radiated (RAD) versus sham (SH) animals. Phosphorylated-tau and APP are involved in Alzheimer's disease (AD) pathogenesis. We determined if the expression levels of hyperphosphorylated-tau, 3R-tau, 4R-tau, synaptic, intraneuronal damage, and DNA damage/oncogenic activation markers were altered in RAD versus SH swine brains. Quantitative analyses demonstrated reduced levels of AT8 and 3R-tau in hippocampus (H) and striatum (Str), increased levels of synaptophysin and PSD-95 in frontal cortex (FCtx), and reduced levels of NF-L in cerebellum (CRB) of RAD versus SH swine. DNA damage and oncogene activation markers levels did not differ between RAD and SH animals, except for histone-H3 (increased in FCtx and CRB, decreased in Str), and p53 (reduced in FCtx, Str, H and CRB). These findings confirm the region-based effects of sLDR on proteins normally expressed in larger mammalian brains and support the potential applicability of LDR to beneficially interfere against neurodegenerative mechanisms.
Collapse
Affiliation(s)
- Diego Iacono
- DoD/USU Brain Tissue Repository and Neuropathology Program, Uniformed Services University (USU), Bethesda, MD, USA.
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA.
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA.
- Neuroscience Program, Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF) Inc., Bethesda, MD, USA.
- Neurodegeneration Disorders Clinic, National Institute of Neurological Disorders and Stroke, NINDS, NIH, Bethesda, MD, USA.
| | - Erin K Murphy
- DoD/USU Brain Tissue Repository and Neuropathology Program, Uniformed Services University (USU), Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF) Inc., Bethesda, MD, USA
| | - Cheryl D Stimpson
- DoD/USU Brain Tissue Repository and Neuropathology Program, Uniformed Services University (USU), Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF) Inc., Bethesda, MD, USA
| | - Daniel P Perl
- DoD/USU Brain Tissue Repository and Neuropathology Program, Uniformed Services University (USU), Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA
| | - Regina M Day
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, MD, USA
| |
Collapse
|
5
|
Yu SP, Jiang MQ, Shim SS, Pourkhodadad S, Wei L. Extrasynaptic NMDA receptors in acute and chronic excitotoxicity: implications for preventive treatments of ischemic stroke and late-onset Alzheimer's disease. Mol Neurodegener 2023; 18:43. [PMID: 37400870 DOI: 10.1186/s13024-023-00636-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/01/2023] [Indexed: 07/05/2023] Open
Abstract
Stroke and late-onset Alzheimer's disease (AD) are risk factors for each other; the comorbidity of these brain disorders in aging individuals represents a significant challenge in basic research and clinical practice. The similarities and differences between stroke and AD in terms of pathogenesis and pathophysiology, however, have rarely been comparably reviewed. Here, we discuss the research background and recent progresses that are important and informative for the comorbidity of stroke and late-onset AD and related dementia (ADRD). Glutamatergic NMDA receptor (NMDAR) activity and NMDAR-mediated Ca2+ influx are essential for neuronal function and cell survival. An ischemic insult, however, can cause rapid increases in glutamate concentration and excessive activation of NMDARs, leading to swift Ca2+ overload in neuronal cells and acute excitotoxicity within hours and days. On the other hand, mild upregulation of NMDAR activity, commonly seen in AD animal models and patients, is not immediately cytotoxic. Sustained NMDAR hyperactivity and Ca2+ dysregulation lasting from months to years, nevertheless, can be pathogenic for slowly evolving events, i.e. degenerative excitotoxicity, in the development of AD/ADRD. Specifically, Ca2+ influx mediated by extrasynaptic NMDARs (eNMDARs) and a downstream pathway mediated by transient receptor potential cation channel subfamily M member (TRPM) are primarily responsible for excitotoxicity. On the other hand, the NMDAR subunit GluN3A plays a "gatekeeper" role in NMDAR activity and a neuroprotective role against both acute and chronic excitotoxicity. Thus, ischemic stroke and AD share an NMDAR- and Ca2+-mediated pathogenic mechanism that provides a common receptor target for preventive and possibly disease-modifying therapies. Memantine (MEM) preferentially blocks eNMDARs and was approved by the Federal Drug Administration (FDA) for symptomatic treatment of moderate-to-severe AD with variable efficacy. According to the pathogenic role of eNMDARs, it is conceivable that MEM and other eNMDAR antagonists should be administered much earlier, preferably during the presymptomatic phases of AD/ADRD. This anti-AD treatment could simultaneously serve as a preconditioning strategy against stroke that attacks ≥ 50% of AD patients. Future research on the regulation of NMDARs, enduring control of eNMDARs, Ca2+ homeostasis, and downstream events will provide a promising opportunity to understand and treat the comorbidity of AD/ADRD and stroke.
Collapse
Affiliation(s)
- Shan P Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA.
| | - Michael Q Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Seong S Shim
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Soheila Pourkhodadad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
6
|
Takahashi H, Yamamoto T, Tsuboi A. Molecular mechanisms underlying activity-dependent ischemic tolerance in the brain. Neurosci Res 2023; 186:3-9. [PMID: 36244569 DOI: 10.1016/j.neures.2022.10.005] [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: 08/08/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. The inhibition of cerebral blood flow triggers intertwined pathological events, resulting in cell death and loss of brain function. Interestingly, animals pre-exposed to short-term ischemia can tolerate subsequent severe ischemia. This phenomenon is called ischemic tolerance and is also triggered by other noxious stimuli. However, whether short-term exposure to non-noxious stimuli can induce ischemic tolerance remains unknown. Recently, we found that pre-exposing mice to an enriched environment for 40 min is sufficient to facilitate cell survival after a subsequent stroke. The neuroprotective process depends on the neuronal activity soon before stroke, of which the activity-dependent transcription factor Npas4 is essential. Excessive Ca2+ influx triggers Npas4 expression in ischemic neurons, leading to the activation of neuroprotective programs. Pre-induction of Npas4 in the normal brain effectively supports cell survival after stroke. Furthermore, our study revealed that Npas4 regulates L-type voltage-gated Ca2+ channels through expression of the small Ras-like GTPase Gem in ischemic neurons. Ischemic tolerance is a good model for understanding how to promote neuroprotective mechanisms in the normal and injured brain. Here, we highlight activity-dependent ischemic tolerance and discuss its role in promoting neuroprotection against stroke.
Collapse
Affiliation(s)
- Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan.
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Akio Tsuboi
- Dynamic Brain Network Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
7
|
Lissek T. Activity-Dependent Induction of Younger Biological Phenotypes. Adv Biol (Weinh) 2022; 6:e2200119. [PMID: 35976161 DOI: 10.1002/adbi.202200119] [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: 04/25/2022] [Revised: 07/11/2022] [Indexed: 01/28/2023]
Abstract
In several mammalian species, including humans, complex stimulation patterns such as cognitive and physical exercise lead to improvements in organ function, organism health and performance, as well as possibly longer lifespans. A framework is introduced here in which activity-dependent transcriptional programs, induced by these environmental stimuli, move somatic cells such as neurons and muscle cells toward a state that resembles younger cells to allow remodeling and adaptation of the organism. This cellular adaptation program targets several process classes that are heavily implicated in aging, such as mitochondrial metabolism, cell-cell communication, and epigenetic information processing, and leads to functional improvements in these areas. The activity-dependent gene program (ADGP) can be seen as a natural, endogenous cellular reprogramming mechanism that provides deep insight into the principles of inducible improvements in cell and organism function and can guide the development of therapeutic approaches for longevity. Here, these ADGPs are analyzed, exemplary critical molecular nexus points such as cAMP response element-binding protein, myocyte enhancer factor 2, serum response factor, and c-Fos are identified, and it is explored how one may leverage them to prevent, attenuate, and reverse human aging-related decline of body function.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| |
Collapse
|
8
|
Seillier C, Lesept F, Toutirais O, Potzeha F, Blanc M, Vivien D. Targeting NMDA Receptors at the Neurovascular Unit: Past and Future Treatments for Central Nervous System Diseases. Int J Mol Sci 2022; 23:ijms231810336. [PMID: 36142247 PMCID: PMC9499580 DOI: 10.3390/ijms231810336] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
The excitatory neurotransmission of the central nervous system (CNS) mainly involves glutamate and its receptors, especially N-methyl-D-Aspartate receptors (NMDARs). These receptors have been extensively described on neurons and, more recently, also on other cell types. Nowadays, the study of their differential expression and function is taking a growing place in preclinical and clinical research. The diversity of NMDAR subtypes and their signaling pathways give rise to pleiotropic functions such as brain development, neuronal plasticity, maturation along with excitotoxicity, blood-brain barrier integrity, and inflammation. NMDARs have thus emerged as key targets for the treatment of neurological disorders. By their large extracellular regions and complex intracellular structures, NMDARs are modulated by a variety of endogenous and pharmacological compounds. Here, we will present an overview of NMDAR functions on neurons and other important cell types involved in the pathophysiology of neurodegenerative, neurovascular, mental, autoimmune, and neurodevelopmental diseases. We will then discuss past and future development of NMDAR targeting drugs, including innovative and promising new approaches.
Collapse
Affiliation(s)
- Célia Seillier
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
| | - Flavie Lesept
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Olivier Toutirais
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU, 14000 Caen, France
| | - Fanny Potzeha
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Manuel Blanc
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, 14000 Caen, France
- Correspondence:
| |
Collapse
|
9
|
Fairless R, Bading H, Diem R. Pathophysiological Ionotropic Glutamate Signalling in Neuroinflammatory Disease as a Therapeutic Target. Front Neurosci 2021; 15:741280. [PMID: 34744612 PMCID: PMC8567076 DOI: 10.3389/fnins.2021.741280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Glutamate signalling is an essential aspect of neuronal communication involving many different glutamate receptors, and underlies the processes of memory, learning and synaptic plasticity. Despite neuroinflammatory diseases covering a range of maladies with very different biological causes and pathophysiologies, a central role for dysfunctional glutamate signalling is becoming apparent. This is not just restricted to the well-described role of glutamate in mediating neurodegeneration, but also includes a myriad of other influences that glutamate can exert on the vasculature, as well as immune cell and glial regulation, reflecting the ability of neurons to communicate with these compartments in order to couple their activity with neuronal requirements. Here, we discuss the role of pathophysiological glutamate signalling in neuroinflammatory disease, using both multiple sclerosis and Alzheimer's disease as examples, and how current steps are being made to harness our growing understanding of these processes in the development of neuroprotective strategies. This review focuses in particular on N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methylisooxazol-4-yl) propionate (AMPA) type ionotropic glutamate receptors, although metabotropic, G-protein-coupled glutamate receptors may also contribute to neuroinflammatory processes. Given the indispensable roles of glutamate-gated ion channels in synaptic communication, means of pharmacologically distinguishing between physiological and pathophysiological actions of glutamate will be discussed that allow deleterious signalling to be inhibited whilst minimising the disturbance of essential neuronal function.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
10
|
Huang M, Lou D, Charli A, Kong D, Jin H, Zenitsky G, Anantharam V, Kanthasamy A, Wang Z, Kanthasamy AG. Mitochondrial dysfunction-induced H3K27 hyperacetylation perturbs enhancers in Parkinson's disease. JCI Insight 2021; 6:138088. [PMID: 34494552 PMCID: PMC8492320 DOI: 10.1172/jci.insight.138088] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial dysfunction is a major pathophysiological contributor to the progression of Parkinson's disease (PD); however, whether it contributes to epigenetic dysregulation remains unknown. Here, we show that both chemically and genetically driven mitochondrial dysfunctions share a common mechanism of epigenetic dysregulation. Under both scenarios, lysine 27 acetylation of likely variant H3.3 (H3.3K27ac) increased in dopaminergic neuronal models of PD, thereby opening that region to active enhancer activity via H3K27ac. These vulnerable epigenomic loci represent potential transcription factor motifs for PD pathogenesis. We further confirmed that mitochondrial dysfunction induces H3K27ac in ex vivo and in vivo (MitoPark) neurodegenerative models of PD. Notably, the significantly increased H3K27ac in postmortem PD brains highlights the clinical relevance to the human PD population. Our results reveal an exciting mitochondrial dysfunction-metabolism-H3K27ac-transcriptome axis for PD pathogenesis. Collectively, the mechanistic insights link mitochondrial dysfunction to epigenetic dysregulation in dopaminergic degeneration and offer potential new epigenetic intervention strategies for PD.
Collapse
Affiliation(s)
- Minhong Huang
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Dan Lou
- Laboratory of Environmental Epigenomes, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adhithiya Charli
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Dehui Kong
- Laboratory of Environmental Epigenomes, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, Hubei Province, China
| | - Huajun Jin
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Gary Zenitsky
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Vellareddy Anantharam
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Arthi Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Zhibin Wang
- Laboratory of Environmental Epigenomes, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, Hubei Province, China
| | - Anumantha G. Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
11
|
Bayazid AB, Jang YA, Kim YM, Kim JG, Lim BO. Neuroprotective Effects of Sodium Butyrate through Suppressing Neuroinflammation and Modulating Antioxidant Enzymes. Neurochem Res 2021; 46:2348-2358. [PMID: 34106394 DOI: 10.1007/s11064-021-03369-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/25/2021] [Accepted: 05/31/2021] [Indexed: 11/28/2022]
Abstract
The discovery of effective therapeutic agents against neurodegenerative diseases (NDDs) remains challenging. Neurotoxicity, inflammations, and oxidative stress are associating factors of NDDs. Sodium butyrate (NaB) is a short-chain fatty acid found in diet and produced in the gut that reportedly protects cancer, inflammation, obesity and so on. Previously, SH-SY5Y cells were studied as in vitro models of cerebral diseases. We have investigated the neuroprotective effects of NaB in SH-SY5Y cells stimulated with TNF-α. The expression of inflammatory mediators, including iNOS, COX-2, and mitogen-activated protein kinases (MAPK) and the apoptotic regulators, including P-53, Bcl-2 associated X (BAX) Protein, and caspase-3 were analyzed by western blot analysis. The anti-apoptotic gene Bcl-2 and the pro-apoptotic gene BAX translocation were also investigated. Our results showed that NaB attenuated cell death and inhibited the NO production and decreased the expression of iNOS and COX-2 in TNF-α-stimulated SH-SY5Y cells. NaB notably ameliorated apoptotic regulatory proteins p-53, Caspase-3 and caspase-1 level, and reversed phosphorylation of extracellular signal-regulated kinases and p-38 proteins. NaB ameliorated Glucocorticoid receptor and NLRP3 inflammasome expressions. NaB also suppressed the BAX nuclear translocation and modulated Nrf-2, HO-1 and MnSOD expression in neuroblastoma cells. In addition, NaB substantially reversed the reactive oxygen species in H2O2 induced SH-SY5Y cells. Altogether, our results suggest that sodium butyrate has potential therapeutic effects against NDDs.
Collapse
Affiliation(s)
- Al Borhan Bayazid
- Department of Integrated Biosciences, Graduate School of Konkuk University, Chungju, 27478, Korea
| | - Young Ah Jang
- Convergence Research Center for Smart Healthcare, R&DB Foundation of Kyungsung University, Busan, Korea
| | - Yu Mi Kim
- Bio-Nano Technology Co, Daegu, Korea
| | - Jae Gon Kim
- BK21 FOUR, GLOCAL Education Program for Nutraceutical and Biopharmaceutical Research, Konkuk University, Chungju, 27478, Korea
| | - Beong Ou Lim
- Department of Integrated Biosciences, Graduate School of Konkuk University, Chungju, 27478, Korea.
| |
Collapse
|
12
|
Back MK, Ruggieri S, Jacobi E, von Engelhardt J. Amyloid Beta-Mediated Changes in Synaptic Function and Spine Number of Neocortical Neurons Depend on NMDA Receptors. Int J Mol Sci 2021; 22:ijms22126298. [PMID: 34208315 PMCID: PMC8231237 DOI: 10.3390/ijms22126298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Onset and progression of Alzheimer's disease (AD) pathophysiology differs between brain regions. The neocortex, for example, is a brain region that is affected very early during AD. NMDA receptors (NMDARs) are involved in mediating amyloid beta (Aβ) toxicity. NMDAR expression, on the other hand, can be affected by Aβ. We tested whether the high vulnerability of neocortical neurons for Aβ-toxicity may result from specific NMDAR expression profiles or from a particular regulation of NMDAR expression by Aβ. Electrophysiological analyses suggested that pyramidal cells of 6-months-old wildtype mice express mostly GluN1/GluN2A NMDARs. While synaptic NMDAR-mediated currents are unaltered in 5xFAD mice, extrasynaptic NMDARs seem to contain GluN1/GluN2A and GluN1/GluN2A/GluN2B. We used conditional GluN1 and GluN2B knockout mice to investigate whether NMDARs contribute to Aβ-toxicity. Spine number was decreased in pyramidal cells of 5xFAD mice and increased in neurons with 3-week virus-mediated Aβ-overexpression. NMDARs were required for both Aβ-mediated changes in spine number and functional synapses. Thus, our study gives novel insights into the Aβ-mediated regulation of NMDAR expression and the role of NMDARs in Aβ pathophysiology in the somatosensory cortex.
Collapse
|
13
|
Mott RE, von Reyn CR, Firestein BL, Meaney DF. Regional Neurodegeneration in vitro: The Protective Role of Neural Activity. Front Comput Neurosci 2021; 15:580107. [PMID: 33854425 PMCID: PMC8039287 DOI: 10.3389/fncom.2021.580107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 02/11/2021] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury is a devastating public health problem, the eighth leading cause of death across the world. To improve our understanding of how injury at the cellular scale affects neural circuit function, we developed a protocol to precisely injure individual neurons within an in vitro neural network. We used high speed calcium imaging to estimate alterations in neural activity and connectivity that occur followed targeted microtrauma. Our studies show that mechanically injured neurons inactivate following microtrauma and eventually re-integrate into the network. Single neuron re-integration is dependent on its activity prior to injury and initial connections in the network: more active and integrated neurons are more resistant to microtrauma and more likely to re-integrate into the network. Micromechanical injury leads to neuronal death 6 h post-injury in a subset of both injured and uninjured neurons. Interestingly, neural activity and network participation after injury were associated with survival in linear discriminate analysis (77.3% correct prediction, Wilks' Lambda = 0.838). Based on this observation, we modulated neuronal activity to rescue neurons after microtrauma. Inhibition of neuronal activity provided much greater survivability than did activation of neurons (ANOVA, p < 0.01 with post-hoc Tukey HSD, p < 0.01). Rescue of neurons by blocking activity in the post-acute period is partially mediated by mitochondrial energetics, as we observed silencing neurons after micromechanical injury led to a significant reduction in mitochondrial calcium accumulation. Overall, the present study provides deeper insight into the propagation of injury within networks, demonstrating that together the initial activity, network structure, and post-injury activity levels contribute to the progressive changes in a neural circuit after mechanical trauma.
Collapse
Affiliation(s)
| | - Catherine R von Reyn
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Ahmed H, Haider A, Ametamey SM. N-Methyl-D-Aspartate (NMDA) receptor modulators: a patent review (2015-present). Expert Opin Ther Pat 2020; 30:743-767. [PMID: 32926646 DOI: 10.1080/13543776.2020.1811234] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION - The NMDA receptor is implicated in various diseases including neurodegenerative, neurodevelopmental and mood disorders. However, only a limited number of clinically approved NMDA receptor modulators are available. Today, apparent NMDA receptor drug development strategies entail 1) exploring the unknown chemical space to identify novel scaffolds; 2) using the clinically available NMDA receptor modulators to expand the therapeutic indication space; 3) and to trace physiological functions of the NMDA receptor. AREAS COVERED - The current review reflects on the functional and pharmacological facets of NMDA receptors and the current clinical status quo of NMDA receptor modulators. Patent literature covering 2015 till April 2020 is discussed with emphasis on new indications. EXPERT OPINION - Supporting evidence shows that subtype-selective NMDA receptor antagonists show an improved safety profile compared to broad-spectrum channel blockers. Although GluN2B-selective antagonists are by far the most extensively investigated subtype-selective modulators, they have shown only modest clinical efficacy so far. To overcome the limitations that have hampered the clinical development of previous subtype-selective NMDA receptor antagonists, future studies with improved animal models that better reflect human NMDA receptor pathophysiology are warranted. The increased availability of subtype-selective probes will allow target engagement studies and proper dose finding in future clinical trials.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich , Zurich, Switzerland
| | - Ahmed Haider
- Department of Nuclear Medicine, University Hospital Zurich , Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich , Schlieren, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich , Zurich, Switzerland
| |
Collapse
|
15
|
Merz SF, Bengtson CP, Tepohl C, Hagenston AM, Bading H, Bas-Orth C. A microscopy-based small molecule screen in primary neurons reveals neuroprotective properties of the FDA-approved anti-viral drug Elvitegravir. Mol Brain 2020; 13:124. [PMID: 32928261 PMCID: PMC7489219 DOI: 10.1186/s13041-020-00641-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 01/28/2023] Open
Abstract
Glutamate toxicity is a pathomechanism that contributes to neuronal cell death in a wide range of acute and chronic neurodegenerative and neuroinflammatory diseases. Activation of the N-methyl-D-aspartate (NMDA)-type glutamate receptor and breakdown of the mitochondrial membrane potential are key events during glutamate toxicity. Due to its manifold functions in nervous system physiology, however, the NMDA receptor is not well suited as a drug target. To identify novel compounds that act downstream of toxic NMDA receptor signaling and can protect mitochondria from glutamate toxicity, we developed a cell viability screening assay in primary mouse cortical neurons. In a proof-of-principle screen we tested 146 natural products and 424 FDA-approved drugs for their ability to protect neurons against NMDA-induced cell death. We confirmed several known neuroprotective drugs that include Dutasteride, Enalapril, Finasteride, Haloperidol, and Oxybutynin, and we identified neuroprotective properties of Elvitegravir. Using live imaging of tetramethylrhodamine ethyl ester-labelled primary cortical neurons, we found that Elvitegravir, Dutasteride, and Oxybutynin attenuated the NMDA-induced breakdown of the mitochondrial membrane potential. Patch clamp electrophysiological recordings in NMDA receptor-expressing HEK293 cell lines and primary mouse hippocampal neurons revealed that Elvitegravir does not act at the NMDA receptor and does not affect the function of glutamatergic synapses. In summary, we have developed a cost-effective and easy-to-implement screening assay in primary neurons and identified Elvitegravir as a neuro- and mitoprotective drug that acts downstream of the NMDA receptor.
Collapse
Affiliation(s)
- Simon F Merz
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany
| | - Clara Tepohl
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany
| | - Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, D-69120, Heidelberg, Germany. .,Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, D-69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
17
|
Bas-Orth C, Koch M, Lau D, Buchthal B, Bading H. A microRNA signature of toxic extrasynaptic N-methyl-D-aspartate (NMDA) receptor signaling. Mol Brain 2020; 13:3. [PMID: 31924235 PMCID: PMC6954508 DOI: 10.1186/s13041-020-0546-0] [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: 10/15/2019] [Accepted: 01/05/2020] [Indexed: 12/27/2022] Open
Abstract
The cellular consequences of N-Methyl-D-Aspartate receptor (NMDAR) stimulation depend on the receptors' subcellular localization. Synaptic NMDARs promote plasticity and survival whereas extrasynaptic NMDARs mediate excitotoxicity and contribute to cell death in neurodegenerative diseases. The mechanisms that couple activation of extrasynaptic NMDARs to cell death remain incompletely understood. We here show that activation of extrasynaptic NMDARs by bath application of NMDA or L-glutamate leads to the upregulation of a group of 19 microRNAs in cultured mouse hippocampal neurons. In contrast, none of these microRNAs is induced upon stimulation of synaptic activity. Increased microRNA expression depends on the pri-miRNA processing enzyme Drosha, but not on de novo gene transcription. These findings suggest that toxic NMDAR signaling involves changes in the expression levels of particular microRNAs.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany. .,Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany.
| | - Mirja Koch
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - David Lau
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| |
Collapse
|
18
|
Courtney NL, Mole AJ, Thomson AK, Murray LM. Reduced P53 levels ameliorate neuromuscular junction loss without affecting motor neuron pathology in a mouse model of spinal muscular atrophy. Cell Death Dis 2019; 10:515. [PMID: 31273192 PMCID: PMC6609617 DOI: 10.1038/s41419-019-1727-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/23/2019] [Accepted: 05/28/2019] [Indexed: 11/09/2022]
Abstract
Spinal Muscular Atrophy (SMA) is a childhood motor neuron disease caused by mutations or deletions within the SMN1 gene. At endstages of disease there is profound loss of motor neurons, loss of axons within ventral roots and defects at the neuromuscular junctions (NMJ), as evidenced by pathological features such as pre-synaptic loss and swelling and post-synaptic shrinkage. Although these motor unit defects have been widely described, the time course and interdependancy of these aspects of motor unit degeneration are unclear. Recent reports have also revealed an early upregulation of transcripts associated with the P53 signalling pathway. The relationship between the upregulation of these transcripts and pathology within the motor unit is also unclear. In this study, we exploit the prolonged disease timecourse and defined pre-symptomatic period in the Smn2B/- mouse model to perform a temporal analysis of the different elements of motor unit pathology. We demonstrate that NMJ loss occurs prior to cell body loss, and coincides with the onset of symptoms. The onset of NMJ pathology also coincides with an increase in P53-related transcripts at the cell body. Finally, using a tamoxifen inducible P53 knockout, we demonstrate that post-natal reduction in P53 levels can reduce NMJ loss, but does not affect other aspects of NMJ pathology, motor neuron loss or the phenotype of the Smn2B/- mouse model. Together this work provides a detailed temporal description of pathology within motor units of an SMA mouse model, and demonstrates that NMJ loss is a P53-dependant process. This work supports the role for P53 as an effector of synaptic and axonal degeneration in a die-back neuropathy.
Collapse
Affiliation(s)
- Natalie L Courtney
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Scotland, EH8 9XD, UK
| | - Alannah J Mole
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Scotland, EH8 9XD, UK
| | - Alison K Thomson
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Scotland, EH8 9XD, UK
| | - Lyndsay M Murray
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, Edinburgh, UK. .,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Scotland, EH8 9XD, UK.
| |
Collapse
|
19
|
Cinel SD, Taylor SJ. Prolonged Bat Call Exposure Induces a Broad Transcriptional Response in the Male Fall Armyworm ( Spodoptera frugiperda; Lepidoptera: Noctuidae) Brain. Front Behav Neurosci 2019; 13:36. [PMID: 30863292 PMCID: PMC6399161 DOI: 10.3389/fnbeh.2019.00036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/11/2019] [Indexed: 12/22/2022] Open
Abstract
Predation risk induces broad behavioral and physiological responses that have traditionally been considered acute and transitory. However, prolonged or frequent exposure to predators and the sensory cues of their presence they broadcast to the environment impact long-term prey physiology and demographics. Though several studies have assessed acute and chronic stress responses in varied taxa, these attempts have often involved a priori expectations of the molecular pathways involved in physiological responses, such as glucocorticoid pathways and neurohormone production in vertebrates. While relatively little is known about physiological and molecular predator-induced stress in insects, many dramatic insect defensive behaviors have evolved to combat selection by predators. For instance, several moth families, such as Noctuidae, include members equipped with tympanic organs that allow the perception of ultrasonic bat calls and facilitate predation avoidance by eliciting evasive aerial flight maneuvers. In this study, we exposed adult male fall armyworm (Spodoptera frugiperda) moths to recorded ultrasonic bat foraging and attack calls for a prolonged period and constructed a de novo transcriptome based on brain tissue from predator cue-exposed relative to control moths kept in silence. Differential expression analysis revealed that 290 transcripts were highly up- or down-regulated among treatment tissues, with many annotating to noteworthy proteins, including a heat shock protein and an antioxidant enzyme involved in cellular stress. Though nearly 50% of differentially expressed transcripts were unannotated, those that were are implied in a broad range of cellular functions within the insect brain, including neurotransmitter metabolism, ionotropic receptor expression, mitochondrial metabolism, heat shock protein activity, antioxidant enzyme activity, actin cytoskeleton dynamics, chromatin binding, methylation, axonal guidance, cilia development, and several signaling pathways. The five most significantly overrepresented Gene Ontology terms included chromatin binding, macromolecular complex binding, glutamate synthase activity, glutamate metabolic process, and glutamate biosynthetic process. As a first assessment of transcriptional responses to ecologically relevant auditory predator cues in the brain of moth prey, this study lays the foundation for examining the influence of these differentially expressed transcripts on insect behavior, physiology, and life history within the framework of predation risk, as observed in ultrasound-sensitive Lepidoptera and other 'eared' insects.
Collapse
Affiliation(s)
- Scott D Cinel
- Illinois Natural History Survey, Prairie Research Institute, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Insect Evolution, Behavior, and Genomics Lab, Florida Museum of Natural History, University of Florida, Gainesville, FL, United States
| | - Steven J Taylor
- Illinois Natural History Survey, Prairie Research Institute, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Colorado College, Colorado Springs, CO, United States
| |
Collapse
|
20
|
Depp C, Bas-Orth C, Schroeder L, Hellwig A, Bading H. Synaptic Activity Protects Neurons Against Calcium-Mediated Oxidation and Contraction of Mitochondria During Excitotoxicity. Antioxid Redox Signal 2018; 29:1109-1124. [PMID: 28990420 DOI: 10.1089/ars.2017.7092] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Excitotoxicity triggered by extrasynaptic N-methyl-d-aspartate-type glutamate receptors has been implicated in many neurodegenerative conditions, including Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, and stroke. Mitochondrial calcium overload leading to mitochondrial dysfunction represents an early event in excitotoxicity. Neurons are rendered resistant to excitotoxicity by previous periods of synaptic activity that activates a nuclear calcium-driven neuroprotective gene program. This process, termed acquired neuroprotection, involves transcriptional repression of the mitochondrial calcium uniporter leading to a reduction in excitotoxcity-associated mitochondrial calcium load. As mitochondrial calcium and the production of reactive oxygen species may be linked, we monitored excitotoxicity-associated changes in the mitochondrial redox status using the ratiometric glutathione redox potential indicator, glutaredoxin 1 (GRX1)-redox-sensitive green fluorescent protein (roGFP)2, targeted to the mitochondrial matrix. Aim of this study was to investigate if suppression of oxidative stress underlies mitoprotection afforded by synaptic activity. RESULTS We found that synaptic activity protects primary rat hippocampal neurons against acute excitotoxicity-induced mitochondrial oxidative stress and mitochondrial contraction associated with it. Downregulation of the mitochondrial uniporter by genetic means mimics the protective effect of synaptic activity on mitochondrial redox status. These findings indicate that oxidative stress acts downstream of mitochondrial calcium overload in excitotoxicity. Innovation and Conclusion: We established mito-GRX1-roGFP2 as a reliable and sensitive tool to monitor rapid redox changes in mitochondria during excitotoxicity. Our results highlight the importance of developing means of blocking mitochondrial calcium overload for therapeutic targeting of oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Antioxid. Redox. Signal. 29, 1109-1124.
Collapse
Affiliation(s)
- Constanze Depp
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Lisa Schroeder
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Andrea Hellwig
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| |
Collapse
|
21
|
p53-inducible DPYSL4 associates with mitochondrial supercomplexes and regulates energy metabolism in adipocytes and cancer cells. Proc Natl Acad Sci U S A 2018; 115:8370-8375. [PMID: 30061407 PMCID: PMC6099896 DOI: 10.1073/pnas.1804243115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The tumor suppressor p53 regulates multiple cellular functions, including energy metabolism. Metabolic deregulation is implicated in the pathogenesis of some cancers and in metabolic disorders and may result from the inactivation of p53 functions. Using RNA sequencing and ChIP sequencing of cancer cells and preadipocytes, we demonstrate that p53 modulates several metabolic processes via the transactivation of energy metabolism genes including dihydropyrimidinase-like 4 (DPYSL4). DPYSL4 is a member of the collapsin response mediator protein family, which is involved in cancer invasion and progression. Intriguingly, DPYSL4 overexpression in cancer cells and preadipocytes up-regulated ATP production and oxygen consumption, while DPYSL4 knockdown using siRNA or CRISPR/Cas9 down-regulated energy production. Furthermore, DPYSL4 was associated with mitochondrial supercomplexes, and deletion of its dihydropyrimidinase-like domain abolished its association and its ability to stimulate ATP production and suppress the cancer cell invasion. Mouse-xenograft and lung-metastasis models indicated that DPYSL4 expression compromised tumor growth and metastasis in vivo. Consistently, database analyses demonstrated that low DPYSL4 expression was significantly associated with poor survival of breast and ovarian cancers in accordance with its reduced expression in certain types of cancer tissues. Moreover, immunohistochemical analysis using the adipose tissue of obese patients revealed that DPYSL4 expression was positively correlated with INFg and body mass index in accordance with p53 activation. Together, these results suggest that DPYSL4 plays a key role in the tumor-suppressor function of p53 by regulating oxidative phosphorylation and the cellular energy supply via its association with mitochondrial supercomplexes, possibly linking to the pathophysiology of both cancer and obesity.
Collapse
|
22
|
HIPK2-Mediated Transcriptional Control of NMDA Receptor Subunit Expression Regulates Neuronal Survival and Cell Death. J Neurosci 2018; 38:4006-4019. [PMID: 29581378 DOI: 10.1523/jneurosci.3577-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/01/2018] [Accepted: 02/20/2018] [Indexed: 11/21/2022] Open
Abstract
NMDA receptors are critical for neuronal communication. Dysfunction in NMDA receptors has been implicated in neuropsychiatric diseases. While it is well recognized that the composition of NMDA receptors undergoes a GluN2B-to-GluN2A switch in early postnatal life, the mechanism regulating this switch remains unclear. Using transcriptomic and functional analyses in brain tissues from male and female Hipk2+/+ and Hipk2-/- mice, we showed that the HIPK2-JNK-c-Jun pathway is important in suppressing the transcription of Grin2a and Grin2c, which encodes the GluN2A and GluN2C subunits of the NMDA receptors, respectively. Loss of HIPK2 leads to a significant decrease in JNK-c-Jun signaling, which in turn derepresses the transcription of Grin2a and Grin2c mRNA and upregulates GluN2A and GluN2C protein levels. These changes result in a significant increase of GluN2A/GluN2B ratio in synapse and mitochondria, a persistent activation of the ERK-CREB pathway and the upregulation of synaptic activity-regulated genes, which collectively contribute to the resistance of Hipk2-/- neurons to cell death induced by mitochondrial toxins.SIGNIFICANCE STATEMENT We identify HIPK2-JNK-c-Jun signaling as a key mechanism that regulates the transcription of NMDA receptor subunits GluN2A and GluN2C in vivo Our results provide insights into a previously unrecognized molecular mechanism that control the switch of NMDA receptor subunits in early postnatal brain development. Furthermore, we provide evidence that changes in the ratio of NMDA subunits GluN2A/GluN2B can also be detected in the synapse and mitochondria, which contributes to a persistent activation of the prosurvival ERK-CREB pathway and its downstream target genes. Collectively, these changes protect HIPK2 deficient neurons from mitochondrial toxins.
Collapse
|
23
|
The Intellectual Disability and Schizophrenia Associated Transcription Factor TCF4 Is Regulated by Neuronal Activity and Protein Kinase A. J Neurosci 2017; 37:10516-10527. [PMID: 28951451 DOI: 10.1523/jneurosci.1151-17.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/10/2017] [Accepted: 09/16/2017] [Indexed: 12/23/2022] Open
Abstract
Transcription factor 4 (TCF4 also known as ITF2 or E2-2) is a basic helix-loop-helix (bHLH) protein associated with Pitt-Hopkins syndrome, intellectual disability, and schizophrenia (SCZ). Here, we show that TCF4-dependent transcription in cortical neurons cultured from embryonic rats of both sexes is induced by neuronal activity via soluble adenylyl cyclase and protein kinase A (PKA) signaling. PKA phosphorylates TCF4 directly and a PKA phosphorylation site in TCF4 is necessary for its transcriptional activity in cultured neurons and in the developing brain in vivo We also demonstrate that Gadd45g (growth arrest and DNA damage inducible gamma) is a direct target of neuronal-activity-induced, TCF4-dependent transcriptional regulation and that TCF4 missense variations identified in SCZ patients alter the transcriptional activity of TCF4 in neurons. This study identifies a new role for TCF4 as a neuronal-activity-regulated transcription factor, offering a novel perspective on the association of TCF4 with cognitive disorders.SIGNIFICANCE STATEMENT The importance of the basic helix-loop-helix transcription factor transcription factor 4 (TCF4) in the nervous system is underlined by its association with common and rare cognitive disorders. In the current study, we show that TCF4-controlled transcription in primary cortical neurons is induced by neuronal activity and protein kinase A. Our results support the hypotheses that dysregulation of neuronal-activity-dependent signaling plays a significant part in the etiology of neuropsychiatric and neurodevelopmental disorders.
Collapse
|
24
|
Olmo IG, Carvalho TG, Costa VV, Alves-Silva J, Ferrari CZ, Izidoro-Toledo TC, da Silva JF, Teixeira AL, Souza DG, Marques JT, Teixeira MM, Vieira LB, Ribeiro FM. Zika Virus Promotes Neuronal Cell Death in a Non-Cell Autonomous Manner by Triggering the Release of Neurotoxic Factors. Front Immunol 2017; 8:1016. [PMID: 28878777 PMCID: PMC5572413 DOI: 10.3389/fimmu.2017.01016] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/08/2017] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV) has recently caused a worldwide outbreak of infections associated with severe neurological complications, including microcephaly in infants born from infected mothers. ZIKV exhibits high neurotropism and promotes neuroinflammation and neuronal cell death. We have recently demonstrated that N-methyl-d-aspartate receptor (NMDAR) blockade by memantine prevents ZIKV-induced neuronal cell death. Here, we show that ZIKV induces apoptosis in a non-cell autonomous manner, triggering cell death of uninfected neurons by releasing cytotoxic factors. Neuronal cultures infected with ZIKV exhibit increased levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and glutamate. Moreover, infected neurons exhibit increased expression of GluN2B and augmented intracellular Ca2+ concentration. Blockade of GluN2B-containing NMDAR by ifenprodil normalizes Ca2+ levels and rescues neuronal cell death. Notably, TNF-α and IL-1β blockade decreases ZIKV-induced Ca2+ flux through GluN2B-containing NMDARs and reduces neuronal cell death, indicating that these cytokines might contribute to NMDAR sensitization and neurotoxicity. In addition, ZIKV-infected cultures treated with ifenprodil exhibits increased activation of the neuroprotective pathway including extracellular signal-regulated kinase and cAMP response element-binding protein, which may underlie ifenprodil-mediated neuroprotection. Together, our data shed some light on the neurotoxic mechanisms triggered by ZIKV and begin to elucidate how GluN2B-containing NMDAR blockade can prevent neurotoxicity.
Collapse
Affiliation(s)
- Isabella G Olmo
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Toniana G Carvalho
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vivian V Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana Alves-Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Tatiane C Izidoro-Toledo
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Antonio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Science, UT Health, Houston, TX, United States
| | | | - Joao T Marques
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
25
|
Dar NJ, Satti NK, Dutt P, Hamid A, Ahmad M. Attenuation of Glutamate-Induced Excitotoxicity by Withanolide-A in Neuron-Like Cells: Role for PI3K/Akt/MAPK Signaling Pathway. Mol Neurobiol 2017; 55:2725-2739. [DOI: 10.1007/s12035-017-0515-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/04/2017] [Indexed: 12/30/2022]
|
26
|
Tyebji S, Hannan AJ. Synaptopathic mechanisms of neurodegeneration and dementia: Insights from Huntington's disease. Prog Neurobiol 2017; 153:18-45. [PMID: 28377290 DOI: 10.1016/j.pneurobio.2017.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 03/19/2017] [Accepted: 03/30/2017] [Indexed: 12/20/2022]
Abstract
Dementia encapsulates a set of symptoms that include loss of mental abilities such as memory, problem solving or language, and reduces a person's ability to perform daily activities. Alzheimer's disease is the most common form of dementia, however dementia can also occur in other neurological disorders such as Huntington's disease (HD). Many studies have demonstrated that loss of neuronal cell function manifests pre-symptomatically and thus is a relevant therapeutic target to alleviate symptoms. Synaptopathy, the physiological dysfunction of synapses, is now being approached as the target for many neurological and psychiatric disorders, including HD. HD is an autosomal dominant and progressive degenerative disorder, with clinical manifestations that encompass movement, cognition, mood and behaviour. HD is one of the most common tandem repeat disorders and is caused by a trinucleotide (CAG) repeat expansion, encoding an extended polyglutamine tract in the huntingtin protein. Animal models as well as human studies have provided detailed, although not exhaustive, evidence of synaptic dysfunction in HD. In this review, we discuss the neuropathology of HD and how the changes in synaptic signalling in the diseased brain lead to its symptoms, which include dementia. Here, we review and discuss the mechanisms by which the 'molecular orchestras' and their 'synaptic symphonies' are disrupted in neurodegeneration and dementia, focusing on HD as a model disease. We also explore the therapeutic strategies currently in pre-clinical and clinical testing that are targeted towards improving synaptic function in HD.
Collapse
Affiliation(s)
- Shiraz Tyebji
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
27
|
Wei H, Gao Z, Zheng L, Zhang C, Liu Z, Yang Y, Teng H, Hou L, Yin Y, Zou X. Protective Effects of Fucoidan on Aβ25-35 and d-Gal-Induced Neurotoxicity in PC12 Cells and d-Gal-Induced Cognitive Dysfunction in Mice. Mar Drugs 2017; 15:md15030077. [PMID: 28300775 PMCID: PMC5367034 DOI: 10.3390/md15030077] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/04/2017] [Accepted: 03/10/2017] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease which contributes to memory loss and cognitive decline in the elderly. Fucoidan, extracted from brown algae, is a complex sulfated polysaccharide and potential bioactive compound. In this study, we investigated whether fucoidan protects PC12 cells from apoptosis induced by a combination of beta-amyloid 25-35 (Aβ25-35) and d-galactose (d-Gal), and improves learning and memory impairment in AD model mice. The results indicated that fucoidan could inhibit the release of cytochrome c from the mitochondria to cytosol and activation of caspases, and increase the expression of apoptosis inhibitor proteins (IAPs), including livin and X-linked IAP (XIAP) in PC12 cells damaged by Aβ25-35 and d-Gal-induction. Fucoidan reversed the decreased activity of acetylcholine (ACh) and choline acetyl transferase (ChAT), as well as the increased activity of acetylcholine esterase (AChE), in AD model mice induced by infusion of d-Gal. Furthermore, fucoidan improved antioxidant activity in vitro and in vivo by activation of superoxide dismutase (SOD) and glutathione (GSH). These results suggested that fucoidan could protect PC12 cells from apoptosis and ameliorate the learning and memory impairment in AD model mice, which appeared to be due to regulating the cholinergic system, reducing oxidative stress, and inhibiting the caspase-dependent apoptosis pathway.
Collapse
Affiliation(s)
- Hengyun Wei
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Zixiang Gao
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Luping Zheng
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| | - Cuili Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Zundong Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Yazong Yang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Hongming Teng
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Lin Hou
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China.
| | - Yuling Yin
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Xiangyang Zou
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
28
|
Bas-Orth C, Tan YW, Lau D, Bading H. Synaptic Activity Drives a Genomic Program That Promotes a Neuronal Warburg Effect. J Biol Chem 2017; 292:5183-5194. [PMID: 28196867 DOI: 10.1074/jbc.m116.761106] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/01/2017] [Indexed: 12/17/2022] Open
Abstract
Synaptic activity drives changes in gene expression to promote long lasting adaptations of neuronal structure and function. One example of such an adaptive response is the buildup of acquired neuroprotection, a synaptic activity- and gene transcription-mediated increase in the resistance of neurons against harmful conditions. A hallmark of acquired neuroprotection is the stabilization of mitochondrial structure and function. We therefore re-examined previously identified sets of synaptic activity-regulated genes to identify genes that are directly linked to mitochondrial function. In mouse and rat primary hippocampal cultures, synaptic activity caused an up-regulation of glycolytic genes and a concomitant down-regulation of genes required for oxidative phosphorylation, mitochondrial biogenesis, and maintenance. Changes in metabolic gene expression were induced by action potential bursting, but not by glutamate bath application activating extrasynaptic NMDA receptors. The specific and coordinate pattern of gene expression changes suggested that synaptic activity promotes a shift of neuronal energy metabolism from oxidative phosphorylation toward aerobic glycolysis, also known as the Warburg effect. The ability of neurons to up-regulate glycolysis has, however, been debated. We therefore used FACS sorting to show that, in mixed neuron glia co-cultures, activity-dependent regulation of metabolic gene expression occurred in neurons. Changes in gene expression were accompanied by changes in the phosphorylation-dependent regulation of the key metabolic enzyme, pyruvate dehydrogenase. Finally, increased synaptic activity caused an increase in the ratio of l-lactate production to oxygen consumption in primary hippocampal cultures. Based on these data we suggest the existence of a synaptic activity-mediated neuronal Warburg effect that may promote mitochondrial homeostasis and neuroprotection.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Yan-Wei Tan
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - David Lau
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
29
|
Wright DJ, Renoir T, Gray LJ, Hannan AJ. Huntington’s Disease: Pathogenic Mechanisms and Therapeutic Targets. ADVANCES IN NEUROBIOLOGY 2017; 15:93-128. [DOI: 10.1007/978-3-319-57193-5_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Bas-Orth C, Tan YW, Oliveira AMM, Bengtson CP, Bading H. The calmodulin-binding transcription activator CAMTA1 is required for long-term memory formation in mice. ACTA ACUST UNITED AC 2016; 23:313-21. [PMID: 27194798 PMCID: PMC4880143 DOI: 10.1101/lm.041111.115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/04/2016] [Indexed: 01/26/2023]
Abstract
The formation of long-term memory requires signaling from the synapse to the nucleus to mediate neuronal activity-dependent gene transcription. Synapse-to-nucleus communication is initiated by influx of calcium ions through synaptic NMDA receptors and/or L-type voltage-gated calcium channels and involves the activation of transcription factors by calcium/calmodulin signaling in the nucleus. Recent studies have drawn attention to a new family of transcriptional regulators, the so-called calmodulin-binding transcription activator (CAMTA) proteins. CAMTAs are expressed at particularly high levels in the mouse and human brain, and we reasoned that, as calmodulin-binding transcription factors, CAMTAs may regulate the formation of long-term memory by coupling synaptic activity and calcium/calmodulin signaling to memory-related transcriptional responses. This hypothesis is supported by genetic studies that reported a correlation between Camta gene polymorphisms or mutations and cognitive capability in humans. Here, we show that acute knockdown of CAMTA1, but not CAMTA2, in the hippocampus of adult mice results in impaired performance in two memory tests, contextual fear conditioning and object-place recognition test. Short-term memory and neuronal morphology were not affected by CAMTA knockdown. Gene expression profiling in the hippocampus of control and CAMTA knockdown mice revealed a number of putative CAMTA1 target genes related to synaptic transmission and neuronal excitability. Patch clamp recordings in organotypic hippocampal slice cultures provided further evidence for CAMTA1-dependent changes in electrophysiological properties. In summary, our study provides experimental evidence that confirms previous human genetic studies and establishes CAMTA1 as a regulator of long-term memory formation.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - Yan-Wei Tan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
31
|
Khacho P, Wang B, Bergeron R. The Good and Bad Sides of NAAG. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:311-49. [PMID: 27288081 DOI: 10.1016/bs.apha.2016.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Why has such a small peptide been the source of controversy in neuroscience over the last 5 decades? Is N-acetyl-aspartyl-glutamate (NAAG) a neurotransmitter? Is NAAG located in neuronal tissue or in astrocytes? Is NAAG involved in neuropsychiatric and neurodegenerative disorders? Is NAAG therapeutically beneficial in the treatment of stroke or in initiating cascades of events leading to psychosis? After many years of intense research there is no clear consensus within the scientific community on how NAAG behaves in the brain. One of the major controversies about NAAG is its physiological action at N-methyl-d-aspartate (NMDA) receptors. While some researchers strongly argue that NAAG acts as a weak agonist at NMDA receptors, others have suggested that NAAG could behave as a potent antagonist. Published data from our laboratory demonstrate that the effect of NAAG on NMDA receptors could be influenced by a number of factors including the subcellular localization and subunit composition of NMDA receptors, as well as protons. In this chapter, we will summarize the knowledge of the literature on NAAG, however, we will place emphasis on our recently published data. More specifically, we have reported interesting findings on the effects of NAAG on NMDA receptors at synaptic and extrasynaptic sites using a pharmacological paradigm to distinguish the two populations of NMDA receptors. Additionally, we have evaluated the role of NAAG on GluN2A- and GluN2B-containing NMDA receptors using a HEK293 cell recombinant system. Finally, we have studied the effects of NAAG on GluN2A- and GluN2B-containing NMDA receptors in different extracellular pH conditions. We believe that our findings could potentially resolve some aspects of the debate regarding the role of NAAG at NMDA receptors.
Collapse
Affiliation(s)
- P Khacho
- University of Ottawa, Ottawa, ON, Canada
| | - B Wang
- University of Ottawa, Ottawa, ON, Canada
| | - R Bergeron
- University of Ottawa, Ottawa, ON, Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
32
|
Hoque A, Hossain MI, Ameen SS, Ang CS, Williamson N, Ng DCH, Chueh AC, Roulston C, Cheng HC. A beacon of hope in stroke therapy-Blockade of pathologically activated cellular events in excitotoxic neuronal death as potential neuroprotective strategies. Pharmacol Ther 2016; 160:159-79. [PMID: 26899498 DOI: 10.1016/j.pharmthera.2016.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Excitotoxicity, a pathological process caused by over-stimulation of ionotropic glutamate receptors, is a major cause of neuronal loss in acute and chronic neurological conditions such as ischaemic stroke, Alzheimer's and Huntington's diseases. Effective neuroprotective drugs to reduce excitotoxic neuronal loss in patients suffering from these neurological conditions are urgently needed. One avenue to achieve this goal is to clearly define the intracellular events mediating the neurotoxic signals originating from the over-stimulated glutamate receptors in neurons. In this review, we first focus on the key cellular events directing neuronal death but not involved in normal physiological processes in the neurotoxic signalling pathways. These events, referred to as pathologically activated events, are potential targets for the development of neuroprotectant therapeutics. Inhibitors blocking some of the known pathologically activated cellular events have been proven to be effective in reducing stroke-induced brain damage in animal models. Notable examples are inhibitors suppressing the ion channel activity of neurotoxic glutamate receptors and those disrupting interactions of specific cellular proteins occurring only in neurons undergoing excitotoxic cell death. Among them, Tat-NR2B9c and memantine are clinically effective in reducing brain damage caused by some acute and chronic neurological conditions. Our second focus is evaluation of the suitability of the other inhibitors for use as neuroprotective therapeutics. We also discuss the experimental approaches suitable for bridging our knowledge gap in our current understanding of the excitotoxic signalling mechanism in neurons and discovery of new pathologically activated cellular events as potential targets for neuroprotection.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ching-Seng Ang
- Bio21 Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Dominic C H Ng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia; School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia
| | - Anderly C Chueh
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Carli Roulston
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
33
|
Lau D, Bengtson CP, Buchthal B, Bading H. BDNF Reduces Toxic Extrasynaptic NMDA Receptor Signaling via Synaptic NMDA Receptors and Nuclear-Calcium-Induced Transcription of inhba/Activin A. Cell Rep 2015; 12:1353-66. [PMID: 26279570 DOI: 10.1016/j.celrep.2015.07.038] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/20/2015] [Accepted: 07/18/2015] [Indexed: 10/23/2022] Open
Abstract
The health of neurons is critically dependent on the relative signaling intensities of survival-promoting synaptic and death-inducing extrasynaptic NMDA receptors. Here, we show that BDNF is a regulator of this balance and promotes neuroprotection by reducing toxic NMDA receptor signaling. BDNF acts by initiating synaptic NMDA-receptor/nuclear-calcium-driven adaptogenomics, leading to increased expression of inhibin β-A (inhba). Inhibin β-A (its homodimer is known as activin A) in turn reduces neurotoxic extrasynaptic NMDA-receptor-mediated calcium influx, thereby shielding neurons against mitochondrial dysfunction, a major cause of excitotoxicity. Thus, BDNF induces acquired neuroprotection by enhancing synaptic activity and lowering extrasynaptic NMDA receptor death signaling through a nuclear calcium-inhibin β-A pathway. This process, which confers protection against ischemic brain damage in a mouse stroke model, may be compromised in Huntington's disease, Alzheimer's disease, or aging-related neurodegenerative conditions that are associated with reduced BDNF levels and/or enhanced extrasynaptic NMDA receptor signaling.
Collapse
Affiliation(s)
- David Lau
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| |
Collapse
|
34
|
Wang Y, Schachner M. The intracellular domain of L1CAM binds to casein kinase 2α and is neuroprotective via inhibition of the tumor suppressors PTEN and p53. J Neurochem 2015; 133:828-43. [PMID: 25727698 DOI: 10.1111/jnc.13083] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/05/2015] [Accepted: 02/24/2015] [Indexed: 02/05/2023]
Abstract
Cell adhesion molecule L1 promotes neuritogenesis and neuronal survival through triggering MAPK pathways. Based on the findings that L1 is associated with casein kinase 2 (CK2), and that deficiency in PTEN promotes neuritogenesis in vitro and regeneration after trauma, we examined the functional relationship between L1 and PTEN. In parallel, we investigated the tumor suppressor p53, which also regulates neuritogenesis. Here, we report that the intracellular domain of L1 binds to the subunit CK2α, and that knockdown of L1 leads to CK2 dephosphorylation and an increase in PTEN and p53 levels. Overexpression of L1, but not the L1 mutants L1 (S1181N, E1184V), which reduced binding between L1 and CK2, reduced expression levels of PTEN and p53 proteins, and enhanced levels of phosphorylated CK2α and mammalian target of rapamycin, which is a downstream effector of PTEN and p53. Treatment of neurons with a CK2 inhibitor or transfection with CK2α siRNA increased levels of PTEN and p53, and inhibited neuritogenesis. The combined observations indicate that L1 downregulates expression of PTEN and p53 via direct binding to CK2α. We suggest that L1 stimulates neuritogenesis by activating CK2α leading to decreased levels of PTEN and p53 via a novel, L1-triggered and CK2α-mediated signal transduction pathway. L1CAM (L1 cell adhesion molecule) is implicated in neural functions through the cognate src/MAP kinase signaling pathway. We now describe a novel signaling platform operating via the alpha subunit of casein kinase 2 which binds to the intracellular domain of L1. Knockdown of L1CAM leads to increased levels of tumor suppressor PTEN (phosphatase and tensin homolog) and p53, known to inhibit neuritogenesis in vitro and recovery from trauma in vivo. By activating this enzyme, L1CAM adds to its beneficial functions by decreasing the levels of PTEN and p53.
Collapse
Affiliation(s)
- Yan Wang
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
35
|
Regulation of neuronal gene expression and survival by basal NMDA receptor activity: a role for histone deacetylase 4. J Neurosci 2015; 34:15327-39. [PMID: 25392500 DOI: 10.1523/jneurosci.0569-14.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuronal gene expression is modulated by activity via calcium-permeable receptors such as NMDA receptors (NMDARs). While gene expression changes downstream of evoked NMDAR activity have been well studied, much less is known about gene expression changes that occur under conditions of basal neuronal activity. In mouse dissociated hippocampal neuronal cultures, we found that a broad NMDAR antagonist, AP5, induced robust gene expression changes under basal activity, but subtype-specific antagonists did not. While some of the gene expression changes are also known to be downstream of stimulated NMDAR activity, others appear specific to basal NMDAR activity. The genes altered by AP5 treatment of basal cultures were enriched for pathways related to class IIa histone deacetylases (HDACs), apoptosis, and synapse-related signaling. Specifically, AP5 altered the expression of all three class IIa HDACs that are highly expressed in the brain, HDAC4, HDAC5, and HDAC9, and also induced nuclear accumulation of HDAC4. HDAC4 knockdown abolished a subset of the gene expression changes induced by AP5, and led to neuronal death under long-term tetrodotoxin or AP5 treatment in rat hippocampal organotypic slice cultures. These data suggest that basal, but not evoked, NMDAR activity regulates gene expression in part through HDAC4, and, that HDAC4 has neuroprotective functions under conditions of low NMDAR activity.
Collapse
|
36
|
Cardinale A, de Stefano MC, Mollinari C, Racaniello M, Garaci E, Merlo D. Biochemical characterization of sirtuin 6 in the brain and its involvement in oxidative stress response. Neurochem Res 2014; 40:59-69. [PMID: 25366464 DOI: 10.1007/s11064-014-1465-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/02/2014] [Accepted: 10/23/2014] [Indexed: 12/21/2022]
Abstract
Sirtuin 6 (SIRT6) is a member of nicotinamide adenine dinucleotide-dependent deacetylase protein family and has been implicated in the control of glucose and lipid metabolism, cancer, genomic stability and DNA repair. Moreover, SIRT6 regulates the expression of a large number of genes involved in stress response and aging. The role of SIRT6 in brain function and neuronal survival is largely unknown. Here, we biochemically characterized SIRT6 in brain tissues and primary neuronal cultures and found that it is highly expressed in cortical and hippocampal regions and enriched in the synaptosomal membrane fraction. Immunoblotting analysis on cortical and hippocampal neurons showed that SIRT6 is downregulated during maturation in vitro, reaching the lowest expression at 11 days in vitro. In addition, SIRT6 overexpression in terminally differentiated cortical and hippocampal neurons, mediated by a neuron-specific recombinant adeno-associated virus, downregulated cell viability under oxidative stress condition. By contrast, under control condition, SIRT6 overexpression had no detrimental effect. Overall these results suggest that SIRT6 may play a role in synaptic function and neuronal maturation and it may be implicated in the regulation of neuronal survival.
Collapse
|
37
|
Ahlgren H, Bas-Orth C, Freitag HE, Hellwig A, Ottersen OP, Bading H. The nuclear calcium signaling target, activating transcription factor 3 (ATF3), protects against dendrotoxicity and facilitates the recovery of synaptic transmission after an excitotoxic insult. J Biol Chem 2014; 289:9970-82. [PMID: 24515113 DOI: 10.1074/jbc.m113.502914] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The focal swellings of dendrites ("dendritic beading") are an early morphological hallmark of neuronal injury and dendrotoxicity. They are associated with a variety of pathological conditions, including brain ischemia, and cause an acute disruption of synaptic transmission and neuronal network function, which contribute to subsequent neuronal death. Here, we show that increased synaptic activity prior to excitotoxic injury protects, in a transcription-dependent manner, against dendritic beading. Expression of activating transcription factor 3 (ATF3), a nuclear calcium-regulated gene and member of the core gene program for acquired neuroprotection, can protect against dendritic beading. Conversely, knockdown of ATF3 exacerbates dendritic beading. Assessment of neuronal network functions using microelectrode array recordings revealed that hippocampal neurons expressing ATF3 were able to regain their ability for functional synaptic transmission and to participate in coherent neuronal network activity within 48 h after exposure to toxic concentrations of NMDA. Thus, in addition to attenuating cell death, synaptic activity and expression of ATF3 render hippocampal neurons more resistant to acute dendrotoxicity and loss of synapses. Dendroprotection can enhance recovery of neuronal network functions after excitotoxic insults.
Collapse
Affiliation(s)
- Hanna Ahlgren
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, INF 364, 69120 Heidelberg, Germany and
| | | | | | | | | | | |
Collapse
|
38
|
Mitochondrial calcium uniporter Mcu controls excitotoxicity and is transcriptionally repressed by neuroprotective nuclear calcium signals. Nat Commun 2013; 4:2034. [PMID: 23774321 PMCID: PMC3709514 DOI: 10.1038/ncomms3034] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 05/20/2013] [Indexed: 01/28/2023] Open
Abstract
The recent identification of the mitochondrial Ca(2+) uniporter gene (Mcu/Ccdc109a) has enabled us to address its role, and that of mitochondrial Ca(2+) uptake, in neuronal excitotoxicity. Here we show that exogenously expressed Mcu is mitochondrially localized and increases mitochondrial Ca(2+) levels following NMDA receptor activation, leading to increased mitochondrial membrane depolarization and excitotoxic cell death. Knockdown of endogenous Mcu expression reduces NMDA-induced increases in mitochondrial Ca(2+), resulting in lower levels of mitochondrial depolarization and resistance to excitotoxicity. Mcu is subject to dynamic regulation as part of an activity-dependent adaptive mechanism that limits mitochondrial Ca(2+) overload when cytoplasmic Ca(2+) levels are high. Specifically, synaptic activity transcriptionally represses Mcu, via a mechanism involving the nuclear Ca(2+) and CaM kinase-mediated induction of Npas4, resulting in the inhibition of NMDA receptor-induced mitochondrial Ca(2+) uptake and preventing excitotoxic death. This establishes Mcu and the pathways regulating its expression as important determinants of excitotoxicity, which may represent therapeutic targets for excitotoxic disorders.
Collapse
|
39
|
Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol 2013; 115:157-88. [PMID: 24361499 DOI: 10.1016/j.pneurobio.2013.11.006] [Citation(s) in RCA: 826] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/28/2013] [Accepted: 11/29/2013] [Indexed: 01/22/2023]
Abstract
Excitotoxicity, the specific type of neurotoxicity mediated by glutamate, may be the missing link between ischemia and neuronal death, and intervening the mechanistic steps that lead to excitotoxicity can prevent stroke damage. Interest in excitotoxicity began fifty years ago when monosodium glutamate was found to be neurotoxic. Evidence soon demonstrated that glutamate is not only the primary excitatory neurotransmitter in the adult brain, but also a critical transmitter for signaling neurons to degenerate following stroke. The finding led to a number of clinical trials that tested inhibitors of excitotoxicity in stroke patients. Glutamate exerts its function in large by activating the calcium-permeable ionotropic NMDA receptor (NMDAR), and different subpopulations of the NMDAR may generate different functional outputs, depending on the signaling proteins directly bound or indirectly coupled to its large cytoplasmic tail. Synaptic activity activates the GluN2A subunit-containing NMDAR, leading to activation of the pro-survival signaling proteins Akt, ERK, and CREB. During a brief episode of ischemia, the extracellular glutamate concentration rises abruptly, and stimulation of the GluN2B-containing NMDAR in the extrasynaptic sites triggers excitotoxic neuronal death via PTEN, cdk5, and DAPK1, which are directly bound to the NMDAR, nNOS, which is indirectly coupled to the NMDAR via PSD95, and calpain, p25, STEP, p38, JNK, and SREBP1, which are further downstream. This review aims to provide a comprehensive summary of the literature on excitotoxicity and our perspectives on how the new generation of excitotoxicity inhibitors may succeed despite the failure of the previous generation of drugs.
Collapse
Affiliation(s)
- Ted Weita Lai
- Graduate Institute of Clinical Medical Science, China Medical University, 91 Hsueh-Shih Road, 40402 Taichung, Taiwan; Translational Medicine Research Center, China Medical University Hospital, 2 Yu-De Road, 40447 Taichung, Taiwan.
| | - Shu Zhang
- Translational Medicine Research Center, China Medical University Hospital, 2 Yu-De Road, 40447 Taichung, Taiwan; Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, V6T 2B5 Vancouver, Canada
| | - Yu Tian Wang
- Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, V6T 2B5 Vancouver, Canada.
| |
Collapse
|
40
|
Yu H, Popescu GK. Inhibition of GluN2A-containing N-methyl-D-aspartate receptors by 2-naphthoic acid. Mol Pharmacol 2013; 84:541-50. [PMID: 23873856 PMCID: PMC3781379 DOI: 10.1124/mol.113.087189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/19/2013] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-D-aspartate (NMDA) receptors mediate excitatory synaptic transmission in the central nervous system and play important roles in synaptic development and plasticity, but also mediate glutamate neurotoxicity. Recently, 2-naphthoic acid (NPA) and its derivatives have been identified as allosteric, noncompetitive NMDA receptor inhibitors. The selectivity of NPA derivatives among NMDA receptor subtypes was mapped structurally to the ligand-binding domain, and was proposed to be mediated by residues on the S1 segment. To delineate the kinetic mechanism by which NPA inhibits NMDA receptor activity, we examined its effects on the NMDA receptor gating reaction. Using whole-cell patch clamping on human embryonic kidney 293 cells expressing recombinant NMDA family of glutamate receptor subunits, GluN1/GluN2A, we found that NPA has a 50% inhibitory effect at 1.9 mM. Further, from one-channel current recordings, we found that 4 mM NPA caused a 62% decrease in open probability by decreasing mean open time 2.5-fold and by increasing mean closed time 2-fold. Kinetic modeling suggested that NPA binding stabilized NMDA receptor closed states and increased the energy barriers toward open states, causing NMDA receptors to dwell longer in pre-open states along the activation pathway. The reaction mechanisms we derived provide quantitative insight into the inhibitory mechanism of NPA and help anticipate its effects on GluN1/GluN2A receptors during both physiologic and pathologic activation modalities.
Collapse
Affiliation(s)
- Han Yu
- Neuroscience Program (H.Y., G.K.P.) and Department of Biochemistry (G.K.P.), School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | | |
Collapse
|
41
|
Ehrnhoefer DE, Skotte NH, Ladha S, Nguyen YTN, Qiu X, Deng Y, Huynh KT, Engemann S, Nielsen SM, Becanovic K, Leavitt BR, Hasholt L, Hayden MR. p53 increases caspase-6 expression and activation in muscle tissue expressing mutant huntingtin. Hum Mol Genet 2013; 23:717-29. [PMID: 24070868 DOI: 10.1093/hmg/ddt458] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Activation of caspase-6 in the striatum of both presymptomatic and affected persons with Huntington's disease (HD) is an early event in the disease pathogenesis. However, little is known about the role of caspase-6 outside the central nervous system (CNS) and whether caspase activation might play a role in the peripheral phenotypes, such as muscle wasting observed in HD. We assessed skeletal muscle tissue from HD patients and well-characterized mouse models of HD. Cleavage of the caspase-6 specific substrate lamin A is significantly increased in skeletal muscle obtained from HD patients as well as in muscle tissues from two different HD mouse models. p53, a transcriptional activator of caspase-6, is upregulated in neuronal cells and tissues expressing mutant huntingtin. Activation of p53 leads to a dramatic increase in levels of caspase-6 mRNA, caspase-6 activity and cleavage of lamin A. Using mouse embryonic fibroblasts (MEFs) from YAC128 mice, we show that this increase in caspase-6 activity can be mitigated by pifithrin-α (pifα), an inhibitor of p53 transcriptional activity, but not through the inhibition of p53's mitochondrial pro-apoptotic function. Remarkably, the p53-mediated increase in caspase-6 expression and activation is exacerbated in cells and tissues of both neuronal and peripheral origin expressing mutant huntingtin (Htt). These findings suggest that the presence of the mutant Htt protein enhances p53 activity and lowers the apoptotic threshold, which activates caspase-6. Furthermore, these results suggest that this pathway is activated both within and outside the CNS in HD and may contribute to both loss of CNS neurons and muscle atrophy.
Collapse
Affiliation(s)
- Dagmar E Ehrnhoefer
- Centre for Molecular Medicine and Therapeutics (CMMT), Department of Medical Genetics, CFRI, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Synaptic activity initiates biochemical processes that have various outcomes, including the formation of memories, increases in neuronal survival and the development of chronic pain and addiction. Virtually all activity-induced, long-lasting adaptations of brain functions require a dialogue between synapses and the nucleus that results in changes in gene expression. Calcium signals that are induced by synaptic activity and propagate into the nucleus are a major route for synapse-to-nucleus communication. Recent findings indicate that diverse forms of neuroadaptation require calcium transients in the nucleus to switch on the necessary genomic programme. Deficits in nuclear calcium signalling as a result of a reduction in synaptic activity or increased extrasynaptic NMDA receptor signalling may underlie the aetiologies of various diseases, including neurodegeneration and cognitive dysfunction.
Collapse
Affiliation(s)
- Hilmar Bading
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), University of Heidelberg, INF 364, 69120 Heidelberg, Germany. Hilmar.Bading@ uni-hd.de
| |
Collapse
|
43
|
Mehta A, Prabhakar M, Kumar P, Deshmukh R, Sharma PL. Excitotoxicity: bridge to various triggers in neurodegenerative disorders. Eur J Pharmacol 2012; 698:6-18. [PMID: 23123057 DOI: 10.1016/j.ejphar.2012.10.032] [Citation(s) in RCA: 476] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 09/26/2012] [Accepted: 10/06/2012] [Indexed: 12/13/2022]
Abstract
Glutamate is one of the most prominent neurotransmitter in the body, present in over 50% of nervous tissue and plays an important role in neuronal excitation. This neuronal excitation is short-lived and is followed by depression. Multiple abnormal triggers such as energy deficiency, oxidative stress, mitochondrial dysfunction, calcium overload, etc can lead to aberration in neuronal excitation process. Such an aberration, serves as a common pool or bridge between abnormal triggers and deleterious signaling processes with which central neurons cannot cope up, leading to death. Excitotoxicity is the pathological process by which nerve cells are damaged and killed by excessive stimulation by neurotransmitters such as glutamate and similar substances. Such excitotoxic neuronal death has been implicated in spinal cord injury, stroke, traumatic brain injury, hearing loss and in neurodegenerative diseases of the central nervous system such as stroke, epilepsy, multiple sclerosis, Alzheimer disease, Amyltropic lateral sclerosis, Parkinson's disease, Huntington disease and alcohol withdrawal. This review mainly emphasizes the triggering events which sustain neuronal excitation, role of calcium, mitochondrial dysfunction, ROS, NO, chloride homeostasis and eicosanoids pathways. Further, a brief introduction about the recent research occurring in the treatment of various neurodegenerative diseases, including a summary of the presumed physiologic mechanisms behind the pharmacology of these disorders.
Collapse
Affiliation(s)
- Ankita Mehta
- Neuropharmacology Division, ISF College of Pharmacy, Ferozpur Road, Ghal Kalan, Moga 142 001, Punjab, India
| | | | | | | | | |
Collapse
|
44
|
Bas-Orth C, Bading H. The divergence-convergence model of acquired neuroprotection. Mech Dev 2012; 130:396-401. [PMID: 23063506 DOI: 10.1016/j.mod.2012.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 09/25/2012] [Accepted: 09/26/2012] [Indexed: 12/27/2022]
Abstract
It is commonly known that mental activity helps to maintain a healthy brain. Recent research has unraveled the underlying molecular mechanisms that explain why an active brain lives longer. These mechanisms involve the activation of a comprehensive transcriptional program that is triggered by enhanced synaptic activity and renders neurons resistant to harmful conditions. Functionally, this state of acquired neuroprotection may be achieved mainly via one mechanism, which is the stabilization of mitochondria. In this review we propose a model that describes the signaling network that links synaptic activity to neuroprotection. We suggest that the divergent-convergent architecture of this signaling network ensures both robust and reliable as well as persistent activation of the neuroprotective machinery.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | | |
Collapse
|
45
|
Tauskela JS, Aylsworth A, Hewitt M, Brunette E, Mealing GAR. Preconditioning induces tolerance by suppressing glutamate release in neuron culture ischemia models. J Neurochem 2012; 122:470-81. [DOI: 10.1111/j.1471-4159.2012.07791.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
46
|
Tan YW, Zhang SJ, Hoffmann T, Bading H. Increasing levels of wild-type CREB up-regulates several activity-regulated inhibitor of death (AID) genes and promotes neuronal survival. BMC Neurosci 2012; 13:48. [PMID: 22607375 PMCID: PMC3407521 DOI: 10.1186/1471-2202-13-48] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 05/18/2012] [Indexed: 12/24/2022] Open
Abstract
Background CREB (cAMP-response element binding protein) is the prototypical signal-regulated transcription factor. In neurons, it is the target of the synaptic activity-induced nuclear calcium-calcium/calmodulin dependent protein kinase (CaMK) IV signaling pathway that controls the expression of genes important for acquired neuroprotection as well as other long-lasting adaptive processes in the nervous system. The function of CREB as a transcriptional activator is controlled by its phosphorylation on serine 133, which can be catalyzed by CaMKIV and leads to the recruitment of the co-activator, CREB binding protein (CBP). Activation of CBP function by nuclear calcium-CaMKIV signaling is a second regulatory step required for CREB/CBP-mediated transcription. Results Here we used recombinant adeno-associated virus (rAAV) to increase the levels of wild type CREB or to overexpress a mutant version of CREB (mCREB) containing a serine to alanine mutation at position amino acid 133 in mouse hippocampal neurons. Increasing the levels of CREB was sufficient to boost neuroprotective activity even under basal conditions (i.e., in the absence of stimulation of synaptic activity). In contrast, overexpression of mCREB increased cell death. The ratio of phospho(serine 133)CREB to CREB immunoreactivity in unstimulated hippocampal neurons was similar for endogenous CREB and overexpressed wild type CREB and, as expected, dramatically reduced for overexpressed mCREB. A gene expression analysis revealed that increased expression of CREB but not that of mCREB in hippocampal neurons led to elevated expression levels of bdnf as well as that of several members of a previously characterized set of Activity-regulated Inhibitor of Death (AID) genes, which include atf3, btg2, gadd45β, and gadd45γ. Conclusions Our findings indicate that the expression levels of wild type CREB are a critical determinant of the ability of hippocampal neurons to survive harmful conditions. Increasing the levels of wild type CREB can, even without inducing synaptic activity, increase pro-survival gene expression and strengthen the neurons’ neuroprotective shield. The observed degradation of CREB protein following NMDA treatment of hippocampal neurons suggests that the known CREB shut-off associated with extrasynaptic NMDA receptor-induced excitotoxicity is followed by CREB proteolysis.
Collapse
Affiliation(s)
- Yan-Wei Tan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| | | | | | | |
Collapse
|
47
|
Graham RK, Ehrnhoefer DE, Hayden MR. Caspase-6 and neurodegeneration. Trends Neurosci 2011; 34:646-56. [DOI: 10.1016/j.tins.2011.09.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 09/02/2011] [Accepted: 09/13/2011] [Indexed: 01/10/2023]
|
48
|
Jebelli JD, Hooper C, Garden GA, Pocock JM. Emerging roles of p53 in glial cell function in health and disease. Glia 2011; 60:515-25. [PMID: 22105777 DOI: 10.1002/glia.22268] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 10/21/2011] [Indexed: 12/12/2022]
Abstract
Emerging evidence suggests that p53, a tumor suppressor protein primarily involved in cancer biology, coordinates a wide range of novel functions in the CNS including the mediation of pathways underlying neurodegenerative disease pathogenesis. Moreover, an evolving concept in cell and molecular neuroscience is that glial cells are far more fundamental to disease progression than previously thought, which may occur via a noncell-autonomous mechanism that is heavily dependent on p53 activities. As a crucial hub connecting many intracellular control pathways, including cell-cycle control and apoptosis, p53 is ideally placed to coordinate the cellular response to a range of stresses. Although neurodegenerative diseases each display a distinct and diverse molecular pathology, apoptosis is a widespread hallmark feature and the multimodal capacity of the p53 system to orchestrate apoptosis and glial cell behavior highlights p53 as a potential unifying target for therapeutic intervention in neurodegeneration.
Collapse
Affiliation(s)
- Joseph D Jebelli
- Department of Neuroinflammation, UCL Institute of Neurology, London, United Kingdom
| | | | | | | |
Collapse
|
49
|
Hagenston AM, Bading H. Calcium signaling in synapse-to-nucleus communication. Cold Spring Harb Perspect Biol 2011; 3:a004564. [PMID: 21791697 DOI: 10.1101/cshperspect.a004564] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Changes in the intracellular concentration of calcium ions in neurons are involved in neurite growth, development, and remodeling, regulation of neuronal excitability, increases and decreases in the strength of synaptic connections, and the activation of survival and programmed cell death pathways. An important aspect of the signals that trigger these processes is that they are frequently initiated in the form of glutamatergic neurotransmission within dendritic trees, while their completion involves specific changes in the patterns of genes expressed within neuronal nuclei. Accordingly, two prominent aims of research concerned with calcium signaling in neurons are determination of the mechanisms governing information conveyance between synapse and nucleus, and discovery of the rules dictating translation of specific patterns of inputs into appropriate and specific transcriptional responses. In this article, we present an overview of the avenues by which glutamatergic excitation of dendrites may be communicated to the neuronal nucleus and the primary calcium-dependent signaling pathways by which synaptic activity can invoke changes in neuronal gene expression programs.
Collapse
Affiliation(s)
- Anna M Hagenston
- CellNetworks-Cluster of Excellence, Department of Neurobiology, Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | | |
Collapse
|
50
|
Nuclear Calcium-VEGFD Signaling Controls Maintenance of Dendrite Arborization Necessary for Memory Formation. Neuron 2011; 71:117-30. [DOI: 10.1016/j.neuron.2011.04.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2011] [Indexed: 01/17/2023]
|