1
|
Xavier AM, Lin Q, Kang CJ, Cheadle L. A single-cell transcriptomic atlas of sensory-dependent gene expression in developing mouse visual cortex. Development 2025; 152:dev204244. [PMID: 40018816 DOI: 10.1242/dev.204244] [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: 07/12/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Sensory experience drives the maturation of neural circuits during postnatal brain development through molecular mechanisms that remain to be fully elucidated. One likely mechanism involves the sensory-dependent expression of genes that encode direct mediators of circuit remodeling within developing cells. To identify potential drivers of sensory-dependent synaptic development, we generated a single-nucleus RNA sequencing dataset describing the transcriptional responses of cells in the mouse visual cortex to sensory deprivation or to stimulation during a developmental window when visual input is necessary for circuit refinement. We sequenced 118,529 nuclei across 16 neuronal and non-neuronal cell types isolated from control, sensory deprived and sensory stimulated mice, identifying 1268 sensory-induced genes within the developing brain. While experience elicited transcriptomic changes in all cell types, excitatory neurons in layer 2/3 exhibited the most robust changes, and the sensory-induced genes in these cells are poised to strengthen synapse-to-nucleus crosstalk and to promote cell type-specific axon guidance pathways. Altogether, we expect this dataset to significantly broaden our understanding of the molecular mechanisms through which sensory experience shapes neural circuit wiring in the developing brain.
Collapse
Affiliation(s)
- Andre M Xavier
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Qianyu Lin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Chris J Kang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
2
|
Zent KH, Dell'Acqua ML. Synapse-to-Nucleus ERK→CREB Transcriptional Signaling Requires Dendrite-to-Soma Ca 2+ Propagation Mediated by L-Type Voltage-Gated Ca 2+ Channels. J Neurosci 2025; 45:e1216242024. [PMID: 39562039 PMCID: PMC11756630 DOI: 10.1523/jneurosci.1216-24.2024] [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: 06/27/2024] [Revised: 10/02/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024] Open
Abstract
The cAMP-response element-binding protein (CREB) transcription factor controls the expression of the neuronal immediate early genes c-fos, Arc, and Bdnf and is essential for long-lasting synaptic plasticity underlying learning and memory. Despite this critical role, there is still ongoing debate regarding the synaptic excitation-transcription (E-T) coupling mechanisms mediating CREB activation in the nucleus. Here we employed optical uncaging of glutamate to mimic synaptic excitation of distal dendrites in conjunction with simultaneous imaging of intracellular Ca2+ dynamics and transcriptional reporter gene expression to elucidate CREB E-T coupling mechanisms in hippocampal neurons cultured from both male and female rats. Using this approach, we found that CREB-dependent transcription was engaged following dendritic stimulation of N-methyl-d-aspartate receptors (NMDARs) only when Ca2+ signals propagated to the soma via subsequent activation of L-type voltage-gated Ca2+ channels resulting in activation of extracellular signal-regulated kinase MAP kinase signaling to sustain CREB phosphorylation in the nucleus. In contrast, dendrite-restricted Ca2+ signals generated by NMDARs failed to stimulate CREB-dependent transcription. Furthermore, Ca2+-CaM-dependent kinase-mediated signaling pathways that may transiently contribute to CREB phosphorylation following stimulation were ultimately dispensable for downstream CREB-dependent transcription and c-Fos induction. These findings emphasize the essential role that L-type Ca2+ channels play in rapidly relaying signals over long distances from synapses located on distal dendrites to the nucleus to control gene expression.
Collapse
Affiliation(s)
- Katlin H Zent
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neurotechnology Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
3
|
Ferreira IL, Marinho D, de Rosa V, Castanheira B, Fang Z, Caldeira GL, Mota SI, Rego AC. Linking activation of synaptic NMDA receptors-induced CREB signaling to brief exposure of cortical neurons to oligomeric amyloid-beta peptide. J Neurochem 2025; 169:e16222. [PMID: 39263896 DOI: 10.1111/jnc.16222] [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: 02/19/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
Amyloid-beta peptide oligomers (AβO) have been considered "primum movens" for a cascade of events that ultimately cause selective neuronal death in Alzheimer's disease (AD). However, initial events triggered by AβO have not been clearly defined. Synaptic (Syn) N-methyl-d-aspartate receptors (NMDAR) are known to activate cAMP response element-binding protein (CREB), a transcriptional factor involved in gene expression related to cell survival, memory formation and synaptic plasticity, whereas activation of extrasynaptic (ESyn) NMDARs was linked to excitotoxic events. In AD brain, CREB phosphorylation/activation was shown to be altered, along with dyshomeostasis of intracellular Ca2+ (Ca2+ i). Thus, in this work, we analyze acute/early and long-term AβO-mediated changes in CREB activation involving Syn or ESyn NMDARs in mature rat cortical neurons. Our findings show that acute AβO exposure produce early increase in phosphorylated CREB, reflecting CREB activity, in a process occurring through Syn NMDAR-mediated Ca2+ influx. Data also demonstrate that AβO long-term (24 h) exposure compromises synaptic function related to Ca2+-dependent CREB phosphorylation/activation and nuclear CREB levels and related target genes, namely Bdnf, Gadd45γ, and Btg2. Data suggest a dual effect of AβO following early or prolonged exposure in mature cortical neurons through the activation of the CREB signaling pathway, linked to the activation of Syn NMDARs.
Collapse
Affiliation(s)
- I Luísa Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Daniela Marinho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Valéria de Rosa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bárbara Castanheira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Zongwei Fang
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Gladys L Caldeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Sandra I Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Xavier AM, Lin Q, Kang CJ, Cheadle L. A single-cell transcriptomic atlas of sensory-dependent gene expression in developing mouse visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600673. [PMID: 38979325 PMCID: PMC11230371 DOI: 10.1101/2024.06.25.600673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensory experience drives the refinement and maturation of neural circuits during postnatal brain development through molecular mechanisms that remain to be fully elucidated. One likely mechanism involves the sensory-dependent expression of genes that encode direct mediators of circuit remodeling within developing cells. However, while studies in adult systems have begun to uncover crucial roles for sensory-induced genes in modifying circuit connectivity, the gene programs induced by brain cells in response to sensory experience during development remain to be fully characterized. Here, we present a single-nucleus RNA-sequencing dataset describing the transcriptional responses of cells in mouse visual cortex to sensory deprivation or sensory stimulation during a developmental window when visual input is necessary for circuit refinement. We sequenced 118,529 individual nuclei across sixteen neuronal and non-neuronal cortical cell types isolated from control, sensory deprived, and sensory stimulated mice, identifying 1,268 unique sensory-induced genes within the developing brain. To demonstrate the utility of this resource, we compared the architecture and ontology of sensory-induced gene programs between cell types, annotated transcriptional induction and repression events based upon RNA velocity, and discovered Neurexin and Neuregulin signaling networks that underlie cell-cell interactions via CellChat . We find that excitatory neurons, especially layer 2/3 pyramidal neurons, are highly sensitive to sensory stimulation, and that the sensory-induced genes in these cells are poised to strengthen synapse-to-nucleus crosstalk by heightening protein serine/threonine kinase activity. Altogether, we expect this dataset to significantly broaden our understanding of the molecular mechanisms through which sensory experience shapes neural circuit wiring in the developing brain.
Collapse
|
5
|
Di Liegro CM, Schiera G, Schirò G, Di Liegro I. Role of Post-Transcriptional Regulation in Learning and Memory in Mammals. Genes (Basel) 2024; 15:337. [PMID: 38540396 PMCID: PMC10970538 DOI: 10.3390/genes15030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 06/14/2024] Open
Abstract
After many decades, during which most molecular studies on the regulation of gene expression focused on transcriptional events, it was realized that post-transcriptional control was equally important in order to determine where and when specific proteins were to be synthesized. Translational regulation is of the most importance in the brain, where all the steps of mRNA maturation, transport to different regions of the cells and actual expression, in response to specific signals, constitute the molecular basis for neuronal plasticity and, as a consequence, for structural stabilization/modification of synapses; notably, these latter events are fundamental for the highest brain functions, such as learning and memory, and are characterized by long-term potentiation (LTP) of specific synapses. Here, we will discuss the molecular bases of these fundamental events by considering both the role of RNA-binding proteins (RBPs) and the effects of non-coding RNAs involved in controlling splicing, editing, stability and translation of mRNAs. Importantly, it has also been found that dysregulation of mRNA metabolism/localization is involved in many pathological conditions, arising either during brain development or in the adult nervous system.
Collapse
Affiliation(s)
- Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (C.M.D.L.); (G.S.)
| | - Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (C.M.D.L.); (G.S.)
| | - Giuseppe Schirò
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
- Neurology and Multiple Sclerosis Center, Unità Operativa Complessa (UOC), Foundation Institute “G. Giglio”, 90015 Cefalù, Italy
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
6
|
Ma H, Khaled HG, Wang X, Mandelberg NJ, Cohen SM, He X, Tsien RW. Excitation-transcription coupling, neuronal gene expression and synaptic plasticity. Nat Rev Neurosci 2023; 24:672-692. [PMID: 37773070 PMCID: PMC12024187 DOI: 10.1038/s41583-023-00742-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Excitation-transcription coupling (E-TC) links synaptic and cellular activity to nuclear gene transcription. It is generally accepted that E-TC makes a crucial contribution to learning and memory through its role in underpinning long-lasting synaptic enhancement in late-phase long-term potentiation and has more recently been linked to late-phase long-term depression: both processes require de novo gene transcription, mRNA translation and protein synthesis. E-TC begins with the activation of glutamate-gated N-methyl-D-aspartate-type receptors and voltage-gated L-type Ca2+ channels at the membrane and culminates in the activation of transcription factors in the nucleus. These receptors and ion channels mediate E-TC through mechanisms that include long-range signalling from the synapse to the nucleus and local interactions within dendritic spines, among other possibilities. Growing experimental evidence links these E-TC mechanisms to late-phase long-term potentiation and learning and memory. These advances in our understanding of the molecular mechanisms of E-TC mean that future efforts can focus on understanding its mesoscale functions and how it regulates neuronal network activity and behaviour in physiological and pathological conditions.
Collapse
Affiliation(s)
- Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China.
| | - Houda G Khaled
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Xiaohan Wang
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Nataniel J Mandelberg
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Samuel M Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
7
|
Grochowska KM, Sperveslage M, Raman R, Failla AV, Głów D, Schulze C, Laprell L, Fehse B, Kreutz MR. Chaperone-mediated autophagy in neuronal dendrites utilizes activity-dependent lysosomal exocytosis for protein disposal. Cell Rep 2023; 42:112998. [PMID: 37590146 DOI: 10.1016/j.celrep.2023.112998] [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: 02/18/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The complex morphology of neurons poses a challenge for proteostasis because the majority of lysosomal degradation machinery is present in the cell soma. In recent years, however, mature lysosomes were identified in dendrites, and a fraction of those appear to fuse with the plasma membrane and release their content to the extracellular space. Here, we report that dendritic lysosomes are heterogeneous in their composition and that only those containing lysosome-associated membrane protein (LAMP) 2A and 2B fuse with the membrane and exhibit activity-dependent motility. Exocytotic lysosomes dock in close proximity to GluN2B-containing N-methyl-D-aspartate-receptors (NMDAR) via an association of LAMP2B to the membrane-associated guanylate kinase family member SAP102/Dlg3. NMDAR-activation decreases lysosome motility and promotes membrane fusion. We find that chaperone-mediated autophagy is a supplier of content that is released to the extracellular space via lysosome exocytosis. This mechanism enables local disposal of aggregation-prone proteins like TDP-43 and huntingtin.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Marit Sperveslage
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Rajeev Raman
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Antonio V Failla
- UKE Microscopic Imaging Facility (umif), University Medical Center Eppendorf, 20251 Hamburg, Germany
| | - Dawid Głów
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Christian Schulze
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Laura Laprell
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
| |
Collapse
|
8
|
Karpova A, Samer S, Turacak R, Yuanxiang P, Kreutz MR. Integration of nuclear Ca 2+ transients and subnuclear protein shuttling provides a novel mechanism for the regulation of CREB-dependent gene expression. Cell Mol Life Sci 2023; 80:228. [PMID: 37491479 PMCID: PMC10368568 DOI: 10.1007/s00018-023-04876-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023]
Abstract
Nuclear Ca2+ waves elicited by NMDAR and L-type voltage-gated Ca2+-channels as well as protein transport from synapse-to-nucleus are both instrumental in control of plasticity-related gene expression. At present it is not known whether fast [Ca2+]n transients converge in the nucleus with signaling of synapto-nuclear protein messenger. Jacob is a protein that translocate a signalosome from N-methyl-D-aspartate receptors (NMDAR) to the nucleus and that docks this signalosome to the transcription factor CREB. Here we show that the residing time of Jacob in the nucleoplasm strictly correlates with nuclear [Ca2+]n transients elicited by neuronal activity. A steep increase in [Ca2+]n induces instantaneous uncoupling of Jacob from LaminB1 at the nuclear lamina and promotes the association with the transcription factor cAMP-responsive element-binding protein (CREB) in hippocampal neurons. The size of the Jacob pool at the nuclear lamina is controlled by previous activity-dependent nuclear import, and thereby captures the previous history of NMDAR-induced nucleocytoplasmic shuttling. Moreover, the localization of Jacob at the nuclear lamina strongly correlates with synaptic activity and [Ca2+]n waves reflecting ongoing neuronal activity. In consequence, the resulting extension of the nuclear residing time of Jacob amplifies the capacity of the Jacob signalosome to regulate CREB-dependent gene expression and will, thereby, compensate for the relatively small number of molecules reaching the nucleus from individual synapses.
Collapse
Affiliation(s)
- Anna Karpova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto von Guericke University, 39106, Magdeburg, Germany.
| | - Sebastian Samer
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Rabia Turacak
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - PingAn Yuanxiang
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto von Guericke University, 39106, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
9
|
Andres-Alonso M, Grochowska KM, Gundelfinger ED, Karpova A, Kreutz MR. Protein transport from pre- and postsynapse to the nucleus: Mechanisms and functional implications. Mol Cell Neurosci 2023; 125:103854. [PMID: 37084990 DOI: 10.1016/j.mcn.2023.103854] [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: 01/13/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
The extreme length of neuronal processes poses a challenge for synapse-to-nucleus communication. In response to this challenge several different mechanisms have evolved in neurons to couple synaptic activity to the regulation of gene expression. One of these mechanisms concerns the long-distance transport of proteins from pre- and postsynaptic sites to the nucleus. In this review we summarize current evidence on mechanisms of transport and consequences of nuclear import of these proteins for gene transcription. In addition, we discuss how information from pre- and postsynaptic sites might be relayed to the nucleus by this type of long-distance signaling. When applicable, we highlight how long-distance protein transport from synapse-to-nucleus can provide insight into the pathophysiology of disease or reveal new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Maria Andres-Alonso
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Katarzyna M Grochowska
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Eckart D Gundelfinger
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; Institute of Pharmacology and Toxicology, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Anna Karpova
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
| |
Collapse
|
10
|
da Rosa MM, de Amorim LC, Alves JVDO, Aguiar IFDS, Oliveira FGDS, da Silva MV, dos Santos MTC. The promising role of natural products in Alzheimer's disease. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2022.100049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
11
|
Grochowska KM, Bär J, Gomes GM, Kreutz MR, Karpova A. Jacob, a Synapto-Nuclear Protein Messenger Linking N-methyl-D-aspartate Receptor Activation to Nuclear Gene Expression. Front Synaptic Neurosci 2021; 13:787494. [PMID: 34899262 PMCID: PMC8662305 DOI: 10.3389/fnsyn.2021.787494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Pyramidal neurons exhibit a complex dendritic tree that is decorated by a huge number of spine synapses receiving excitatory input. Synaptic signals not only act locally but are also conveyed to the nucleus of the postsynaptic neuron to regulate gene expression. This raises the question of how the spatio-temporal integration of synaptic inputs is accomplished at the genomic level and which molecular mechanisms are involved. Protein transport from synapse to nucleus has been shown in several studies and has the potential to encode synaptic signals at the site of origin and decode them in the nucleus. In this review, we summarize the knowledge about the properties of the synapto-nuclear messenger protein Jacob with special emphasis on a putative role in hippocampal neuronal plasticity. We will elaborate on the interactome of Jacob, the signals that control synapto-nuclear trafficking, the mechanisms of transport, and the potential nuclear function. In addition, we will address the organization of the Jacob/NSMF gene, its origin and we will summarize the evidence for the existence of splice isoforms and their expression pattern.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Research Group (RG) Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Julia Bär
- Research Group (RG) Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Research Group (RG) Neuronal Protein Transport, University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology Hamburg, Hamburg, Germany.,Research Group (RG) Optobiology, Institute of Biology, HU Berlin, Berlin, Germany
| | - Guilherme M Gomes
- Research Group (RG) Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Michael R Kreutz
- Research Group (RG) Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology Hamburg, Hamburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,German Research Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Anna Karpova
- Research Group (RG) Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
12
|
Li NQ, Peng Z, Xu WW, An K, Wan L. Bone mesenchymal stem cells attenuate resiniferatoxin-induced neuralgia via inhibiting TRPA1-PKCδ-P38/MAPK-p-P65 pathway in mice. Brain Res Bull 2021; 174:92-102. [PMID: 34098041 DOI: 10.1016/j.brainresbull.2021.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Treatment of neuropathic pain (NP) resulting from nerve injury is one of the most complicated and challenging in modern practice. Pharmacological treatments for NP are not fully effectively and novel approaches are requisite. Recently, transplantation of bone mesenchymal stem cells (BMSCs) has represented a promising approach for pain relief and neural repair, but how it produces beneficial effects on resiniferatoxin (RTX) induced nerve injury is still unclear. Here, we identified the BMSCs' analgesic effects and their potential mechanisms of microglial cells activation on RTX induced neuralgia. Immunostaining, biochemical studies demonstrated that microglia rather than astrocyte cells activation involved in RTX induced mechanical hyperalgesia, whereas the GFP-labeled BMSCs alleviated this mechanical hyperalgesia. Moreover, pain-related TRPA1, PKCδ, CaMKIIɑ (Calcium/calmodulin dependent protein kinase II), P38/MAPK (mitogen-activated protein kinase), p-P65 activation and expression in the spinal cord were significantly inhibited after BMSC administration. In addition, BMSCs treated RTX mice displayed a lower density of mushroom dendritic spines. Our research suggested that activation of PKCδ-CaMKIIɑ-P38/MAPK-p-P65 pathway and mushroom dendritic spines abnormal increase in the spinal cord is the main mechanism of RTX induced neuropathic pain, and transplant of BMSCs to the damaged nerve may offer promising approach for neuropathic pain.
Collapse
Affiliation(s)
- Nan-Qi Li
- Department of Anesthesiology, The first Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, PR China
| | - Zhe Peng
- Department of Pain Management, The Second Affiliated Hospital, Guangzhou Medical University, State Key Clinical Specialty in Pain Medicine, Guangzhou, Guangdong, 510260, PR China
| | - Wen-Wen Xu
- Department of Anesthesiology, The first Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, PR China
| | - Ke An
- Department of Anesthesiology, The first Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, PR China.
| | - Li Wan
- Department of Pain Management, The Second Affiliated Hospital, Guangzhou Medical University, State Key Clinical Specialty in Pain Medicine, Guangzhou, Guangdong, 510260, PR China.
| |
Collapse
|
13
|
Unveiling the pathogenesis of perineural invasion from the perspective of neuroactive molecules. Biochem Pharmacol 2021; 188:114547. [PMID: 33838132 DOI: 10.1016/j.bcp.2021.114547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022]
Abstract
Perineural invasion (PNI) is characterized by an encounter between the cancer cells and neuronal fibers and holds an extremely poor prognosis for malignant tumors. The exact molecular mechanism behind PNI yet remains to be explored. However, it is worth-noting that an involvement of the neuroactive molecules plays a major part in this process. A complex signaling network comprising the interplay between immunological cascades and neurogenic molecules such as tumor-derived neurotrophins, neuromodulators, and growth factors constitutes an active microenvironment for PNI associated with malignancy. The present review aims at discussing the following points in relation to PNI: a) Communication between PNI and neuroplasticity mechanisms can explain the pathophysiology of poor, short and long-term outcomes in cancer patients; b) Neuroactive molecules can significantly alter the neurons and cancer cells so as to sustain PNI progression; c) Finally, careful manipulation of neurogenic pathways and/or their crosstalk with the immunological molecules implicated in PNI could provide a potential breakthrough in cancer therapeutics.
Collapse
|
14
|
Olevska A, Spanagel R, Bernardi RE. Impaired contextual fear conditioning in RasGRF2 mutant mice is likely Ras-ERK-dependent. Neurobiol Learn Mem 2021; 181:107435. [PMID: 33831510 DOI: 10.1016/j.nlm.2021.107435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Ras/Raf/MEK/ERK (Ras-ERK) signaling has been shown to play an important role in fear acquisition. However, little information is known regarding the mechanisms that contribute to the regulation of this pathway in terms of the learning of conditioned fears. Ras Guanine Nucleotide Releasing Factor 2 (RasGRF2) is one of two guanine nucleotide exchange factors (GEF) that regulates the Ras-ERK signaling pathway in a Ca2+-dependent manner via control of the cycling of Ras isoforms between an inactive and active state. Here we sought to determine the role of RasGRF2 on contextual fear conditioning in RasGRF2 knockout (KO) and their wild type (WT) counterparts. Male KO and WT mice underwent a single session of contextual fear conditioning (12 min, 4 unsignaled shocks), followed by either daily 12-min retention trials or the molecular analysis of Ras activation and pERK1/2 activity. KO mice showed an impaired acquisition of contextual fear, as demonstrated by reduced freezing during fear conditioning and 24-hr retention tests relative to WT mice. Ras analysis following fear conditioning demonstrated a reduction in Ras activation in the hippocampus as well as a reduction in pERK1/2 in the CA1 region of the hippocampus in KO mice, suggesting that the decrease in fear conditioning in KO mice is at least in part due to the impairment of Ras-ERK signaling in the hippocampus during learning. These data indicate a role for RasGRF2 in contextual fear conditioning in mice that may be Ras-ERK-dependent.
Collapse
Affiliation(s)
- Anastasia Olevska
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.
| |
Collapse
|
15
|
Ashourpour F, Jafari A, Babaei P. Chronic administration of Tat-GluR23Y ameliorates cognitive dysfunction targeting CREB signaling in rats with amyloid beta neurotoxicity. Metab Brain Dis 2021; 36:701-709. [PMID: 33420884 DOI: 10.1007/s11011-020-00662-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 12/25/2020] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is behaviorally characterized by memory impairments, and pathologically by amyloid β1-42 (Aβ1-42) plaques and tangles. Aβ binds to excitatory synapses and disrupts their transmission due to dysregulation of the glutamate receptors. Here we hypothesized that chronic inhibition of the endocytosis of AMPA receptors together with GluN2B subunit of NMDA receptors might improve cognition deficit induced by Aβ(1-42) neurotoxicity. Forty male Wistar rats were used in this study and divided into 5 groups: Saline + Saline, Aβ+Saline, Aβ+Ifen (Ifenprodil, 3 nmol /2 weeks), Aβ+GluR23Y (Tat-GluR23Y 3 μmol/kg/2 weeks) and Aβ+Ifen+GluR23Y (same doses and durations). Aβ(1-42) neurotoxicity was induced by intracerebroventricular (ICV) injection of Aβ1-42 (2 μg/μl/side), and then animals received the related treatments for 14 days. Cognitive performance of rats and hippocampal level of cAMP-response element-binding (CREB) were evaluated using Morris Water Maze (MWM), and western blotting respectively. Obtained data from the acquisition trials were analyzed by two way Anova and Student T test. Also one way Analysis of variance (ANOVA) with post hoc Tuckey were used to clarify between groups differences in probe test. The Group receiving Aβ, showed significant cognition deficit (long latency to platform and short total time spent in target quadrant (TTS), parallel with lower level of hippocampal CREB, versus vehicle group. While, Aβ+ GluR23Y exhibited the shortest latency to platform and the longest TTS during the probe test, parallel with the higher hippocampal level of CREB compared with other groups. The present study provides evidence that chronic administration of Tat-GluR23Y; an inhibitor of GluA2-AMPARs endocytosis, successfully restores spatial memory impaired by amyloid beta neurotoxicity targeting CREB signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Ashourpour
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, 8th Km of Rasht -Tehran road, Guilan University Complex, Rasht, Guilan, 41996-13769, Iran
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, 8th Km of Rasht -Tehran road, Guilan University Complex, Rasht, Guilan, 41996-13769, Iran.
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
16
|
Liu S, Liu C, Xiong L, Xie J, Huang C, Pi R, Huang Z, Li L. Icaritin Alleviates Glutamate-Induced Neuronal Damage by Inactivating GluN2B-Containing NMDARs Through the ERK/DAPK1 Pathway. Front Neurosci 2021; 15:525615. [PMID: 33692666 PMCID: PMC7937872 DOI: 10.3389/fnins.2021.525615] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/02/2021] [Indexed: 01/29/2023] Open
Abstract
Excitatory toxicity due to excessive glutamate release is considered the core pathophysiological mechanism of cerebral ischemia. It is primarily mediated by N-methyl-D-aspartate receptors (NMDARs) on neuronal membranes. Our previous studies have found that icaritin (ICT) exhibits neuroprotective effects against cerebral ischemia in rats, but the underlying mechanism is unclear. This study aims to investigate the protective effect of ICT on glutamate-induced neuronal injury and uncover its possible molecular mechanism. An excitatory toxicity injury model was created using rat primary cortical neurons treated with glutamate and glycine. The results showed that ICT has neuroprotective effects on glutamate-treated primary cortical neurons by increasing cell viability while reducing the rate of lactate dehydrogenase (LDH) release and reducing apoptosis. Remarkably, ICT rescued the changes in the ERK/DAPK1 signaling pathway after glutamate treatment by increasing the expression levels of p-ERK, p-DAPK1 and t-DAPK1. In addition, ICT also regulates NMDAR function during glutamate-induced injury by decreasing the expression level of the GluN2B subunit and enhancing the expression level of the GluN2A subunit. As cotreatment with the ERK-specific inhibitor U0126 and ICT abolishes the beneficial effects of ITC on the ERK/DAPK1 pathway, NMDAR subtypes and neuronal cell survival, ERK is recognized as a crucial mediator in the protective mechanism of ICT. In conclusion, our findings demonstrate that ICT has a neuroprotective effect on neuronal damage induced by glutamate, and its mechanism may be related to inactivating GluN2B-containing NMDAR through the ERK/DAPK1 pathway. This study provides a new clue for the prevention and treatment of clinical ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Song Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Chaoming Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Lijiao Xiong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China.,First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jiali Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Cheng Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China.,Institute for Medical Sciences of Pain, Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Rongbiao Pi
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China.,Institute for Medical Sciences of Pain, Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Liangdong Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China.,First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
17
|
Samer S, Raman R, Laube G, Kreutz MR, Karpova A. The nuclear lamina is a hub for the nuclear function of Jacob. Mol Brain 2021; 14:9. [PMID: 33436037 PMCID: PMC7802242 DOI: 10.1186/s13041-020-00722-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Jacob is a synapto-nuclear messenger protein that couples NMDAR activity to CREB-dependent gene expression. In this study, we investigated the nuclear distribution of Jacob and report a prominent targeting to the nuclear envelope that requires NMDAR activity and nuclear import. Immunogold electron microscopy and proximity ligation assay combined with STED imaging revealed preferential association of Jacob with the inner nuclear membrane where it directly binds to LaminB1, an intermediate filament and core component of the inner nuclear membrane (INM). The association with the INM is transient; it involves a functional nuclear export signal in Jacob and a canonical CRM1-RanGTP-dependent export mechanism that defines the residing time of the protein at the INM. Taken together, the data suggest a stepwise redistribution of Jacob within the nucleus following nuclear import and prior to nuclear export.
Collapse
Affiliation(s)
- Sebastian Samer
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Rajeev Raman
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Gregor Laube
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39106, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Anna Karpova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39106, Magdeburg, Germany.
| |
Collapse
|
18
|
Communication of Glioma cells with neuronal plasticity: What is the underlying mechanism? Neurochem Int 2020; 141:104879. [PMID: 33068685 DOI: 10.1016/j.neuint.2020.104879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/26/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022]
Abstract
There has been a significantly rising discussion on how neuronal plasticity communicates with the glioma growth and invasion. This literature review aims to determine which neurotransmitters, ion channels and signaling pathways are involved in this context, how information is transferred from synaptic sites to the glioma cells and how glioma cells apply established mechanics of synaptic plasticity for their own increment. This work is a compilation of some outstanding findings related to the influence of the glutamate, calcium, potassium, chloride and sodium channels and other important brain plasticity molecules over the glioma progression. These topics also include the relevant molecular signaling data which could prove to be helpful for an effective clinical management of brain tumors in the future.
Collapse
|
19
|
Borodinova AA, Balaban PM. Epigenetic Regulation as a Basis for Long-Term Changes in the Nervous System: In Search of Specificity Mechanisms. BIOCHEMISTRY (MOSCOW) 2020; 85:994-966. [DOI: 10.1134/s0006297920090023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Adaptive long-term changes in the functioning of nervous system (plasticity, memory) are not written in the genome, but are directly associated with the changes in expression of many genes comprising epigenetic regulation. Summarizing the known data regarding the role of epigenetics in regulation of plasticity and memory, we would like to highlight several key aspects. (i) Different chromatin remodeling complexes and DNA methyltransferases can be organized into high-order multiprotein repressor complexes that are cooperatively acting as the “molecular brake pads”, selectively restricting transcriptional activity of specific genes at rest. (ii) Relevant physiological stimuli induce a cascade of biochemical events in the activated neurons resulting in translocation of different signaling molecules (protein kinases, NO-containing complexes) to the nucleus. (iii) Stimulus-specific nitrosylation and phosphorylation of different epigenetic factors is linked to a decrease in their enzymatic activity or changes in intracellular localization that results in temporary destabilization of the repressor complexes. (iv) Removing “molecular brakes” opens a “critical time window” for global and local epigenetic changes, triggering specific transcriptional programs and modulation of synaptic connections efficiency. It can be assumed that the reversible post-translational histone modifications serve as the basis of plastic changes in the neural network. On the other hand, DNA methylation and methylation-dependent 3D chromatin organization can serve a stable molecular basis for long-term maintenance of plastic changes and memory.
Collapse
|
20
|
Parra-Damas A, Saura CA. Synapse-to-Nucleus Signaling in Neurodegenerative and Neuropsychiatric Disorders. Biol Psychiatry 2019; 86:87-96. [PMID: 30846302 DOI: 10.1016/j.biopsych.2019.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/18/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023]
Abstract
Synapse-to-nucleus signaling is critical for converting signals received at synapses into transcriptional programs essential for cognition, memory, and emotion. This neuronal mechanism usually involves activity-dependent translocation of synaptonuclear factors from synapses to the nucleus resulting in regulation of transcriptional programs underlying synaptic plasticity. Acting as synapse-to-nucleus messengers, amyloid precursor protein intracellular domain associated-1 protein, cAMP response element binding protein (CREB)-regulated transcription coactivator-1, Jacob, nuclear factor kappa-light-chain-enhancer of activated B cells, RING finger protein 10, and SH3 and multiple ankyrin repeat domains 3 play essential roles in synapse remodeling and plasticity, which are considered the cellular basis of memory. Other synaptic proteins, such as extracellular signal-regulated kinase, calcium/calmodulin-dependent protein kinase II gamma, and CREB2, translocate from dendrites or cytosol to the nucleus upon synaptic activity, suggesting that they could contribute to synapse-to-nucleus signaling. Notably, some synaptonuclear factors converge on the transcription factor CREB, indicating that CREB signaling is a key hub mediating integration of synaptic signals into transcriptional programs required for neuronal function and plasticity. Although major efforts have been focused on identification and regulatory mechanisms of synaptonuclear factors, the relevance of synapse-to-nucleus communication in brain physiology and pathology is still unclear. Recent evidence, however, indicates that synaptonuclear factors are implicated in neuropsychiatric, neurodevelopmental, and neurodegenerative disorders, suggesting that uncoupling synaptic activity from nuclear signaling may prompt synapse pathology, contributing to a broad spectrum of brain disorders. This review summarizes current knowledge of synapse-to-nucleus signaling in neuron survival, synaptic function and plasticity, and memory. Finally, we discuss how altered synapse-to-nucleus signaling may lead to memory and emotional disturbances, which is relevant for clinical and therapeutic strategies in neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
21
|
Schicknick H, Henschke JU, Budinger E, Ohl FW, Gundelfinger ED, Tischmeyer W. β-adrenergic modulation of discrimination learning and memory in the auditory cortex. Eur J Neurosci 2019; 50:3141-3163. [PMID: 31162753 PMCID: PMC6900137 DOI: 10.1111/ejn.14480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 05/27/2019] [Accepted: 05/31/2019] [Indexed: 01/11/2023]
Abstract
Despite vast literature on catecholaminergic neuromodulation of auditory cortex functioning in general, knowledge about its role for long‐term memory formation is scarce. Our previous pharmacological studies on cortex‐dependent frequency‐modulated tone‐sweep discrimination learning of Mongolian gerbils showed that auditory‐cortical D1/5‐dopamine receptor activity facilitates memory consolidation and anterograde memory formation. Considering overlapping functions of D1/5‐dopamine receptors and β‐adrenoceptors, we hypothesised a role of β‐adrenergic signalling in the auditory cortex for sweep discrimination learning and memory. Supporting this hypothesis, the β1/2‐adrenoceptor antagonist propranolol bilaterally applied to the gerbil auditory cortex after task acquisition prevented the discrimination increment that was normally monitored 1 day later. The increment in the total number of hurdle crossings performed in response to the sweeps per se was normal. Propranolol infusion after the seventh training session suppressed the previously established sweep discrimination. The suppressive effect required antagonist injection in a narrow post‐session time window. When applied to the auditory cortex 1 day before initial conditioning, β1‐adrenoceptor‐antagonising and β1‐adrenoceptor‐stimulating agents retarded and facilitated, respectively, sweep discrimination learning, whereas β2‐selective drugs were ineffective. In contrast, single‐sweep detection learning was normal after propranolol infusion. By immunohistochemistry, β1‐ and β2‐adrenoceptors were identified on the neuropil and somata of pyramidal and non‐pyramidal neurons of the gerbil auditory cortex. The present findings suggest that β‐adrenergic signalling in the auditory cortex has task‐related importance for discrimination learning of complex sounds: as previously shown for D1/5‐dopamine receptor signalling, β‐adrenoceptor activity supports long‐term memory consolidation and reconsolidation; additionally, tonic input through β1‐adrenoceptors may control mechanisms permissive for memory acquisition.
Collapse
Affiliation(s)
- Horst Schicknick
- Special Lab Molecular Biological Techniques, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Julia U Henschke
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Eike Budinger
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Frank W Ohl
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Eckart D Gundelfinger
- Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Molecular Neurobiology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Wolfgang Tischmeyer
- Special Lab Molecular Biological Techniques, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
22
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
23
|
Wang X, Zheng W. Ca 2+ homeostasis dysregulation in Alzheimer's disease: a focus on plasma membrane and cell organelles. FASEB J 2019; 33:6697-6712. [PMID: 30848934 DOI: 10.1096/fj.201801751r] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Emerging evidence indicates that Ca2+ is a vital factor in modulating the pathogenesis of Alzheimer's disease (AD). In healthy neurons, Ca2+ concentration is balanced to maintain a lower level in the cytosol than in the extracellular space or certain intracellular compartments such as endoplasmic reticulum (ER) and the lysosome, whereas this homeostasis is broken in AD. On the plasma membrane, the AD hallmarks amyloid-β (Aβ) and tau interact with ligand-gated or voltage-gated Ca2+-influx channels and inhibit the Ca2+-efflux ATPase or exchangers, leading to an elevated intracellular Ca2+ level and disrupted Ca2+ signal. In the ER, the disabled presenilin "Ca2+ leak" function and the direct implications of Aβ and presenilin mutants contribute to Ca2+-signal disorder. The enhanced ryanodine receptor (RyR)-mediated and inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca2+ release from the ER aggravates cytosolic Ca2+ disorder and triggers apoptosis; the down-regulated ER Ca2+ sensor, stromal interaction molecule (STIM), alleviates store-operated Ca2+ entry in plasma membrane, leading to spine loss. The increased transfer of Ca2+ from ER to mitochondria through mitochondria-associated ER membrane (MAM) causes Ca2+ overload in the mitochondrial matrix and consequently opens the cellular damage-related channel, mitochondrial permeability transition pore (mPTP). In this review, we discuss the effects of Aβ, tau and presenilin on neuronal Ca2+ signal, focusing on the receptors and regulators in plasma membrane and ER; we briefly introduce the involvement of MAM-mediated Ca2+ transfer and mPTP opening in AD pathogenesis.-Wang, X., Zheng, W. Ca2+ homeostasis dysregulation in Alzheimer's disease: a focus on plasma membrane and cell organelles.
Collapse
Affiliation(s)
- Xingjian Wang
- Department of Histology and Embryology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Zheng
- Department of Histology and Embryology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
24
|
Sun Y, Xu Y, Cheng X, Chen X, Xie Y, Zhang L, Wang L, Hu J, Gao Z. The differences between GluN2A and GluN2B signaling in the brain. J Neurosci Res 2018; 96:1430-1443. [PMID: 29682799 DOI: 10.1002/jnr.24251] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 03/28/2018] [Accepted: 04/06/2018] [Indexed: 12/24/2022]
Abstract
The N-methyl-d-aspartate (NMDA) receptor, a typical ionotropic glutamate receptor, is a crucial protein for maintaining brain function. GluN2A and GluN2B are the main types of NMDA receptor subunit in the adult forebrain. Studies have demonstrated that they play different roles in a number of pathophysiological processes. Although the underlying mechanism for this has not been clarified, the most fundamental reason may be the differences between the signaling pathways associated with GluN2A and GluN2B. With the aim of elucidating the reasons behind the diverse roles of these two subunits, we described the signaling differences between GluN2A and GluN2B from the aspects of C-terminus-associated molecules, effects on typical downstream signaling proteins, and metabotropic signaling. Because there are several factors interfering with the determination of subunit-specific signaling, there is still a long way to go toward clarifying the signaling differences between these two subunits. Developing better pharmacology tools, such as highly selective antagonists for triheteromeric GluN2A- and GluN2B-containing NMDA receptors, and establishing new molecular biological methods, for example, engineering photoswitchable NMDA receptors, may be useful for clarifying the signaling differences between GluN2A and GluN2B.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China.,Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China
| | - Yingge Xu
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China
| | - Xiaokun Cheng
- Department of Physical and Chemical Analysis, North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, People's Republic of China
| | - Xi Chen
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China
| | - Yinghua Xie
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China.,Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, College of Basic Medical Science, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Long Wang
- Department of Family and Consumer Sciences, California State University, Long Beach, California
| | - Jie Hu
- Nursing Research Center, School of Nursing, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China.,Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China.,State Key Laboratory Breeding Base, Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, People's Republic of China
| |
Collapse
|
25
|
Lisman J, Cooper K, Sehgal M, Silva AJ. Memory formation depends on both synapse-specific modifications of synaptic strength and cell-specific increases in excitability. Nat Neurosci 2018; 21:309-314. [PMID: 29434376 PMCID: PMC5915620 DOI: 10.1038/s41593-018-0076-6] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/08/2017] [Indexed: 11/09/2022]
Abstract
The modification of synaptic strength produced by long-term potentiation (LTP) is widely thought to underlie memory storage. Indeed, given that hippocampal pyramidal neurons have >10,000 independently modifiable synapses, the potential for information storage by synaptic modification is enormous. However, recent work suggests that CREB-mediated global changes in neuronal excitability also play a critical role in memory formation. Because these global changes have a modest capacity for information storage compared with that of synaptic plasticity, their importance for memory function has been unclear. Here we review the newly emerging evidence for CREB-dependent control of excitability and discuss two possible mechanisms. First, the CREB-dependent transient change in neuronal excitability performs a memory-allocation function ensuring that memory is stored in ways that facilitate effective linking of events with temporal proximity (hours). Second, these changes may promote cell-assembly formation during the memory-consolidation phase. It has been unclear whether such global excitability changes and local synaptic mechanisms are complementary. Here we argue that the two mechanisms can work together to promote useful memory function.
Collapse
Affiliation(s)
- John Lisman
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | | - Megha Sehgal
- Department of Neurobiology, Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, Integrative Center for Learning and Memory, and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alcino J Silva
- Department of Biology, Brandeis University, Waltham, MA, USA.
- Department of Neurobiology, Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, Integrative Center for Learning and Memory, and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Chen SP, Sun J, Zhou YQ, Cao F, Braun C, Luo F, Ye DW, Tian YK. Sinomenine attenuates cancer-induced bone pain via suppressing microglial JAK2/STAT3 and neuronal CAMKII/CREB cascades in rat models. Mol Pain 2018; 14:1744806918793232. [PMID: 30027795 PMCID: PMC6096675 DOI: 10.1177/1744806918793232] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/04/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer-induced bone pain is one of the most severe types of pathological pain, which often occurs in patients with advanced prostate, breast, and lung cancer. It is of great significance to improve the therapies of cancer-induced bone pain due to the opioids' side effects including addiction, sedation, pruritus, and vomiting. Sinomenine, a traditional Chinese medicine, showed obvious analgesic effects on a rat model of chronic inflammatory pain, but has never been proven to treat cancer-induced bone pain. In the present study, we investigated the analgesic effect of sinomenine after tumor cell implantation and specific cellular mechanisms in cancer-induced bone pain. Our results indicated that single administration of sinomenine significantly and dose-dependently alleviated mechanical allodynia in rats with cancer-induced bone pain and the effect lasted for 4 h. After tumor cell implantation, the protein levels of phosphorylated-Janus family tyrosine kinase 2 (p-JAK2), phosphorylated-signal transducers and activators of transcription 3 (p-STAT3), phosphorylated-Ca2+/calmodulin-dependent protein kinase II (p-CAMKII), and phosphorylated-cyclic adenosine monophosphate response element-binding protein (p-CREB) were persistently up-regulated in the spinal cord horn. Chronic intraperitoneal treatment with sinomenine markedly suppressed the activation of microglia and effectively inhibited the expression of JAK2/STAT3 and CAMKII/CREB signaling pathways. We are the first to reveal that up-regulation of microglial JAK2/STAT3 pathway are involved in the development and maintenance of cancer-induced bone pain. Moreover, our investigation provides the first evidence that sinomenine alleviates cancer-induced bone pain by inhibiting microglial JAK2/STAT3 and neuronal CAMKII/CREB cascades.
Collapse
Affiliation(s)
- Shu-Ping Chen
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Sun
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Cao
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Psychiatry, UMKC School of Medicine, Kansas City, MO
| | - Cody Braun
- UMKC School of Medicine, Kansas City, MO
| | - Fang Luo
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ke Tian
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Welters A, Klüppel C, Mrugala J, Wörmeyer L, Meissner T, Mayatepek E, Heiss C, Eberhard D, Lammert E. NMDAR antagonists for the treatment of diabetes mellitus-Current status and future directions. Diabetes Obes Metab 2017; 19 Suppl 1:95-106. [PMID: 28880473 DOI: 10.1111/dom.13017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/18/2017] [Accepted: 05/20/2017] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus is characterized by chronically elevated blood glucose levels accelerated by a progressive decline of insulin-producing β-cells in the pancreatic islets. Although medications are available to transiently adjust blood glucose to normal levels, the effects of current drugs are limited when it comes to preservation of a critical mass of functional β-cells to sustainably maintain normoglycemia. In this review, we recapitulate recent evidence on the role of pancreatic N-methyl-D-aspartate receptors (NMDARs) in β-cell physiology, and summarize effects of morphinan-based NMDAR antagonists that are beneficial for insulin secretion, glucose tolerance and islet cell survival. We further discuss NMDAR-mediated molecular pathways relevant for neuronal cell survival, which may also be important for the preservation of β-cell function and mass. Finally, we summarize the literature for evidence on the role of NMDARs in the development of diabetic long-term complications, and highlight beneficial pharmacologic aspects of NMDAR antagonists in diabetic nephropathy, retinopathy as well as neuropathy.
Collapse
Affiliation(s)
- Alena Welters
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Carina Klüppel
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Jessica Mrugala
- Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Helmholtz Zentrum München, Neuherberg, Düsseldorf, Germany
| | - Laura Wörmeyer
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Helmholtz Zentrum München, Neuherberg, Düsseldorf, Germany
| |
Collapse
|
28
|
Schaefer N, Rotermund C, Blumrich EM, Lourenco MV, Joshi P, Hegemann RU, Jamwal S, Ali N, García Romero EM, Sharma S, Ghosh S, Sinha JK, Loke H, Jain V, Lepeta K, Salamian A, Sharma M, Golpich M, Nawrotek K, Paidi RK, Shahidzadeh SM, Piermartiri T, Amini E, Pastor V, Wilson Y, Adeniyi PA, Datusalia AK, Vafadari B, Saini V, Suárez-Pozos E, Kushwah N, Fontanet P, Turner AJ. The malleable brain: plasticity of neural circuits and behavior - a review from students to students. J Neurochem 2017. [PMID: 28632905 DOI: 10.1111/jnc.14107] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One of the most intriguing features of the brain is its ability to be malleable, allowing it to adapt continually to changes in the environment. Specific neuronal activity patterns drive long-lasting increases or decreases in the strength of synaptic connections, referred to as long-term potentiation and long-term depression, respectively. Such phenomena have been described in a variety of model organisms, which are used to study molecular, structural, and functional aspects of synaptic plasticity. This review originated from the first International Society for Neurochemistry (ISN) and Journal of Neurochemistry (JNC) Flagship School held in Alpbach, Austria (Sep 2016), and will use its curriculum and discussions as a framework to review some of the current knowledge in the field of synaptic plasticity. First, we describe the role of plasticity during development and the persistent changes of neural circuitry occurring when sensory input is altered during critical developmental stages. We then outline the signaling cascades resulting in the synthesis of new plasticity-related proteins, which ultimately enable sustained changes in synaptic strength. Going beyond the traditional understanding of synaptic plasticity conceptualized by long-term potentiation and long-term depression, we discuss system-wide modifications and recently unveiled homeostatic mechanisms, such as synaptic scaling. Finally, we describe the neural circuits and synaptic plasticity mechanisms driving associative memory and motor learning. Evidence summarized in this review provides a current view of synaptic plasticity in its various forms, offers new insights into the underlying mechanisms and behavioral relevance, and provides directions for future research in the field of synaptic plasticity. Read the Editorial Highlight for this article on page 788. Cover Image for this issue: doi: 10.1111/jnc.13815.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Würzburg, Germany
| | - Carola Rotermund
- German Center of Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany.,Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pooja Joshi
- Inserm UMR 1141, Robert Debre Hospital, Paris, France
| | - Regina U Hegemann
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Sumit Jamwal
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nilufar Ali
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | | | - Sorabh Sharma
- Neuropharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Shampa Ghosh
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Jitendra K Sinha
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Hannah Loke
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Vishal Jain
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmad Salamian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mahima Sharma
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mojtaba Golpich
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Katarzyna Nawrotek
- Department of Process Thermodynamics, Faculty of Process and Environmental Engineering, Lodz University of Technology, Lodz, Poland
| | - Ramesh K Paidi
- CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sheila M Shahidzadeh
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Tetsade Piermartiri
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elham Amini
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Veronica Pastor
- Instituto de Biología Celular y Neurociencia Prof. Eduardo De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Yvette Wilson
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Philip A Adeniyi
- Cell Biology and Neurotoxicity Unit, Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado - Ekiti, Ekiti State, Nigeria
| | | | - Benham Vafadari
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Vedangana Saini
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Edna Suárez-Pozos
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Toxicología, México
| | - Neetu Kushwah
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Paula Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cellular Biology and Neuroscience (IBCN), CONICET-UBA, School of Medicine, Buenos Aires, Argentina
| | - Anthony J Turner
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
29
|
Wei C, Luo T, Zou S, Zhou X, Shen W, Ji X, Li Q, Wu A. Differentially expressed lncRNAs and miRNAs with associated ceRNA networks in aged mice with postoperative cognitive dysfunction. Oncotarget 2017; 8:55901-55914. [PMID: 28915561 PMCID: PMC5593532 DOI: 10.18632/oncotarget.18362] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/28/2017] [Indexed: 12/11/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative complication observed in elderly patients. Using microarray analyses, we comprehensively compared long non-coding RNA (lncRNA), messenger RNA (mRNA), and microRNA (miRNA) expression profiles in hippocampal tissues from a mouse model of POCD and control mice. A total of 175 lncRNAs, 117 mRNAs, and 26 miRNAs were differentially expressed between POCD and control mice. Gene ontology (GO) and KEGG pathway enrichment analyses were performed to explore the principal functions of dysregulated genes. Correlated coding-noncoding co-expression (CNC) and competing endogenous RNA (ceRNA) expression networks were constructed using bioinformatics methods. lncRNA NONMMUT000708 correlated positively with expression of the inflammation-related gene Hif3a. lncRNAs NONMMUT043249 and NONMMUT028705 mediated gene expression by binding the transcription factor cAMP response element-binding protein (CREB). The constructed ceRNA network suggested lncRNA NONMMUT055714 binds competitively with miR-7684-5p, increasing expression of its target gene, Sorl1. Finally, eight dysregulated lncRNAs, four miRNAs, and ten mRNAs were confirmed via quantitative real-time polymerase chain reaction (PCR) in 10 POCD-healthy mouse paired samples. These results suggest that lncRNAs and miRNAs are involved in POCD pathogenesis and progression. Our ceRNA network will improve understanding of lncRNA-mediated ceRNA regulatory mechanisms operating during the pathogenesis of POCD.
Collapse
Affiliation(s)
- Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ting Luo
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shanshan Zou
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaobin Zhou
- Department of Orthopedics, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wenzhen Shen
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaolin Ji
- Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Qi Li
- Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|