1
|
de Andrés R, Martínez-Blanco E, Díez-Guerra FJ. HDAC4 Inhibits NMDA Receptor-mediated Stimulation of Neurogranin Expression. Mol Neurobiol 2025; 62:5609-5628. [PMID: 39581920 DOI: 10.1007/s12035-024-04598-3] [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: 07/09/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024]
Abstract
The coordination of neuronal wiring and activity within the central nervous system (CNS) is crucial for cognitive function, particularly in the context of aging and neurological disorders. Neurogranin (Ng), an abundant forebrain protein, modulates calmodulin (CaM) activity and deeply influences synaptic plasticity and neuronal processing. This study investigates the regulatory mechanisms of Ng expression, a critical but underexplored area for combating cognitive impairment. Utilizing both in vitro and in vivo hippocampal models, we show that Ng expression arises during late developmental stages, coinciding with the processes of synaptic maturation and neuronal circuit consolidation. We observed that Ng expression increases in neuronal networks with heightened synaptic activity and identified GluN2B-containing N-methyl-D-aspartate (NMDA) receptors as key drivers of this expression. Additionally, we discovered that nuclear-localized HDAC4 inhibits Ng expression, establishing a regulatory axis that is counteracted by NMDA receptor stimulation. Analysis of the Ng gene promoter activity revealed regulatory elements between the - 2.4 and - 0.85 Kbp region, including a binding site for RE1-Silencing Transcription factor (REST), which may mediate HDAC4's repressive effect on Ng expression. Further analysis of the promoter sequence revealed conserved binding sites for the myocyte enhancer factor-2 (MEF2) transcription factor, a target of HDAC4-mediated transcription regulation. Our findings elucidate the interplay between synaptic activity, NMDAR function, and transcriptional regulation in controlling Ng expression, offering insights into synaptic plasticity mechanisms and potential therapeutic strategies to prevent cognitive dysfunction.
Collapse
Affiliation(s)
- Raquel de Andrés
- Laboratory Molecular Basis of Neuronal Plasticity, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Elena Martínez-Blanco
- Laboratory Molecular Basis of Neuronal Plasticity, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - F Javier Díez-Guerra
- Laboratory Molecular Basis of Neuronal Plasticity, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain.
| |
Collapse
|
2
|
Aderibigbe O, Wood LB, Margulies SS. Cyclosporine A Accelerates Neurorecovery Transcriptional Trajectory in a Swine Model of Diffuse Traumatic Brain Injury. Int J Mol Sci 2025; 26:3531. [PMID: 40331981 PMCID: PMC12026708 DOI: 10.3390/ijms26083531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025] Open
Abstract
Mild traumatic brain injury (mTBI) is a leading cause of morbidity in children with both short- and long-term neurological, cognitive, cerebrovascular, and emotional deficits. These deficits have been attributed to ongoing pathophysiological cascades that occur acutely and persist post-injury. Given our limited understanding of the transcriptional changes associated with these pathophysiological cascades, we studied formalin-fixed paraffin-embedded (FFPE) tissues from the frontal cortex (FC) and the hippocampus + amygdala (H&A) regions of swine (N = 40) after a sagittal rapid non-impact head rotation (RNR). We then sequenced RNA to define transcriptional changes at 1 day and 1 week after injury and investigated the protective influence of cyclosporine A (CsA) treatment. Differentially expressed genes (DEGs) were classified into five temporal patterns (Early, Transient, Persistent, Intensified, Delayed, or Late). DEGs were more abundant at 1 week than 1 day. Shared significant gene ontology annotations in both regions included terms associated with neuronal distress at 1 day and neurorecovery at 1 week. CsA (20 mg/kg/day) infused for 1 day (beginning at 6 h after injury) accelerated 466 DEGs in the FC and 2794 DEGs in the H&A, such that the CsA-treated transcriptional profile was associated with neurorecovery. Overall, our data reveal the effects of anatomic region and elapsed time on gene expression post-mTBI and motivate future studies of CsA treatment.
Collapse
Affiliation(s)
- Oluwagbemisola Aderibigbe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| | - Levi B. Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan S. Margulies
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| |
Collapse
|
3
|
Brown JC, Kweon J, Sharma P, Siddiqi SH, Isserles M, Ressler KJ. Critically Assessing the Unanswered Questions of How, Where, and When to Induce Plasticity in the Posttraumatic Stress Disorder Network With Transcranial Magnetic Stimulation. Biol Psychiatry 2025; 97:392-404. [PMID: 38909668 DOI: 10.1016/j.biopsych.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024]
Abstract
Extinction of traumatic memory, a primary treatment approach (termed exposure therapy) in posttraumatic stress disorder (PTSD), occurs through relearning and may be subserved at the molecular level by long-term potentiation of relevant circuits. In parallel, repetitive transcranial magnetic stimulation (TMS) is thought to work through long-term potentiation-like mechanisms and may provide a novel, safe, and effective treatment for PTSD. In a recent failed randomized controlled trial we emphasized the necessity of correctly identifying cortical targets, the directionality of TMS protocols, and the role of memory activation. Here, we provide a systematic review of TMS for PTSD to further identify how, where, and when TMS treatment should be delivered to alleviate PTSD symptoms. We conducted a systematic review of the literature by searching for repetitive TMS clinical trials involving patients with PTSD and outcomes. We searched MEDLINE through October 25, 2023, for "TMS and PTSD" and "transcranial magnetic stimulation and posttraumatic stress disorder." Thirty-one publications met our inclusion criteria (k = 17 randomized controlled trials, k = 14 open label). Randomized controlled trial protocols were varied in terms of TMS protocols, cortical TMS targets, and memory activation protocols. There was no clear superiority of low-frequency (k = 5) versus high-frequency (k = 6) protocols or by stimulation location. Memory provocation or exposure protocols (k = 7) appear to enhance response. Overall, TMS appears to be effective in treating PTSD symptoms across a variety of TMS frequencies, hemispheric target differences, and exposure protocols. Disparate protocols may be conceptually harmonized when viewed as potentiating proposed anxiolytic networks or suppressing anxiogenic networks.
Collapse
Affiliation(s)
- Joshua C Brown
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| | - Jamie Kweon
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts
| | - Prayushi Sharma
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts
| | - Shan H Siddiqi
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, Boston, Massachusetts
| | - Moshe Isserles
- Department of Psychiatry, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
4
|
Svirsky SE, Henchir J, Parry M, Holets E, Zhang T, Gittes GK, Carlson SW, Dixon CE. Viral-mediated increased hippocampal neurogranin modulate synapses at one month in a rat model of controlled cortical impact. Sci Rep 2024; 14:28998. [PMID: 39578516 PMCID: PMC11584851 DOI: 10.1038/s41598-024-77682-2] [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: 02/27/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024] Open
Abstract
Reductions of neurogranin (Ng), a calcium-sensitive calmodulin-binding protein, result in significant impairment across various hippocampal-dependent learning and memory tasks. Conversely, increasing levels of Ng facilitates synaptic plasticity, increases synaptogenesis and boosts cognitive abilities. Controlled cortical impact (CCI), an experimental traumatic brain injury (TBI) model, results in significantly reduced hippocampal Ng protein expression up to 4 weeks post-injury, supporting a strategy to increase Ng to improve function. In this study, hippocampal Ng expression was increased in adult, male Sham and CCI injured animals using intraparenchymal injection of adeno-associated virus (AAV) 30 min post-injury, thereby also affording the ability to differentiate endogenous and exogenous Ng. At 4 weeks, molecular, anatomical, and behavioral measures of synaptic plasticity were evaluated to determine the therapeutic potential of Ng modulation post-TBI. Increasing Ng had a TBI-dependent effect on hippocampal expression of synaptic proteins and dendritic spine morphology. Increasing Ng did not improve behavior across all outcomes in both Sham and CCI groups at the 4 week time-point. Overall, increasing Ng expression modulated protein expression and dendritic spine morphology, but exerted limited functional benefit after CCI. This study furthers our understanding of Ng, and mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Madison Parry
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Erik Holets
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Ting Zhang
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - George K Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- V.A. Pittsburgh Healthcare System, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
5
|
Yu X, Guo J, Song Y, Wei B, Shi Y, Zhao Y, Zhao Z, Gao Q, Wang B, Sun M. HDAC1/2/3-mediated downregulation of neurogranin is involved in cognitive impairment in offspring exposed to maternal subclinical hypothyroidism. FASEB J 2024; 38:e23736. [PMID: 38865202 DOI: 10.1096/fj.202400389r] [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: 02/20/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Subclinical hypothyroidism (SCH) in pregnancy is the most common form of thyroid dysfunction in pregnancy, which can affect fetal nervous system development and increase the risk of neurodevelopmental disorders after birth. However, the mechanism of the effect of maternal subclinical hypothyroidism on fetal brain development and behavioral phenotypes is still unclear and requires further study. In this study, we constructed a mouse model of maternal subclinical hypothyroidism by exposing dams to drinking water containing 50 ppm propylthiouracil (PTU) during pregnancy and found that its offspring were accompanied by severe cognitive deficits by behavioral testing. Mechanistically, gestational SCH resulted in the upregulation of protein expression and activity of HDAC1/2/3 in the hippocampus of the offspring. ChIP analysis revealed that H3K9ac on the neurogranin (Ng) promoter was reduced in the hippocampus of the offspring of SCH, with a significant reduction in Ng protein, leading to reduced expression levels of synaptic plasticity markers PSD95 (a membrane-associated protein in the postsynaptic density) and SYN (synaptophysin, a specific marker for presynaptic terminals), and impaired synaptic plasticity. In addition, administration of MS-275 (an HDAC1/2/3-specific inhibitor) to SCH offspring alleviated impaired synaptic plasticity and cognitive dysfunction in offspring. Thus, our study suggests that maternal subclinical hypothyroidism may mediate offspring cognitive dysfunction through the HDAC1/2/3-H3K9ac-Ng pathway. Our study contributes to the understanding of the signaling mechanisms underlying maternal subclinical hypothyroidism-mediated cognitive impairment in the offspring.
Collapse
Affiliation(s)
- Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Guo
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yueyang Song
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zejun Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic, Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
6
|
Rueda-García V, Rondón-Barragán IS. Molecular Characterization of Neurogranin (NRGN) Gene from Red‑Bellied Pacu (Piaractus brachypomus). Mol Neurobiol 2024; 61:2620-2630. [PMID: 37922064 PMCID: PMC11043121 DOI: 10.1007/s12035-023-03700-5] [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: 06/15/2023] [Accepted: 10/06/2023] [Indexed: 11/05/2023]
Abstract
Neurogranin (NRGN) is a small brain protein expressed in various telencephalic areas and plays an essential role in synaptic plasticity by regulating the availability of calmodulin (CaM). The study aims to characterize the neurogranin gene in Colombian native fish, red-bellied pacu, Piaractus brachypomus, its basal tissue expression and differential expression in brain injury and sublethal toxicity by organophosphates. NRGN gene contains an open reading frame of 183 nucleotides encoding for 60 amino acids. Bioinformatics analysis showed an IQ motif necessary in the interaction with CaM. NRGN mRNA was detected in tissues with higher expression in brain, gills, and head kidney. In brain regions, NRGN showed high expression in the telencephalon (TE) and olfactory bulb (OB). In the sublethal toxicity experiment, NRGN mRNA was upregulated in individuals under organophosphate exposure in the OB and optic chiasm (OC). In brain injury experiment, NRGN showed upregulation at 14 days in OC and at 24 h and 7 days in TE. These findings demonstrate the differential expression of NRGN under different experimental conditions which make it a candidate for a biomarker in the brain of P. brachypomus.
Collapse
Affiliation(s)
- Valentina Rueda-García
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Building 33 L105, 730002, Ibagué, Tolima, Colombia
| | - Iang Schroniltgen Rondón-Barragán
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Building 33 L105, 730002, Ibagué, Tolima, Colombia.
| |
Collapse
|
7
|
Alba‐González A, Folgueira M, Castro A, Anadón R, Yáñez J. Distribution of neurogranin-like immunoreactivity in the brain and sensory organs of the adult zebrafish. J Comp Neurol 2022; 530:1569-1587. [PMID: 35015905 PMCID: PMC9415131 DOI: 10.1002/cne.25297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 11/11/2022]
Abstract
We studied the expression of neurogranin in the brain and some sensory organs (barbel taste buds, olfactory organs, and retina) of adult zebrafish. Database analysis shows zebrafish has two paralog neurogranin genes (nrgna and nrgnb) that translate into three peptides with a conserved IQ domain, as in mammals. Western blots of zebrafish brain extracts using an anti-neurogranin antiserum revealed three separate bands, confirming the presence of three neurogranin peptides. Immunohistochemistry shows neurogranin-like expression in the brain and sensory organs (taste buds, neuromasts and olfactory epithelium), not being able to discern its three different peptides. In the retina, the most conspicuous positive cells were bipolar neurons. In the brain, immunopositive neurons were observed in all major regions (pallium, subpallium, preoptic area, hypothalamus, diencephalon, mesencephalon and rhombencephalon, including the cerebellum), a more extended distribution than in mammals. Interestingly, dendrites, cell bodies and axon terminals of some neurons were immunopositive, thus zebrafish neurogranins may play presynaptic and postsynaptic roles. Most positive neurons were found in primary sensory centers (viscerosensory column and medial octavolateral nucleus) and integrative centers (pallium, subpallium, optic tectum and cerebellum), which have complex synaptic circuitry. However, we also observed expression in areas not related to sensory or integrative functions, such as in cerebrospinal fluid-contacting cells associated with the hypothalamic recesses, which exhibited high neurogranin-like immunoreactivity. Together, these results reveal important differences with the patterns reported in mammals, suggesting divergent evolution from the common ancestor.
Collapse
Affiliation(s)
- Anabel Alba‐González
- Department of Biology, Faculty of SciencesUniversity of A CoruñaA CoruñaSpain,Centro de Investigaciones Científicas Avanzadas (CICA)University of A CoruñaA CoruñaSpain
| | - Mónica Folgueira
- Department of Biology, Faculty of SciencesUniversity of A CoruñaA CoruñaSpain,Centro de Investigaciones Científicas Avanzadas (CICA)University of A CoruñaA CoruñaSpain
| | - Antonio Castro
- Department of Biology, Faculty of SciencesUniversity of A CoruñaA CoruñaSpain,Centro de Investigaciones Científicas Avanzadas (CICA)University of A CoruñaA CoruñaSpain
| | - Ramón Anadón
- Department of Functional Biology, Faculty of BiologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Julián Yáñez
- Department of Biology, Faculty of SciencesUniversity of A CoruñaA CoruñaSpain,Centro de Investigaciones Científicas Avanzadas (CICA)University of A CoruñaA CoruñaSpain
| |
Collapse
|
8
|
Stollenwerk TM, Hillard CJ. Adolescent THC Treatment Does Not Potentiate the Behavioral Effects in Adulthood of Maternal Immune Activation. Cells 2021; 10:3503. [PMID: 34944011 PMCID: PMC8700174 DOI: 10.3390/cells10123503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Both in utero exposure to maternal immune activation and cannabis use during adolescence have been associated with increased risk for the development of schizophrenia; however, whether these exposures exert synergistic effects on brain function is not known. In the present study, mild maternal immune activation (MIA) was elicited in mice with prenatal exposure to polyinosinic-polycytidylic acid (poly(I:C)), and ∆9-tetrahydrocannabinol (THC) was provided throughout adolescence in cereal (3 mg/kg/day for 5 days). Neither THC nor MIA pretreatments altered activity in assays used to characterize hyperdopaminergic states in adulthood: amphetamine hyperlocomotion and prepulse inhibition of the acoustic startle reflex. Adolescent THC treatment elicited deficits in spatial memory and enhanced spatial reversal learning in adult female mice in the Morris water maze, while exposure to MIA elicited female-specific deficits in fear extinction learning in adulthood. There were no effects in these assays in adult males, nor were there interactions between THC and MIA in adult females. While doses of poly(I:C) and THC were sufficient to elicit behavioral effects, particularly relating to cognitive performance in females, there was no evidence that adolescent THC exposure synergized with the risk imposed by MIA to worsen behavioral outcomes in adult mice of either sex.
Collapse
Affiliation(s)
| | - Cecilia J. Hillard
- Neuroscience Research Center, Department of Pharmacology and Toxicology, Medical College of Wisconsin, Wauwatosa, WI 53226, USA;
| |
Collapse
|
9
|
Gribaudo S, Saraulli D, Nato G, Bonzano S, Gambarotta G, Luzzati F, Costanzi M, Peretto P, Bovetti S, De Marchis S. Neurogranin Regulates Adult-Born Olfactory Granule Cell Spine Density and Odor-Reward Associative Memory in Mice. Int J Mol Sci 2021; 22:ijms22084269. [PMID: 33924098 PMCID: PMC8074334 DOI: 10.3390/ijms22084269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/16/2022] Open
Abstract
Neurogranin (Ng) is a brain-specific postsynaptic protein, whose role in modulating Ca2+/calmodulin signaling in glutamatergic neurons has been linked to enhancement in synaptic plasticity and cognitive functions. Accordingly, Ng knock-out (Ng-ko) mice display hippocampal-dependent learning and memory impairments associated with a deficit in long-term potentiation induction. In the adult olfactory bulb (OB), Ng is expressed by a large population of GABAergic granule cells (GCs) that are continuously generated during adult life, undergo high synaptic remodeling in response to the sensory context, and play a key role in odor processing. However, the possible implication of Ng in OB plasticity and function is yet to be investigated. Here, we show that Ng expression in the OB is associated with the mature state of adult-born GCs, where its active-phosphorylated form is concentrated at post-synaptic sites. Constitutive loss of Ng in Ng-ko mice resulted in defective spine density in adult-born GCs, while their survival remained unaltered. Moreover, Ng-ko mice show an impaired odor-reward associative memory coupled with reduced expression of the activity-dependent transcription factor Zif268 in olfactory GCs. Overall, our data support a role for Ng in the molecular mechanisms underlying GC plasticity and the formation of olfactory associative memory.
Collapse
Affiliation(s)
- Simona Gribaudo
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
| | - Daniele Saraulli
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council, 00143 Rome, Italy;
| | - Giulia Nato
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
| | - Giovanna Gambarotta
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
- Department of Clinical and Biological Sciences (DSCB), University of Torino, 10043 Turin, Italy
| | - Federico Luzzati
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
| | - Marco Costanzi
- Department of Human Sciences, LUMSA University, 00193 Rome, Italy;
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
- Correspondence: (S.B.); (S.D.M.)
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.G.); (G.N.); (S.B.); (F.L.); (P.P.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy;
- Correspondence: (S.B.); (S.D.M.)
| |
Collapse
|
10
|
Hwang H, Szucs MJ, Ding LJ, Allen A, Ren X, Haensgen H, Gao F, Rhim H, Andrade A, Pan JQ, Carr SA, Ahmad R, Xu W. Neurogranin, Encoded by the Schizophrenia Risk Gene NRGN, Bidirectionally Modulates Synaptic Plasticity via Calmodulin-Dependent Regulation of the Neuronal Phosphoproteome. Biol Psychiatry 2021; 89:256-269. [PMID: 33032807 PMCID: PMC9258036 DOI: 10.1016/j.biopsych.2020.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Neurogranin (Ng), encoded by the schizophrenia risk gene NRGN, is a calmodulin-binding protein enriched in the postsynaptic compartments, and its expression is reduced in the postmortem brains of patients with schizophrenia. Experience-dependent translation of Ng is critical for encoding contextual memory, and Ng regulates developmental plasticity in the primary visual cortex during the critical period. However, the overall impact of Ng on the neuronal signaling that regulates synaptic plasticity is unknown. METHODS Altered Ng expression was achieved via virus-mediated gene manipulation in mice. The effect on long-term potentiation (LTP) was accessed using spike timing-dependent plasticity protocols. Quantitative phosphoproteomics analyses led to discoveries in significant phosphorylated targets. An identified candidate was examined with high-throughput planar patch clamp and was validated with pharmacological manipulation. RESULTS Ng bidirectionally modulated LTP in the hippocampus. Decreasing Ng levels significantly affected the phosphorylation pattern of postsynaptic density proteins, including glutamate receptors, GTPases, kinases, RNA binding proteins, selective ion channels, and ionic transporters, some of which highlighted clusters of schizophrenia- and autism-related genes. Hypophosphorylation of NMDA receptor subunit Grin2A, one significant phosphorylated target, resulted in accelerated decay of NMDA receptor currents. Blocking protein phosphatase PP2B activity rescued the accelerated NMDA receptor current decay and the impairment of LTP mediated by Ng knockdown, implicating the requirement of synaptic PP2B activity for the deficits. CONCLUSIONS Altered Ng levels affect the phosphorylation landscape of neuronal proteins. PP2B activity is required for mediating the deficit in synaptic plasticity caused by decreasing Ng levels, revealing a novel mechanistic link of a schizophrenia risk gene to cognitive deficits.
Collapse
Affiliation(s)
- Hongik Hwang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| | | | - Lei J. Ding
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrew Allen
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Xiaobai Ren
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Henny Haensgen
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fan Gao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hyewhon Rhim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Jen Q. Pan
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Weifeng Xu
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
11
|
Wanderlind ML, Gonçalves R, Tomasi CD, Dal-Pizzol F, Ritter C. Association of neurogranin with delirium among critically ill patients. Biomark Med 2020; 14:1613-1617. [PMID: 33336596 DOI: 10.2217/bmm-2020-0328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Neurogranin (Ng) concentrates at dendritic spines. In patients with Alzheimer disease Ng levels are elevated. The role of Ng in delirium development has not been assessed, therefore we hypothesized that Ng levels are associated with delirium in critically ill patients. Materials & methods: From 94 critically ill patients, 47 developed delirium and 47 controls were included. Blood was collected during the first 24 h of intensive care unit (ICU) admission, and on the day of delirium diagnoses. Ng and IL-1β were determined. Results: Ng and IL-1β levels were higher in the delirium group at ICU admission and on the day of delirium diagnoses. IL-1β and Ng were independently associated with delirium occurrence. Conclusion: Ng levels are associated with delirium development in ICU patients.
Collapse
Affiliation(s)
- Márcia Lz Wanderlind
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Renata Gonçalves
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Cristiane D Tomasi
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| | - Cristiane Ritter
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| |
Collapse
|
12
|
Xiang Y, Xin J, Le W, Yang Y. Neurogranin: A Potential Biomarker of Neurological and Mental Diseases. Front Aging Neurosci 2020; 12:584743. [PMID: 33132903 PMCID: PMC7573493 DOI: 10.3389/fnagi.2020.584743] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
Neurogranin (Ng) is a small protein usually expressed in granule-like structures in pyramidal cells of the hippocampus and cortex. However, its clinical value is not fully clear so far. Currently, Ng is proved to be involved in synaptic plasticity, synaptic regeneration, and long-term potentiation mediated by the calcium- and calmodulin-signaling pathways. Due to both the synaptic integrity and function as the growing concerns in the pathogenesis of a wide variety of neurological and mental diseases, a series of researches published focused on the associations between Ng and these kinds of diseases in the past decade. Therefore, in this review, we highlight several diseases, which include, but are not limited to, Alzheimer’s disease, Parkinson disease, Creutzfeldt–Jakob disease, neuro-HIV, neurosyphilis, schizophrenia, depression, traumatic brain injury, and acute ischemic stroke, and summarize the associations between cerebrospinal fluid or blood-derived Ng with these diseases. We propose that Ng is a potential and promising biomarker to improve the diagnosis, prognosis, and severity evaluation of these diseases in the future.
Collapse
Affiliation(s)
- Yang Xiang
- Institute of Neuroscience, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Clinical Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| | - Jiayan Xin
- North Sichuan Medical College, Nanchong, China.,Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| | - Weidong Le
- Institute of Neuroscience, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Clinical Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yongjian Yang
- Department of Cardiovasology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
13
|
Niu H, Xie R, Li L, Zhang X, Wei X, Deng J, Li Z. WITHDRAWN: Resveratrol partially prevents learning and memory deficits in rats exposed to gaseous formaldehyde. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020:S1382-6689(20)30179-4. [PMID: 32976996 DOI: 10.1016/j.etap.2020.103503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Huatao Niu
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Ran Xie
- Department of PET-CT, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Lanjiang Li
- College of Forensic Medicine, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Xiang Zhang
- Experimental Demonstration Center, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Xiaohan Wei
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Jie Deng
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Zhongming Li
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China.
| |
Collapse
|
14
|
Kalish BT, Barkat TR, Diel EE, Zhang EJ, Greenberg ME, Hensch TK. Single-nucleus RNA sequencing of mouse auditory cortex reveals critical period triggers and brakes. Proc Natl Acad Sci U S A 2020; 117:11744-11752. [PMID: 32404418 PMCID: PMC7261058 DOI: 10.1073/pnas.1920433117] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Auditory experience drives neural circuit refinement during windows of heightened brain plasticity, but little is known about the genetic regulation of this developmental process. The primary auditory cortex (A1) of mice exhibits a critical period for thalamocortical connectivity between postnatal days P12 and P15, during which tone exposure alters the tonotopic topography of A1. We hypothesized that a coordinated, multicellular transcriptional program governs this window for patterning of the auditory cortex. To generate a robust multicellular map of gene expression, we performed droplet-based, single-nucleus RNA sequencing (snRNA-seq) of A1 across three developmental time points (P10, P15, and P20) spanning the tonotopic critical period. We also tone-reared mice (7 kHz pips) during the 3-d critical period and collected A1 at P15 and P20. We identified and profiled both neuronal (glutamatergic and GABAergic) and nonneuronal (oligodendrocytes, microglia, astrocytes, and endothelial) cell types. By comparing normal- and tone-reared mice, we found hundreds of genes across cell types showing altered expression as a result of sensory manipulation during the critical period. Functional voltage-sensitive dye imaging confirmed GABA circuit function determines critical period onset, while Nogo receptor signaling is required for its closure. We further uncovered previously unknown effects of developmental tone exposure on trajectories of gene expression in interneurons, as well as candidate genes that might execute tonotopic plasticity. Our single-nucleus transcriptomic resource of developing auditory cortex is thus a powerful discovery platform with which to identify mediators of tonotopic plasticity.
Collapse
Affiliation(s)
- Brian T Kalish
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Tania R Barkat
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Center for Brain Science, Harvard University, Cambridge, MA 02138
| | - Erin E Diel
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Center for Brain Science, Harvard University, Cambridge, MA 02138
| | | | | | - Takao K Hensch
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138;
- Center for Brain Science, Harvard University, Cambridge, MA 02138
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Child Brain Development, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
15
|
Svirsky S, Henchir J, Li Y, Ma X, Carlson S, Dixon CE. Neurogranin Protein Expression Is Reduced after Controlled Cortical Impact in Rats. J Neurotrauma 2020; 37:939-949. [PMID: 31691647 PMCID: PMC7175627 DOI: 10.1089/neu.2019.6759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic brain injury (TBI) is known to cause short- and long-term synaptic changes in the brain, possibly underlying downstream cognitive impairments. Neuronal levels of neurogranin, a calcium-sensitive calmodulin-binding protein essential for synaptic plasticity and postsynaptic signaling, are correlated with cognitive function. This study aims to understand the effect of TBI on neurogranin by characterizing changes in protein expression at various time points after injury. Adult, male rats were subjected to either controlled cortical impact (CCI) or control surgery. Expression of neurogranin and post-synaptic density 95 (PSD-95) were evaluated by Western blot in the cortex and hippocampus at 24 h and 1, 2, and 4 weeks post-injury. We hypothesized that CCI reduces neurogranin levels in the cortex and hippocampus, and demonstrate different expression patterns from PSD-95. Neurogranin levels were reduced in the ipsilateral cortex and hippocampus up to 2 weeks after injury but recovered to sham levels by 4 weeks. The contralateral cortex and hippocampus were relatively resistant to changes in neurogranin expression post-injury. Qualitative immunohistochemical assessment corroborated the immunoblot findings. Particularly, the pericontusional cortex and ipsilateral Cornu Ammonis (CA)3 region showed marked reduction in immunoreactivity. PSD-95 demonstrated similar expression patterns to neurogranin in the cortex; however, in the hippocampus, protein expression was increased compared with sham at the 2 and 4 week time points. Our results indicate that CCI lowers neurogranin expression with temporal and regional specificity and that this occurs independently of dendritic loss. Further understanding of the role of neurogranin in synaptic biology after TBI will elucidate pathological mechanisms contributing to cognitive dysfunction.
Collapse
Affiliation(s)
- Sarah Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Youming Li
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Xiecheng Ma
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Shaun Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Zhong L, Gerges NZ. Neurogranin Regulates Metaplasticity. Front Mol Neurosci 2020; 12:322. [PMID: 32038160 PMCID: PMC6992556 DOI: 10.3389/fnmol.2019.00322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/17/2019] [Indexed: 01/14/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are two major forms of synaptic plasticity that are widely accepted as cellular mechanisms involved in learning and memory. Metaplasticity is a process whereby modifications in synaptic processes shift the threshold for subsequent plasticity. While metaplasticity has been functionally observed, its molecular basis is not well understood. Here, we report that neurogranin (Ng) regulates metaplasticity by shifting the threshold toward potentiation, i.e., increasing Ng in hippocampal neurons lowers the threshold for LTP and augments the threshold for LTD. We also show that Ng does not change the ultrastructural localization of calmodulin (CaM)-dependent protein Kinase II (CaMKII) or calcineurin, critical enzymes for the induction of LTP and LTD, respectively. Interestingly, while CaMKII concentrates close to the plasma membrane, calcineurin concentrates away from the plasma membrane. These data, along with the previous observation showing Ng targets CaM closer to the plasma membrane, suggesting that shifting the localization of CaM within the dendritic spines and closer to the plasma membrane, where there is more CaMKII, may be favoring the activation of CaMKII vs. that of calcineurin. Thus, the regulation of CaM localization/targeting within dendritic spines by Ng may provide a mechanistic basis for the regulation of metaplasticity.
Collapse
Affiliation(s)
| | - Nashaat Z. Gerges
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
17
|
Hippocampal Protein Kinase C Gamma Signaling Mediates the Impairment of Spatial Learning and Memory in Prenatally Stressed Offspring Rats. Neuroscience 2019; 414:186-199. [DOI: 10.1016/j.neuroscience.2019.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/06/2019] [Accepted: 06/23/2019] [Indexed: 12/13/2022]
|
18
|
Nazir FH, Becker B, Brinkmalm A, Höglund K, Sandelius Å, Bergström P, Satir TM, Öhrfelt A, Blennow K, Agholme L, Zetterberg H. Expression and secretion of synaptic proteins during stem cell differentiation to cortical neurons. Neurochem Int 2018; 121:38-49. [PMID: 30342961 PMCID: PMC6232556 DOI: 10.1016/j.neuint.2018.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/01/2018] [Accepted: 10/17/2018] [Indexed: 11/23/2022]
Abstract
Synaptic function and neurotransmitter release are regulated by specific proteins. Cortical neuronal differentiation of human induced pluripotent stem cells (hiPSC) provides an experimental model to obtain more information about synaptic development and physiology in vitro. In this study, expression and secretion of the synaptic proteins, neurogranin (NRGN), growth-associated protein-43 (GAP-43), synaptosomal-associated protein-25 (SNAP-25) and synaptotagmin-1 (SYT-1) were analyzed during cortical neuronal differentiation. Protein levels were measured in cells, modeling fetal cortical development and in cell-conditioned media which was used as a model of cerebrospinal fluid (CSF), respectively. Human iPSC-derived cortical neurons were maintained over a period of at least 150 days, which encompasses the different stages of neuronal development. The differentiation was divided into the following stages: hiPSC, neuro-progenitors, immature and mature cortical neurons. We show that NRGN was first expressed and secreted by neuro-progenitors while the maximum was reached in mature cortical neurons. GAP-43 was expressed and secreted first by neuro-progenitors and its expression increased markedly in immature cortical neurons. SYT-1 was expressed and secreted already by hiPSC but its expression and secretion peaked in mature neurons. SNAP-25 was first detected in neuro-progenitors and the expression and secretion increased gradually during neuronal stages reaching a maximum in mature neurons. The sensitive analytical techniques used to monitor the secretion of these synaptic proteins during cortical development make these data unique, since the secretion of these synaptic proteins has not been investigated before in such experimental models. The secretory profile of synaptic proteins, together with low release of intracellular content, implies that mature neurons actively secrete these synaptic proteins that previously have been associated with neurodegenerative disorders, including Alzheimer's disease. These data support further studies of human neuronal and synaptic development in vitro, and would potentially shed light on the mechanisms underlying altered concentrations of the proteins in bio-fluids in neurodegenerative diseases.
Collapse
Affiliation(s)
- Faisal Hayat Nazir
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-405 30, Gothenburg, Sweden; Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden.
| | - Bruno Becker
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Kina Höglund
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Åsa Sandelius
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden
| | - Petra Bergström
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-405 30, Gothenburg, Sweden
| | - Tugce Munise Satir
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-405 30, Gothenburg, Sweden
| | - Annika Öhrfelt
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Lotta Agholme
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-405 30, Gothenburg, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden; UCL, Institute of Neurology, Department of Neurodegerative Disease, University College London, Queen Square, London, WC1N 3BG, UK; UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
| |
Collapse
|
19
|
Intrahippocampal injection of a lentiviral vector expressing neurogranin enhances cognitive function in 5XFAD mice. Exp Mol Med 2018; 50:e461. [PMID: 29568074 PMCID: PMC5898899 DOI: 10.1038/emm.2017.302] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023] Open
Abstract
Progressive cognitive declines are the main clinical symptoms of Alzheimer's disease (AD). Cognitive impairment in AD is directly correlated with amyloid beta (Aβ)-mediated synaptic deficits. It is known that upregulation of neurogranin (Ng), a postsynaptic protein, contributes to the enhancement of synaptic plasticity and cognitive function. By contrast, downregulation of Ng expression results in learning and memory impairments. Interestingly, Ng expression is significantly reduced in the parenchyma of brains with AD. However, the pathological role that downregulated Ng plays in the cognitive dysfunctions observed in AD remains unclear. Therefore, the present study examined whether enhancing Ng expression affected cognitive functions in 5XFAD mice, an animal model of AD. We found that the Ng reductions and cognitive decline observed in 5XFAD mice were restored in mice that were intrahippocampally injected with an Ng-expressing lentiviral vector. Furthermore, overexpression of Ng upregulated expression of postsynaptic density protein-95 in the hippocampus of 5XFAD mice. These results suggest that the cause of cognitive decline in AD may be at least partially associated with reduced Ng levels, and thus, supplementation of Ng may be an appropriate therapeutic strategy for individuals with AD.
Collapse
|
20
|
Stefanoska K, Volkerling A, Bertz J, Poljak A, Ke YD, Ittner LM, Ittner A. An N-terminal motif unique to primate tau enables differential protein-protein interactions. J Biol Chem 2018; 293:3710-3719. [PMID: 29382714 DOI: 10.1074/jbc.ra118.001784] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 01/25/2018] [Indexed: 01/05/2023] Open
Abstract
Compared with other mammalian species, humans are particularly susceptible to tau-mediated neurodegenerative disorders. Differential interactions of the tau protein with other proteins are critical for mediating tau's physiological functions as well as tau-associated pathological processes. Primate tau harbors an 11-amino acid-long motif in its N-terminal region (residues 18-28), which is not present in non-primate species and whose function is unknown. Here, we used deletion mutagenesis to remove this sequence region from the longest human tau isoform, followed by glutathione S-transferase (GST) pulldown assays paired with isobaric tags for relative and absolute quantitation (iTRAQ) multiplex labeling, a quantitative method to measure protein abundance by mass spectrometry. Using this method, we found that the primate-specific N-terminal tau motif differentially mediates interactions with neuronal proteins. Among these binding partners are proteins involved in synaptic transmission (synapsin-1 and synaptotagmin-1) and signaling proteins of the 14-3-3 family. Furthermore, we identified an interaction of tau with a member of the annexin family (annexin A5) that was linked to the 11-residue motif. These results suggest that primate Tau has evolved specific residues that differentially regulate protein-protein interactions compared with tau proteins from other non-primate mammalian species. Our findings provide in vitro insights into tau's interactions with other proteins that may be relevant to human disease.
Collapse
Affiliation(s)
| | | | - Josefine Bertz
- From the Dementia Research Unit, School of Medical Sciences
| | - Anne Poljak
- the Bioanalytical Mass Spectrometry Facility, and
| | - Yazi D Ke
- the Motor Neuron Disease Unit, School of Medical Sciences, The University of New South Wales, Sydney, New South Wales 2052 and
| | - Lars M Ittner
- From the Dementia Research Unit, School of Medical Sciences, .,Neuroscience Australia, Sydney, New South Wales 2031, Australia
| | - Arne Ittner
- From the Dementia Research Unit, School of Medical Sciences
| |
Collapse
|
21
|
Han KS, Cooke SF, Xu W. Experience-Dependent Equilibration of AMPAR-Mediated Synaptic Transmission during the Critical Period. Cell Rep 2017; 18:892-904. [PMID: 28122240 DOI: 10.1016/j.celrep.2016.12.084] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/21/2016] [Accepted: 12/26/2016] [Indexed: 11/17/2022] Open
Abstract
Experience-dependent synapse refinement is essential for functional optimization of neural circuits. However, how sensory experience sculpts excitatory synaptic transmission is poorly understood. Here, we show that despite substantial remodeling of synaptic connectivity, AMPAR-mediated synaptic transmission remains at equilibrium during the critical period in the mouse primary visual cortex. The maintenance of this equilibrium requires neurogranin (Ng), a postsynaptic calmodulin-binding protein important for synaptic plasticity. With normal visual experience, loss of Ng decreased AMPAR-positive synapse numbers, prevented AMPAR-silent synapse maturation, and increased spine elimination. Importantly, visual deprivation halted synapse loss caused by loss of Ng, revealing that Ng coordinates experience-dependent AMPAR-silent synapse conversion to AMPAR-active synapses and synapse elimination. Loss of Ng also led to sensitized long-term synaptic depression (LTD) and impaired visually guided behavior. Our synaptic interrogation reveals that experience-dependent coordination of AMPAR-silent synapse conversion and synapse elimination hinges upon Ng-dependent mechanisms for constructive synaptic refinement during the critical period.
Collapse
Affiliation(s)
- Kyung-Seok Han
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Samuel F Cooke
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Weifeng Xu
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
22
|
Reker AN, Oliveros A, Sullivan JM, Nahar L, Hinton DJ, Kim T, Bruner RC, Choi DS, Goeders NE, Nam HW. Neurogranin in the nucleus accumbens regulates NMDA receptor tolerance and motivation for ethanol seeking. Neuropharmacology 2017; 131:58-67. [PMID: 29225043 DOI: 10.1016/j.neuropharm.2017.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/08/2017] [Accepted: 12/05/2017] [Indexed: 01/15/2023]
Abstract
Dysfunction of N-methyl-d-aspartate receptor (NMDAR) signaling in the nucleus accumbens (NAc) has been implicated in the pathophysiology of alcohol use disorders (AUD). Neurogranin (Ng), a calmodulin-binding protein, is exclusively expressed in the post-synapse, and mediates NMDAR driven synaptic plasticity by regulating the calcium-calmodulin (Ca2+-CaM) pathway. To study the functional role of Ng in AUD, we administrated behavior tests including Pavlovian instrument transfer (PIT), operant conditioning, and rotarod test using Ng null mice (Ng-/- mice). We used adeno-associated virus (AAV)-mediated Ng expression and pharmacological manipulation to validate behavioral responses in Ng-/- mice. The results from our multidisciplinary approaches demonstrated that deficit of Ng increases tolerance to NMDAR inhibition and elicit faster cue reactivity during PIT without changes in ethanol reward. Operant conditioning results demonstrated that Ng-/- mice self-administered significantly more ethanol and displayed reduced sensitivity to aversive motivation. We identified that ethanol exposure decreases mGluR5 (metabotropic glutamate receptor 5) expression in the NAc of Ng-/- mice and pharmacological inhibition of mGluR5 reverses NMDAR desensitization in Ng-/- mice. Together these findings specifically suggest that accumbal Ng plays an essential role in the counterbalance between NMDAR and mGluR5 signaling; which alters NMDAR resistance, and thereby altering aversive motivation for ethanol and may ultimately contribute to susceptibility for alcohol addiction.
Collapse
Affiliation(s)
- Ashlie N Reker
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alfredo Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - John M Sullivan
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Lailun Nahar
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - David J Hinton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Taehyun Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Robert C Bruner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| |
Collapse
|
23
|
Oliveros A, Wininger K, Sens J, Larsson MK, Liu XC, Choi S, Faka A, Schwieler L, Engberg G, Erhardt S, Choi DS. LPS-induced cortical kynurenic acid and neurogranin-NFAT signaling is associated with deficits in stimulus processing during Pavlovian conditioning. J Neuroimmunol 2017; 313:1-9. [PMID: 29153599 DOI: 10.1016/j.jneuroim.2017.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/04/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
The N-Methyl-d-Aspartate receptor (NMDAR) antagonist kynurenic acid (KYNA) and the post-synaptic calmodulin binding protein neurogranin (Nrgn) have been implicated in neurological and neuropsychiatric conditions including Alzheimer's disease and schizophrenia. This study indicates that systemic dual-lipopolysaccharide (LPS) injections increases KYNA in the medial prefrontal cortex (mPFC), which is accompanied with increased phosphorylation of nuclear factor kappa chain of activated B cells (NFκB) and activation of the nuclear factor of activated T- cells (NFAT). Our results also indicate that dual-LPS increases Nrgn phosphorylation and concomitantly reduces phosphorylation of calmodulin kinase-II (CaMKII). We confirmed that systemic blockade of kynurenine-3 monooxygenase in conjunction with kynurenine administration results in significant increases in Nrgn phosphorylation and a significant reduction of CaMKII phosphorylation in the mPFC. Consequently, dual-LPS administration induced significant impairments in stimulus processing during Pavlovian conditioning. Taken together, our study indicates that elevations in KYNA in the mPFC can directly regulate NMDA-Nrgn-CaMKII signaling, suggesting that neuroinflammatory conditions affecting this pathway may be associated with cognitive dysfunction.
Collapse
Affiliation(s)
- A Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - K Wininger
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - J Sens
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - M K Larsson
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - X C Liu
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - S Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - A Faka
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - L Schwieler
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - G Engberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - S Erhardt
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - D S Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
24
|
Jȩdrzejewska-Szmek J, Luczak V, Abel T, Blackwell KT. β-adrenergic signaling broadly contributes to LTP induction. PLoS Comput Biol 2017; 13:e1005657. [PMID: 28742159 PMCID: PMC5546712 DOI: 10.1371/journal.pcbi.1005657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 08/07/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
Long-lasting forms of long-term potentiation (LTP) represent one of the major cellular mechanisms underlying learning and memory. One of the fundamental questions in the field of LTP is why different molecules are critical for long-lasting forms of LTP induced by diverse experimental protocols. Further complexity stems from spatial aspects of signaling networks, such that some molecules function in the dendrite and some are critical in the spine. We investigated whether the diverse experimental evidence can be unified by creating a spatial, mechanistic model of multiple signaling pathways in hippocampal CA1 neurons. Our results show that the combination of activity of several key kinases can predict the occurrence of long-lasting forms of LTP for multiple experimental protocols. Specifically Ca2+/calmodulin activated kinase II, protein kinase A and exchange protein activated by cAMP (Epac) together predict the occurrence of LTP in response to strong stimulation (multiple trains of 100 Hz) or weak stimulation augmented by isoproterenol. Furthermore, our analysis suggests that activation of the β-adrenergic receptor either via canonical (Gs-coupled) or non-canonical (Gi-coupled) pathways underpins most forms of long-lasting LTP. Simulations make the experimentally testable prediction that a complete antagonist of the β-adrenergic receptor will likely block long-lasting LTP in response to strong stimulation. Collectively these results suggest that converging molecular mechanisms allow CA1 neurons to flexibly utilize signaling mechanisms best tuned to temporal pattern of synaptic input to achieve long-lasting LTP and memory storage. Long-term potentiation of the strength of synaptic connections is a mechanism of learning and memory storage. One of the most confusing aspects of hippocampal synaptic potentiation is that numerous experiments have revealed the requirement for a plethora of signaling molecules. Furthermore the degree to which molecules activated by the stress response modify hippocampal synaptic potentiation and memory is still unclear. We used a computational model to demonstrate that this molecular diversity can be explained by considering a combination of several key molecules. We also show that activation of β-adrenergic receptors by the stress response appears to be involved in most forms of synaptic potentiation, though in some cases unconventional mechanisms are utilized. This suggests that novel treatments for stress-related disorders may have more success if they target unconventional mechanisms activated by β-adrenergic receptors.
Collapse
Affiliation(s)
- Joanna Jȩdrzejewska-Szmek
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
| | - Vincent Luczak
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kim T Blackwell
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
25
|
Cannady R, McGonigal JT, Newsom RJ, Woodward JJ, Mulholland PJ, Gass JT. Prefrontal Cortex K Ca2 Channels Regulate mGlu 5-Dependent Plasticity and Extinction of Alcohol-Seeking Behavior. J Neurosci 2017; 37:4359-4369. [PMID: 28320841 PMCID: PMC5413180 DOI: 10.1523/jneurosci.2873-16.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 12/17/2022] Open
Abstract
Identifying novel treatments that facilitate extinction learning could enhance cue-exposure therapy and reduce high relapse rates in alcoholics. Activation of mGlu5 receptors in the infralimbic prefrontal cortex (IL-PFC) facilitates learning during extinction of cue-conditioned alcohol-seeking behavior. Small-conductance calcium-activated potassium (KCa2) channels have also been implicated in extinction learning of fear memories, and mGlu5 receptor activation can reduce KCa2 channel function. Using a combination of electrophysiological, pharmacological, and behavioral approaches, this study examined KCa2 channels as a novel target to facilitate extinction of alcohol-seeking behavior in rats. This study also explored related neuronal and synaptic mechanisms within the IL-PFC that underlie mGlu5-dependent enhancement of extinction learning. Using whole-cell patch-clamp electrophysiology, activation of mGlu5 in ex vivo slices significantly reduced KCa2 channel currents in layer V IL-PFC pyramidal neurons, confirming functional downregulation of KCa2 channel activity by mGlu5 receptors. Additionally, positive modulation of KCa2 channels prevented mGlu5 receptor-dependent facilitation of long-term potentiation in the IL-PFC. Systemic and intra-IL-PFC treatment with apamin (KCa2 channel allosteric inhibitor) significantly enhanced extinction of alcohol-seeking behavior across multiple extinction sessions, an effect that persisted for 3 weeks, but was not observed after apamin microinfusions into the prelimbic PFC. Positive modulation of IL-PFC KCa2 channels significantly attenuated mGlu5-dependent facilitation of alcohol cue-conditioned extinction learning. These data suggest that mGlu5-dependent facilitation of extinction learning and synaptic plasticity in the IL-PFC involves functional inhibition of KCa2 channels. Moreover, these findings demonstrate that KCa2 channels are a novel target to facilitate long-lasting extinction of alcohol-seeking behavior.SIGNIFICANCE STATEMENT Alcohol use disorder is a chronic relapsing disorder that is associated with compulsive alcohol-seeking behavior. One of the main causes of alcohol relapse is the craving caused by environmental cues that are associated with alcohol. These cues are formed by normal learning and memory principles, and the understanding of the brain mechanisms that help form these associations can lead to the development of drugs and/or behavior therapies that reduce the impact that these cues have on relapse in alcoholics.
Collapse
Affiliation(s)
- Reginald Cannady
- Department of Neuroscience
- Department of Psychiatry & Behavioral Sciences, and
| | | | | | - John J Woodward
- Department of Neuroscience
- Department of Psychiatry & Behavioral Sciences, and
| | | | - Justin T Gass
- Department of Neuroscience,
- Department of Psychiatry & Behavioral Sciences, and
- Charleston Alcohol Research Center, Addiction Sciences Division, College of Health Professions, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
26
|
Kaleka KS, Gerges NZ. Neurogranin restores amyloid β-mediated synaptic transmission and long-term potentiation deficits. Exp Neurol 2015; 277:115-123. [PMID: 26721336 DOI: 10.1016/j.expneurol.2015.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 12/08/2015] [Accepted: 12/18/2015] [Indexed: 11/16/2022]
Abstract
Amyloid β (Aβ) is widely considered one of the early causes of cognitive deficits observed in Alzheimer's disease. Many of the deficits caused by Aβ are attributed to its disruption of synaptic function represented by its blockade of long-term potentiation (LTP) and its induction of synaptic depression. Identifying pathways that reverse these synaptic deficits may open the door to new therapeutic targets. In this study, we explored the possibility that Neurogranin (Ng)-a postsynaptic calmodulin (CaM) targeting protein that enhances synaptic function-may rescue Aβ-mediated deficits in synaptic function. Our results show that Ng is able to reverse synaptic depression and LTP deficits induced by Aβ. Furthermore, Ng's restoration of synaptic transmission is through the insertion of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPARs). These restorative effects of Ng are dependent on the interaction of Ng and CaM and CaM-dependent activation of CaMKII. Overall, this study identifies a novel mechanism to rescue synaptic deficits induced by Aβ oligomers. It also suggests Ng and CaM signaling as potential therapeutic targets for Alzheimer's disease as well as important tools to further explore the pathophysiology underlying the disease.
Collapse
Affiliation(s)
- Kanwardeep Singh Kaleka
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States.
| |
Collapse
|
27
|
Janelidze S, Hertze J, Zetterberg H, Landqvist Waldö M, Santillo A, Blennow K, Hansson O. Cerebrospinal fluid neurogranin and YKL-40 as biomarkers of Alzheimer's disease. Ann Clin Transl Neurol 2015; 3:12-20. [PMID: 26783546 PMCID: PMC4704480 DOI: 10.1002/acn3.266] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/09/2015] [Accepted: 10/19/2015] [Indexed: 12/18/2022] Open
Abstract
Objective Widespread implementation of cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) in clinical settings requires improved accuracy for diagnosis of prodromal disease and for distinguishing AD from non‐AD dementias. Novel and promising CSF biomarkers include neurogranin, a marker of synaptic degeneration, and YKL‐40, a marker of neuroinflammation. Methods CSF neurogranin and YKL‐40 were measured in a cohort of 338 individuals including cognitively healthy controls and patients with stable mild cognitive impairment (sMCI), MCI who later developed AD (MCI‐AD), AD dementia, Parkinson's disease dementia (PDD), dementia with Lewy bodies (DLB), vascular dementia (VaD), and frontotemporal dementia (FTD). The diagnostic accuracy of neurogranin and YKL‐40 were compared with the core AD biomarkers, β‐amyloid (Aβ42 and Aβ40) and tau. Results Neurogranin levels were increased in AD and decreased in non‐AD dementia compared with healthy controls. As a result, AD patients showed considerably higher CSF levels of neurogranin than DLB/PDD, VaD and FTD patients. CSF YKL‐40 levels were increased in AD compared with DLB/PDD but not with VaD or FTD. Neither CSF neurogranin nor YKL‐40 levels differed significantly between sMCI patients and MCI‐AD patients. Both biomarkers correlated positively with CSF Aβ40 and tau. CSF neurogranin and YKL‐40 could separate AD dementia from non‐AD dementias (neurogranin, area under the curve [AUC] = 0.761; YKL‐40, AUC = 0.604; Aβ42/neurogranin, AUC = 0.849; Aβ42/YKL‐40, AUC = 0.785), but the diagnostic accuracy was not better compared to CSF Aβ and tau (Aβ42, AUC = 0.755; tau AUC = 0.858; Aβ42/tau, AUC = 0.895; Aβ42/Aβ40, AUC = 0.881). Similar results were obtained when separating sMCI from MCI‐AD cases. Interpretation CSF neurogranin and YKL‐40 do not improve the diagnostic accuracy of either prodromal AD or AD dementia when compared to the core CSF AD biomarkers. Nevertheless, the CSF level of neurogranin is selectively increased in AD dementia, whereas YKL‐40 is increased in both AD and FTD suggesting that synaptic degeneration and glial activation may be important in these neurodegenerative conditions.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden
| | - Joakim Hertze
- Clinical Memory Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden; Memory Clinic Skåne University Hospital Malmö Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory Institute of Neuroscience and Physiology Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden; Department of Molecular Neuroscience UCL Institute of Neurology Queen Square London United Kingdom
| | - Maria Landqvist Waldö
- Memory Clinic Skåne University Hospital Malmö Sweden; Section of Geriatric Psychiatry Department of Clinical Sciences Lund University Lund Sweden
| | - Alexander Santillo
- Clinical Memory Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden; Memory Clinic Skåne University Hospital Malmö Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory Institute of Neuroscience and Physiology Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden; Memory Clinic Skåne University Hospital Malmö Sweden
| |
Collapse
|