51
|
Pena DA, Duarte ML, Pramio DT, Devi LA, Schechtman D. Exploring Morphine-Triggered PKC-Targets and Their Interaction with Signaling Pathways Leading to Pain via TrkA. Proteomes 2018; 6:proteomes6040039. [PMID: 30301203 PMCID: PMC6313901 DOI: 10.3390/proteomes6040039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/29/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022] Open
Abstract
It is well accepted that treatment of chronic pain with morphine leads to μ opioid receptor (MOR) desensitization and the development of morphine tolerance. MOR activation by the selective peptide agonist, D-Ala2, N-MePhe4, Gly-ol]-enkephalin(DAMGO), leads to robust G protein receptor kinase activation, β-arrestin recruitment, and subsequent receptor endocytosis, which does not occur in an activation by morphine. However, MOR activation by morphine induces receptor desensitization, in a Protein kinase C (PKC) dependent manner. PKC inhibitors have been reported to decrease receptor desensitization, reduce opiate tolerance, and increase analgesia. However, the exact role of PKC in these processes is not clearly delineated. The difficulties in establishing a particular role for PKC have been, in part, due to the lack of reagents that allow the selective identification of PKC targets. Recently, we generated a conformation state-specific anti-PKC antibody that preferentially recognizes the active state of this kinase. Using this antibody to selectively isolate PKC substrates and a proteomics strategy to establish the identity of the proteins, we examined the effect of morphine treatment on the PKC targets. We found an enhanced interaction of a number of proteins with active PKC, in the presence of morphine. In this article, we discuss the role of these proteins in PKC-mediated MOR desensitization and analgesia. In addition, we posit a role for some of these proteins in mediating pain by TrKA activation, via the activation of transient receptor potential cation channel subfamily V member 1 (TRPV1). Finally, we discuss how these new PKC interacting proteins and pathways could be targeted for the treatment of pain.
Collapse
Affiliation(s)
- Darlene A Pena
- Department of Biochemistry, Chemistry Institute, University of São Paulo, Sao Paulo 05508-220, Brazil.
| | - Mariana Lemos Duarte
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Dimitrius T Pramio
- Department of Biochemistry, Chemistry Institute, University of São Paulo, Sao Paulo 05508-220, Brazil.
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Deborah Schechtman
- Department of Biochemistry, Chemistry Institute, University of São Paulo, Sao Paulo 05508-220, Brazil.
| |
Collapse
|
52
|
Becker B, Nazir FH, Brinkmalm G, Camporesi E, Kvartsberg H, Portelius E, Boström M, Kalm M, Höglund K, Olsson M, Zetterberg H, Blennow K. Alzheimer-associated cerebrospinal fluid fragments of neurogranin are generated by Calpain-1 and prolyl endopeptidase. Mol Neurodegener 2018; 13:47. [PMID: 30157938 PMCID: PMC6116393 DOI: 10.1186/s13024-018-0279-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/17/2018] [Indexed: 12/16/2022] Open
Abstract
Background Neurogranin (Ng) is a small 7.6 kDa postsynaptic protein that has been detected at elevated concentrations in cerebrospinal fluid (CSF) of patients with Alzheimer’s disease (AD), both as a full-length molecule and as fragments from its C-terminal half. Ng is involved in postsynaptic calcium (Ca) signal transduction and memory formation via binding to calmodulin in a Ca-dependent manner. The mechanism of Ng secretion from neurons to CSF is currently unknown, but enzymatic cleavage of Ng may be of relevance. Therefore, the aim of the study was to identify the enzymes responsible for the cleavage of Ng, yielding the Ng fragment pattern of C-terminal fragments detectable and increased in CSF of AD patients. Methods Fluorigenic quenched FRET probes containing sequences of Ng were utilized to identify Ng cleaving activities among enzymes known to have increased activity in AD and in chromatographically fractionated mouse brain extracts. Results Human Calpain-1 and prolyl endopeptidase were identified as the candidate enzymes involved in the formation of endogenous Ng peptides present in CSF, cleaving mainly in the central region of Ng, and between amino acids 75_76 in the Ng sequence, respectively. The cleavage by Calpain-1 affects the IQ domain of Ng, which may deactivate or change the function of Ng in Ca2+/calmodulin -dependent signaling for synaptic plasticity. While shorter Ng fragments were readily cleaved in vitro by prolyl endopeptidase, the efficiency of cleavage on larger Ng fragments was much lower. Conclusions Calpain-1 and prolyl endopeptidase cleave Ng in the IQ domain and near the C-terminus, respectively, yielding specific fragments of Ng in CSF. These fragments may give clues to the roles of increased activities of these enzymes in the pathophysiology of AD, and provide possible targets for pharmacologic intervention. Electronic supplementary material The online version of this article (10.1186/s13024-018-0279-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Faisal Hayat Nazir
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Martina Boström
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marie Kalm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Maria Olsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
53
|
Schipke CG, De Vos A, Fuentes M, Jacobs D, Vanmechelen E, Peters O. Neurogranin and BACE1 in CSF as Potential Biomarkers Differentiating Depression with Cognitive Deficits from Early Alzheimer's Disease: A Pilot Study. Dement Geriatr Cogn Dis Extra 2018; 8:277-289. [PMID: 30186306 PMCID: PMC6120408 DOI: 10.1159/000489847] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background/Aims Major depressive disorder (MDD) can cooccur with early Alzheimer's disease (AD) or may cause memory problems independently of AD. Previous studies have suggested that the AD-related cerebrospinal fluid (CSF) biomarkers tau and Aβ(1–42) could help discriminate between early AD and depression unrelated to AD. Moreover, the postsynaptic protein neurogranin and presynaptic BACE1 have increasingly gained attention as potential new AD biomarkers, but they have not yet been investigated concerning depression. Methods Using ELISAs, we studied CSF neurogranin and BACE1 levels in patients with mild (n = 21) and moderate (n = 19) AD, as well as in MDD patients with (n = 20) and without (n = 20) cognitive deficits. The clinical examinations included analyses of t-tau, Aβ(1–42), and Aβ(1–40), besides neuropsychological tests and cranial magnetic resonance imaging. Depressive symptom severity was assessed using the Geriatric Depression Scale (GDS). Results Along with classic AD biomarkers, neurogranin and BACE1 CSF levels differed between moderate AD and MDD (p ≤ 0.01). MDD associated with cognitive deficits was distinguished from mild AD through the CSF neurogranin/BACE1 ratio (p < 0.05), which was strongly correlated with GDS scores (ρ = −0.656; p < 0.01). Conclusion The neurogranin/BACE1 ratio in CSF can distinguish between depression and AD among patients with similar cognitive deficits, along with the classic AD biomarkers. Further longitudinal studies are ongoing to identify which biomarkers have prognostic value.
Collapse
Affiliation(s)
- Carola G Schipke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | | | - Manuel Fuentes
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and German Center for Neurodegenerative Diseases (DZNE), Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Dirk Jacobs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | - Eugeen Vanmechelen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | - Oliver Peters
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and German Center for Neurodegenerative Diseases (DZNE), Department of Psychiatry and Psychotherapy, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| |
Collapse
|
54
|
Abstract
De novo protein synthesis is critical for memory formation. We found that protein synthesis during acquisition is transiently required for contextual memory formation. We identified one candidate gene, Nrgn (encoding protein neurogranin, Ng) with enhanced translation upon novel-context exposure, and found that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. Furthermore, fragile-X mental retardation protein interacts with the 3′UTR of the Nrgn mRNA, which is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Together, these results indicate that experience-dependent and acute translation of Ng in the hippocampus during memory acquisition enables durable context memory encoding. Experience induces de novo protein synthesis in the brain and protein synthesis is required for long-term memory. It is important to define the critical temporal window of protein synthesis and identify newly synthesized proteins required for memory formation. Using a behavioral paradigm that temporally separates the contextual exposure from the association with fear, we found that protein synthesis during the transient window of context exposure is required for contextual memory formation. Among an array of putative activity-dependent translational neuronal targets tested, we identified one candidate, a schizophrenia-associated candidate mRNA, neurogranin (Ng, encoded by the Nrgn gene) responding to novel-context exposure. The Ng mRNA was recruited to the actively translating mRNA pool upon novel-context exposure, and its protein levels were rapidly increased in the hippocampus. By specifically blocking activity-dependent translation of Ng using virus-mediated molecular perturbation, we show that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. We further interrogated the molecular mechanism underlying the experience-dependent translation of Ng, and found that fragile-X mental retardation protein (FMRP) interacts with the 3′UTR of the Nrgn mRNA and is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Our results reveal that FMRP-mediated, experience-dependent, rapid enhancement of Ng translation in the hippocampus during the memory acquisition enables durable context memory encoding.
Collapse
|
55
|
Guha D, Wagner MCE, Ayyavoo V. Human immunodeficiency virus type 1 (HIV-1)-mediated neuroinflammation dysregulates neurogranin and induces synaptodendritic injury. J Neuroinflammation 2018; 15:126. [PMID: 29703241 PMCID: PMC5923011 DOI: 10.1186/s12974-018-1160-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/13/2018] [Indexed: 11/30/2022] Open
Abstract
Background Human immunodeficiency virus type 1 (HIV-1)-associated neurocognitive disorder (HAND) is a common outcome of a majority of HIV-1-infected subjects and is associated with synaptodendritic damage. Neurogranin (Ng), a postsynaptic protein, and calmodulin (CaM) are two important players of synaptic integrity/functions. The biological role of Ng in the context of HAND is unknown. Methods We compared the expression of Ng in frontal cortex (FC) tissues from control and HIV-1-positive subjects with and without HAND by immunohistochemistry, western blot, and qRT-PCR. The interaction between Ng and CaM was analyzed by co-immunoprecipitation. Ng, microtubule-associated protein 2 (MAP2), CaM, CaM-dependent protein kinase II (CaMKII), CREB, synaptophysin (Syp), and synapsin I (Syn I) expressions were evaluated by western blot using FC tissue lysates and differentiated SH-SY5Y (dSH-SY5Y) cells. Identification of inflammatory factors related to Ng loss was accomplished by exposing dSH-SY5Y cells to HIV-1 and mock-infected monocyte-derived macrophage (MDM) supernatants or HIV-1 NLYU2 pseudotyped with VSV-G-Env. Levels of interleukin (IL)-1β, IL-8, tumor necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP)-1, MCP-2, and CXCL5 in MDM supernatants were measured by ELISA. Association of IL-1β and IL-8 to Ng expression in context of HIV-1 infection was evaluated in the presence or absence of neutralizing antibodies against these cytokines. Results Expression level of Ng was reduced significantly in FC of HAND-positive (HAND+) patients compared to uninfected individuals. Although no difference was found in CaM expression, interaction between Ng and CaM was reduced in HAND+ patients, which was associated with decreased level of CaMKII, a downstream signaling molecule of CaM pathway. This in turn resulted in reduction of synaptic markers, Syp and Syn I. HIV-1 infection directly had no considerable effect on dysregulation of Ng expression in dSH-SY5Y cells, whereas high amount of pro-inflammatory IL-1β and IL-8 in HIV-1-infected MDM supernatants was associated with significant reduction in Ng expression. Conclusions Synaptic damage in HAND+ patients could be a result of abrogation of Ng through HIV-1-induced inflammation that dysregulates Ng-CaM interaction and downstream signaling cascades associated with synaptodendritic functions. This is the first study evaluating the potential role of Ng in the context of HIV-1 neuropathogenesis.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, 2117 Pitt Public Health, 130 DeSoto Street, Pittsburgh, PA, 15261, USA
| | - Marc C E Wagner
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, 2117 Pitt Public Health, 130 DeSoto Street, Pittsburgh, PA, 15261, USA
| | - Velpandi Ayyavoo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, 2117 Pitt Public Health, 130 DeSoto Street, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
56
|
Hippocampal Protein Kinase C Signaling Mediates the Short-Term Memory Impairment Induced by Delta9-Tetrahydrocannabinol. Neuropsychopharmacology 2018; 43:1021-1031. [PMID: 28816239 PMCID: PMC5854793 DOI: 10.1038/npp.2017.175] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 08/02/2017] [Accepted: 08/09/2017] [Indexed: 01/17/2023]
Abstract
Cannabis affects cognitive performance through the activation of the endocannabinoid system, and the molecular mechanisms involved in this process are poorly understood. Using the novel object-recognition memory test in mice, we found that the main psychoactive component of cannabis, delta9-tetrahydrocannabinol (THC), alters short-term object-recognition memory specifically involving protein kinase C (PKC)-dependent signaling. Indeed, the systemic or intra-hippocampal pre-treatment with the PKC inhibitors prevented the short-term, but not the long-term, memory impairment induced by THC. In contrast, systemic pre-treatment with mammalian target of rapamycin complex 1 inhibitors, known to block the amnesic-like effects of THC on long-term memory, did not modify such a short-term cognitive deficit. Immunoblot analysis revealed a transient increase in PKC signaling activity in the hippocampus after THC treatment. Thus, THC administration induced the phosphorylation of a specific Ser residue in the hydrophobic-motif at the C-terminal tail of several PKC isoforms. This significant immunoreactive band that paralleled cognitive performance did not match in size with the major PKC isoforms expressed in the hippocampus except for PKCθ. Moreover, THC transiently enhanced the phosphorylation of the postsynaptic calmodulin-binding protein neurogranin in a PKC dependent manner. These data demonstrate that THC alters short-term object-recognition memory through hippocampal PKC/neurogranin signaling.
Collapse
|
57
|
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: 3.3] [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
|
58
|
Headley A, De Leon-Benedetti A, Dong C, Levin B, Loewenstein D, Camargo C, Rundek T, Zetterberg H, Blennow K, Wright CB, Sun X. Neurogranin as a predictor of memory and executive function decline in MCI patients. Neurology 2018; 90:e887-e895. [PMID: 29429972 DOI: 10.1212/wnl.0000000000005057] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 12/05/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether high CSF levels of neurogranin (Ng) predict longitudinal decline in memory and executive function during early-stage Alzheimer disease (AD). METHODS Baseline levels of CSF Ng were studied in relation to cross-sectional and longitudinal cognitive performance over 8 years. Data were obtained from the Alzheimer's Disease Neuroimaging Initiative database, and participants with normal cognition (n = 111) and mild cognitive impairment (MCI) (n = 193) were included. RESULTS High levels of CSF Ng were associated with poor baseline memory scores (β = -0.21, p < 0.0001). CSF Ng predicted both memory and executive function decline over time (β = -0.0313, p = 0.0068 and β = -0.0346, p = 0.0169, respectively) independently of age, sex, education, and APOE ε4 status. When the rate of decline by tertiles was examined, CSF Ng was a level-dependent predictor of memory function, whereby the group with highest levels of Ng showed the fastest rates of decline in both memory and executive function. When examined separately, elevated Ng was associated with cognitive decline in participants with MCI but not in those with normal cognition. The levels of CSF Ng were not associated with cognitive measures when tau and amyloid 42 (Aβ42) were controlled for in these analyses. CONCLUSIONS High CSF Ng associates with poor memory scores in participants with MCI cross-sectionally and with poor memory and executive function longitudinally. The association of Ng with cognitive measures disappears when tau and Aβ42 are included in the statistical models. Our findings suggest that CSF Ng may serve as a biomarker of cognition. Synaptic dysfunction contributes to cognitive impairment in early-stage AD.
Collapse
Affiliation(s)
- Alison Headley
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Andres De Leon-Benedetti
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Chuanhui Dong
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Bonnie Levin
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - David Loewenstein
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Christian Camargo
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Tatjana Rundek
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Henrik Zetterberg
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Kaj Blennow
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Clinton B Wright
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD
| | - Xiaoyan Sun
- From the Department of Neuroscience (A.H.), University of California San Diego, La Jolla; Department of Neurology (A.D.L.-B., C.D., B.L., C.C., T.R., X.S.), Evelyn F. McKnight Brain Institute (B.L., C.C., T.R., X.S.), and Psychiatry and Behavioral Sciences (D.L.), University of Miami Miller School of Medicine, FL; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University Gothenburg, Molndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute (H.Z.), London; and National Institute of Neurological Disorders and Stroke (C.B.W.), Bethesda, MD.
| | | |
Collapse
|
59
|
De Vos A, Struyfs H, Jacobs D, Fransen E, Klewansky T, De Roeck E, Robberecht C, Van Broeckhoven C, Duyckaerts C, Engelborghs S, Vanmechelen E. The Cerebrospinal Fluid Neurogranin/BACE1 Ratio is a Potential Correlate of Cognitive Decline in Alzheimer's Disease. J Alzheimers Dis 2018; 53:1523-38. [PMID: 27392859 PMCID: PMC4981899 DOI: 10.3233/jad-160227] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background: In diagnosing Alzheimer’s disease (AD), ratios of cerebrospinal fluid (CSF) biomarkers, such as CSF Aβ1-42/tau, have an improved diagnostic performance compared to the single analytes, yet, still a limited value to predict cognitive decline. Since synaptic dysfunction/loss is closely linked to cognitive impairment, synaptic proteins are investigated as candidate CSF AD progression markers. Objective: We studied CSF levels of the postsynaptic protein neurogranin and protein BACE1, predominantly localized presynaptically, and their relation to CSF total-tau, Aβ1-42, Aβ1-40, and Aβ1-38. All six analytes were considered as single parameters as well as ratios. Methods: Every ELISA involved was based on monoclonal antibodies, including the BACE1 and neurogranin immunoassay. The latter specifically targets neurogranin C-terminally truncated at P75, a more abundant species of the protein in CSF. We studied patients with MCI due to AD (n = 38) and 50 dementia due to AD patients, as well as age-matched cognitively healthy elderly (n = 20). A significant subset of the patients was followed up by clinical and neuropsychologically (MMSE) examinations for at least one year. Results: The single analytes showed statistically significant differences between the clinical groups, but the ratios of analytes indeed had a higher diagnostic performance. Furthermore, only the ratio of CSF neurogranin trunc P75/BACE1 was significantly correlated with the yearly decline in MMSE scores in patients with MCI and dementia due to AD, pointing toward the prognostic value of the ratio. Conclusion: This is the first study demonstrating that the CSF neurogranin trunc P75/BACE1 ratio, reflecting postsynaptic/presynaptic integrity, is related to cognitive decline.
Collapse
Affiliation(s)
- Ann De Vos
- ADx NeuroSciences NV, Technologiepark Zwijnaarde 4, 9052 Gent, Belgium
| | - Hanne Struyfs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Dirk Jacobs
- ADx NeuroSciences NV, Technologiepark Zwijnaarde 4, 9052 Gent, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Tom Klewansky
- Department of Neuropathology, Pitié-Salpêtrière Hospital, Paris, France
| | - Ellen De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Developmental and Lifespan Psychology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Caroline Robberecht
- Neurodegenerative Brain Diseases Group, VIB Department of Molecular Genetics, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB Department of Molecular Genetics, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | | |
Collapse
|
60
|
Mossakowska-Wójcik J, Orzechowska A, Talarowska M, Szemraj J, Gałecki P. The importance of TCF4 gene in the etiology of recurrent depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:304-308. [PMID: 28341444 DOI: 10.1016/j.pnpbp.2017.03.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/13/2017] [Accepted: 03/20/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND A recurrent depressive disorder is one of the most commonly diagnosed disease entities among psychiatric disorders. The prevalence and morbidity of depression are constantly increasing. Numerous studies have demonstrated the role of genetic factors in the etiology of depressive disorders. Many studies are being conducted to identify genes that predispose to depression. The purpose of this study was to investigate the role of TCF4 gene in the etiology of recurrent depressive disorders and, in particular, to assess expression of the TCF4 gene at the mRNA and protein level in patients with recurrent depressive disorders versus healthy individuals. MATERIAL AND METHODS The examined population consisted of 170 individuals suffering from depression and 90 healthy individuals. The expressions of the TCF4 gene at the mRNA and protein level were assessed. RESULTS Decreased TCF4 expression at the mRNA and protein level was found in patients with depressive disorder versus healthy individuals. Expression of the studied gene was not affected by the patients' sex and age. The statistical analysis also showed no correlation between the expression of TCF4 at the mRNA and protein level and the number of episodes or the severity of symptoms. Among the clinical manifestations of depression, only the duration of the illness correlated with the expression of TCF4 at the mRNA level. CONCLUSIONS Expression of TCF4 at the mRNA and protein level may be significant in the pathomechanism of recurrent depressive disorder and it is not dependent on sex and age.
Collapse
Affiliation(s)
- Joanna Mossakowska-Wójcik
- Department of Adult Psychiatry, Medical University of Lodz, Aleksandrowska 159, Lodz 91-229, Poland.
| | - Agata Orzechowska
- Department of Adult Psychiatry, Medical University of Lodz, Aleksandrowska 159, Lodz 91-229, Poland
| | - Monika Talarowska
- Department of Adult Psychiatry, Medical University of Lodz, Aleksandrowska 159, Lodz 91-229, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Czechoslowacka 8/10, 92-216, Lodz, Poland
| | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Lodz, Aleksandrowska 159, Lodz 91-229, Poland
| |
Collapse
|
61
|
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: 0.9] [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
|
62
|
Romano DR, Pharris MC, Patel NM, Kinzer-Ursem TL. Competitive tuning: Competition's role in setting the frequency-dependence of Ca2+-dependent proteins. PLoS Comput Biol 2017; 13:e1005820. [PMID: 29107982 PMCID: PMC5690689 DOI: 10.1371/journal.pcbi.1005820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 11/16/2017] [Accepted: 10/13/2017] [Indexed: 01/20/2023] Open
Abstract
A number of neurological disorders arise from perturbations in biochemical signaling and protein complex formation within neurons. Normally, proteins form networks that when activated produce persistent changes in a synapse’s molecular composition. In hippocampal neurons, calcium ion (Ca2+) flux through N-methyl-D-aspartate (NMDA) receptors activates Ca2+/calmodulin signal transduction networks that either increase or decrease the strength of the neuronal synapse, phenomena known as long-term potentiation (LTP) or long-term depression (LTD), respectively. The calcium-sensor calmodulin (CaM) acts as a common activator of the networks responsible for both LTP and LTD. This is possible, in part, because CaM binding proteins are “tuned” to different Ca2+ flux signals by their unique binding and activation dynamics. Computational modeling is used to describe the binding and activation dynamics of Ca2+/CaM signal transduction and can be used to guide focused experimental studies. Although CaM binds over 100 proteins, practical limitations cause many models to include only one or two CaM-activated proteins. In this work, we view Ca2+/CaM as a limiting resource in the signal transduction pathway owing to its low abundance relative to its binding partners. With this view, we investigate the effect of competitive binding on the dynamics of CaM binding partner activation. Using an explicit model of Ca2+, CaM, and seven highly-expressed hippocampal CaM binding proteins, we find that competition for CaM binding serves as a tuning mechanism: the presence of competitors shifts and sharpens the Ca2+ frequency-dependence of CaM binding proteins. Notably, we find that simulated competition may be sufficient to recreate the in vivo frequency dependence of the CaM-dependent phosphatase calcineurin. Additionally, competition alone (without feedback mechanisms or spatial parameters) could replicate counter-intuitive experimental observations of decreased activation of Ca2+/CaM-dependent protein kinase II in knockout models of neurogranin. We conclude that competitive tuning could be an important dynamic process underlying synaptic plasticity. Learning and memory formation are likely associated with dynamic fluctuations in the connective strength of neuronal synapses. These fluctuations, called synaptic plasticity, are regulated by calcium ion (Ca2+) influx through ion channels localized to the post-synaptic membrane. Within the post-synapse, the dominant Ca2+ sensor protein, calmodulin (CaM), may activate a variety of downstream binding partners, each contributing to synaptic plasticity outcomes. The conditions at which certain binding partners most strongly activate are increasingly studied using computational models. Nearly all computational studies describe these binding partners in combinations of only one or two CaM binding proteins. In contrast, we combine seven well-studied CaM binding partners into a single model wherein they simultaneously compete for access to CaM. Our dynamic model suggests that competition narrows the window of conditions for optimal activation of some binding partners, mimicking the Ca2+-frequency dependence of some proteins in vivo. Further characterization of CaM-dependent signaling dynamics in neuronal synapses may benefit our understanding of learning and memory formation. Furthermore, we propose that competitive binding may be another framework, alongside feedback and feed-forward loops, signaling motifs, and spatial localization, that can be applied to other signal transduction networks, particularly second messenger cascades, to explain the dynamical behavior of protein activation.
Collapse
Affiliation(s)
- Daniel R. Romano
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Matthew C. Pharris
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Neal M. Patel
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Tamara L. Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
- * E-mail:
| |
Collapse
|
63
|
Casaletto KB, Elahi FM, Bettcher BM, Neuhaus J, Bendlin BB, Asthana S, Johnson SC, Yaffe K, Carlsson C, Blennow K, Zetterberg H, Kramer JH. Neurogranin, a synaptic protein, is associated with memory independent of Alzheimer biomarkers. Neurology 2017; 89:1782-1788. [PMID: 28939668 DOI: 10.1212/wnl.0000000000004569] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To determine the association between synaptic functioning as measured via neurogranin in CSF and cognition relative to established Alzheimer disease (AD) biomarkers in neurologically healthy older adults. METHODS We analyzed CSF concentrations of neurogranin, β-amyloid (Aβ42), phosphorylated tau (p-tau), and total tau (t-tau) among 132 neurologically normal older adults (mean 64.5, range 55-85), along with bilateral hippocampal volumes and a measure of episodic memory (Auditory Verbal Learning Test, delayed recall). Univariable analyses examined the relationship between neurogranin and the other AD-related biomarkers. Multivariable regression models examined the relationship between neurogranin and delayed recall, adjusting for age and sex, and interaction terms (neurogranin × AD biomarkers). RESULTS Higher neurogranin concentrations were associated with older age (ρ = 0.20, p = 0.02), lower levels of p-tau and t-tau, and smaller hippocampal volumes (p < 0.03), but not with CSF Aβ42 (p = 0.18). In addition, CSF neurogranin demonstrated a significant relationship with memory performance independent of the AD-related biomarkers; individuals with the lowest CSF neurogranin concentrations performed better on delayed recall than those with medium or high CSF neurogranin concentrations (p < 0.01). Notably, CSF p-tau, t-tau, and Aβ42 and hippocampal volumes were not significantly associated with delayed recall scores (p > 0.40), and did not interact with neurogranin to predict memory (p > 0.10). CONCLUSIONS Synaptic dysfunction (assessed via neurogranin) may be an early pathologic process in age-related neurodegeneration, and a sensitive marker of age-related cognitive abilities, potentially preceding or even acting independently from AD pathogenesis. Synaptic functioning may be a useful early marker of cognitive aging and possibly a target for future brain aging interventions.
Collapse
Affiliation(s)
- Kaitlin B Casaletto
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| | - Fanny M Elahi
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Brianne M Bettcher
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - John Neuhaus
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Barbara B Bendlin
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Sanjay Asthana
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Sterling C Johnson
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kristine Yaffe
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Cynthia Carlsson
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Henrik Zetterberg
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Joel H Kramer
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
64
|
Lista S, Toschi N, Baldacci F, Zetterberg H, Blennow K, Kilimann I, Teipel SJ, Cavedo E, dos Santos AM, Epelbaum S, Lamari F, Dubois B, Nisticò R, Floris R, Garaci F, Hampel H. Cerebrospinal Fluid Neurogranin as a Biomarker of Neurodegenerative Diseases: A Cross-Sectional Study. J Alzheimers Dis 2017; 59:1327-1334. [DOI: 10.3233/jad-170368] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Filippo Baldacci
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, Sweden
| | - Ingo Kilimann
- Department of Psychosomatic Medicine, University of Rostock & DZNE Rostock, Rostock, Germany
| | - Stefan J. Teipel
- Department of Psychosomatic Medicine, University of Rostock & DZNE Rostock, Rostock, Germany
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
- IRCCS “San Giovanni di Dio-Fatebenefratelli”, Brescia, Italy
| | - Antonio Melo dos Santos
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de laMaladie d’Alzheimer (IM2A), HôpitalPitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Stéphane Epelbaum
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de laMaladie d’Alzheimer (IM2A), HôpitalPitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Foudil Lamari
- AP-HP, UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Bruno Dubois
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de laMaladie d’Alzheimer (IM2A), HôpitalPitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Robert Nisticò
- Department of Biology, University ofRome “Tor Vergata” & Pharmacology of Synaptic Disease Lab, European Brain Research Institute (E.B.R.I.), Rome, Italy
| | - Roberto Floris
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Casa di Cura “San Raffaele Cassino”, Cassino, Italy
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | | |
Collapse
|
65
|
Gomis-Perez C, Soldovieri MV, Malo C, Ambrosino P, Taglialatela M, Areso P, Villarroel A. Differential Regulation of PI(4,5)P 2 Sensitivity of Kv7.2 and Kv7.3 Channels by Calmodulin. Front Mol Neurosci 2017; 10:117. [PMID: 28507506 PMCID: PMC5410570 DOI: 10.3389/fnmol.2017.00117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 11/19/2022] Open
Abstract
HIGHLIGHTS- Calmodulin-dependent Kv7.2 current density without the need of binding calcium. - Kv7.2 current density increase is accompanied with resistance to PI(4,5)P2 depletion. - Kv7.3 current density is insensitive to calmodulin elevation. - Kv7.3 is more sensitive to PI(4,5)P2 depletion in the presence of calmodulin. - Apo-calmodulin influences PI(4,5)P2 dependence in a subunit specific manner.
The identification and understanding of critical factors regulating M-current functional density, whose main components are Kv7.2 and Kv7.3 subunits, has profound pathophysiological impact given the important role of the M-current in neuronal excitability control. We report the increase in current density of Kv7.2 channels by calmodulin (CaM) and by a mutant CaM unable to bind Ca2+ (CaM1234) revealing that this potentiation is calcium independent. Furthermore, after co-expressing a CaM binding protein (CaM sponge) to reduce CaM cellular availability, Kv7.2 current density was reduced. Current inhibition after transient depletion of the essential Kv7 co-factor phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) by activating Danio rerio voltage sensitive phosphatase (DrVSP) was blunted by co-expressing CaM1234 or the CaM sponge. In addition, CaM-dependent potentiation was occluded by tonic elevation of PI(4,5)P2 levels by PI(4)P5-kinase (PIP5K) expression. In contrast to the effect on homomeric Kv7.2 channels, CaM1234 failed to potentiate heteromeric Kv7.2/3 or homomeric Kv7.3 channels. Sensitivity to PI(4,5)P2 depletion of Kv7.2/3 channels was increased after expression of CaM1234 or the CaM sponge, while that of homomeric Kv7.3 was unaltered. Altogether, the data reveal that apo-CaM influences PI(4,5)P2 dependence of Kv7.2, Kv7.2/3, and of Kv7.3 channels in a subunit specific manner.
Collapse
Affiliation(s)
- Carolina Gomis-Perez
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| | - Maria V Soldovieri
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Covadonga Malo
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| | - Paolo Ambrosino
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Maurizio Taglialatela
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy.,Department of Neuroscience, University of Naples "Federico II,"Naples, Italy
| | - Pilar Areso
- Department Farmacología, UPV/EHU, Universidad del País VascoLeioa, Spain
| | - Alvaro Villarroel
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| |
Collapse
|
66
|
Seeger C, Talibov VO, Danielson UH. Biophysical analysis of the dynamics of calmodulin interactions with neurogranin and Ca 2+ /calmodulin-dependent kinase II. J Mol Recognit 2017; 30. [PMID: 28449373 PMCID: PMC5518211 DOI: 10.1002/jmr.2621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/26/2016] [Accepted: 02/06/2017] [Indexed: 01/17/2023]
Abstract
Calmodulin (CaM) functions depend on interactions with CaM‐binding proteins, regulated by
Ca2+. Induced structural changes influence the affinity, kinetics, and specificities of the interactions. The dynamics of CaM interactions with neurogranin (Ng) and the CaM‐binding region of
Ca2+/calmodulin‐dependent kinase II (CaMKII290−309) have been studied using biophysical methods. These proteins have opposite
Ca2+ dependencies for CaM binding. Surface plasmon resonance biosensor analysis confirmed that
Ca2+ and CaM interact very rapidly, and with moderate affinity (
KDSPR=3μM). Calmodulin‐CaMKII290−309 interactions were only detected in the presence of
Ca2+, exhibiting fast kinetics and nanomolar affinity (
KDSPR=7.1nM). The CaM–Ng interaction had higher affinity under
Ca2+‐depleted (
KDSPR=480nM,k1=3.4×105M−1s−1 and k−1 = 1.6 × 10−1s−1) than
Ca2+‐saturated conditions (
KDSPR=19μM). The IQ motif of Ng (Ng27−50) had similar affinity for CaM as Ng under
Ca2+‐saturated conditions (
KDSPR=14μM), but no interaction was seen under
Ca2+‐depleted conditions. Microscale thermophoresis using fluorescently labeled CaM confirmed the surface plasmon resonance results qualitatively, but estimated lower affinities for the Ng (
KDMST=890nM) and CaMKII290−309(
KDMST=190nM) interactions. Although CaMKII290−309 showed expected interaction characteristics, they may be different for full‐length CaMKII. The data for full‐length Ng, but not Ng27−50, agree with the current model on Ng regulation of
Ca2+/CaM signaling.
Collapse
Affiliation(s)
- Christian Seeger
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden.,Beactica AB, Uppsala, Sweden
| | | | - U Helena Danielson
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden.,Beactica AB, Uppsala, Sweden.,Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
67
|
Salas IH, Callaerts-Vegh Z, Arranz AM, Guix FX, D’Hooge R, Esteban JA, De Strooper B, Dotti CG. Tetraspanin 6: A novel regulator of hippocampal synaptic transmission and long term plasticity. PLoS One 2017; 12:e0171968. [PMID: 28207852 PMCID: PMC5312877 DOI: 10.1371/journal.pone.0171968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/27/2017] [Indexed: 11/19/2022] Open
Abstract
Tetraspanins (Tspan) are transmembrane proteins with important scaffold and signalling functions. Deletions of Tetraspanin 6 (Tspan6) gene, a member of the tetraspanin family, have been reported in patients with Epilepsy Female-restricted with Mental Retardation (EFMR). Interestingly, mutations in Tspan7, highly homologous to Tspan6, are associated with X-linked intellectual disability, suggesting that these two proteins are important for cognition. Considering recent evidences showing that Tspan7 plays a key role in synapse development and AMPAR trafficking, we initiated the study of Tspan6 in synaptic function using a Tspan6 knock out mouse model. Here we report that hippocampal field recordings from Tspan6 knock out mice show an enhanced basal synaptic transmission and impaired long term potentiation (LTP). A normal paired-pulse facilitation response suggests that Tspan6 affects the properties of the postsynaptic rather than the presynaptic terminal. However, no changes in spine morphology or postsynaptic markers could be detected in Tspan6 KO mice compared with wild types. In addition, Tspan6 KO mice show normal locomotor behaviour and no defects in hippocampus-dependent memory tests.
Collapse
Affiliation(s)
- Isabel H. Salas
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | | | - Amaia M. Arranz
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Francesc X. Guix
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Rudi D’Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - José A. Esteban
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
| | - Bart De Strooper
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
- * E-mail: (CGD); (BDS)
| | - Carlos G. Dotti
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
- * E-mail: (CGD); (BDS)
| |
Collapse
|
68
|
Lista S, Hampel H. Synaptic degeneration and neurogranin in the pathophysiology of Alzheimer’s disease. Expert Rev Neurother 2016; 17:47-57. [DOI: 10.1080/14737175.2016.1204234] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Simone Lista
- AXA Research Fund & UPMC Chair, Paris, France
- IHU-A-ICM – Paris Institute of Translational Neurosciences, Pitié-Salpêtrière University Hospital, Paris, France
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Paris, France
- Department of Neurology, Sorbonne Universities, Institute of Memory and Alzheimer’s Disease (IM2A) & Brain and Spine Institute (ICM) UMR S 1127, Pitié-Salpêtrière University Hospital, Pierre and Marie Curie University, Paris 06, Paris, France
| |
Collapse
|
69
|
Kim H, Kang H, Heo RW, Jeon BT, Yi CO, Shin HJ, Kim J, Jeong SY, Kwak W, Kim WH, Kang SS, Roh GS. Caloric restriction improves diabetes-induced cognitive deficits by attenuating neurogranin-associated calcium signaling in high-fat diet-fed mice. J Cereb Blood Flow Metab 2016; 36:1098-110. [PMID: 26661177 PMCID: PMC4908619 DOI: 10.1177/0271678x15606724] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
Diabetes-induced cognitive decline has been recognized in human patients of type 2 diabetes mellitus and mouse model of obesity, but the underlying mechanisms or therapeutic targets are not clearly identified. We investigated the effect of caloric restriction on diabetes-induced memory deficits and searched a molecular mechanism of caloric restriction-mediated neuroprotection. C57BL/6 mice were fed a high-fat diet for 40 weeks and RNA-seq analysis was performed in the hippocampus of high-fat diet-fed mice. To investigate caloric restriction effect on differential expression of genes, mice were fed high-fat diet for 20 weeks and continued on high-fat diet or subjected to caloric restriction (2 g/day) for 12 weeks. High-fat diet-fed mice exhibited insulin resistance, glial activation, blood-brain barrier leakage, and memory deficits, in that we identified neurogranin, a down-regulated gene in high-fat diet-fed mice using RNA-seq analysis; neurogranin regulates Ca(2+)/calmodulin-dependent synaptic function. Caloric restriction increased insulin sensitivity, reduced high-fat diet-induced blood-brain barrier leakage and glial activation, and improved memory deficit. Furthermore, caloric restriction reversed high-fat diet-induced expression of neurogranin and the activation of Ca(2+)/calmodulin-dependent protein kinase II and calpain as well as the downstream effectors. Our results suggest that neurogranin is an important factor of high-fat diet-induced memory deficits on which caloric restriction has a therapeutic effect by regulating neurogranin-associated calcium signaling.
Collapse
Affiliation(s)
- Hwajin Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Heeyoung Kang
- Department of Neurology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Rok Won Heo
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Byeong Tak Jeon
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, USA
| | - Chin-Ok Yi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Hyun Joo Shin
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Jeonghyun Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seon-Yong Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | - Won-Ho Kim
- Division of Metabolic Diseases, Center for Biomedical Sciences, National Institute of Health, Osong, Republic of Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| |
Collapse
|
70
|
Dunnick JK, Merrick BA, Brix A, Morgan DL, Gerrish K, Wang Y, Flake G, Foley J, Shockley KR. Molecular Changes in the Nasal Cavity after N, N-dimethyl-p-toluidine Exposure. Toxicol Pathol 2016; 44:835-47. [PMID: 27099258 DOI: 10.1177/0192623316637708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
N, N-dimethyl-p-toluidine (DMPT; Cas No. 99-97-8), an accelerant for methyl methacrylate monomers in medical devices, is a nasal cavity carcinogen according to a 2-yr cancer study of male and female F344/N rats, with the nasal tumors arising from the transitional cell epithelium. In this study, we exposed male F344/N rats for 5 days to DMPT (0, 1, 6, 20, 60, or 120 mg/kg [oral gavage]) to explore the early changes in the nasal cavity after short-term exposure. Lesions occurred in the nasal cavity including hyperplasia of transitional cell epithelium (60 and 120 mg/kg). Nasal tissue was rapidly removed and preserved for subsequent laser capture microdissection and isolation of the transitional cell epithelium (0 and 120 mg/kg) for transcriptomic studies. DMPT transitional cell epithelium gene transcript patterns were characteristic of an antioxidative damage response (e.g., Akr7a3, Maff, and Mgst3), cell proliferation, and decrease in signals for apoptosis. The transcripts of amino acid transporters were upregulated (e.g., Slc7a11). The DMPT nasal transcript expression pattern was similar to that found in the rat nasal cavity after formaldehyde exposure, with over 1,000 transcripts in common. Molecular changes in the nasal cavity after DMPT exposure suggest that oxidative damage is a mechanism of the DMPT toxic and/or carcinogenic effects.
Collapse
Affiliation(s)
- June K Dunnick
- Toxicology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - B Alex Merrick
- Biomolecular Screening Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Amy Brix
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | - Daniel L Morgan
- NTP Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kevin Gerrish
- Molecular Genomics Core, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Yu Wang
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Gordon Flake
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Julie Foley
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Keith R Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
71
|
Brown JC, Petersen A, Zhong L, Himelright ML, Murphy JA, Walikonis RS, Gerges NZ. Bidirectional regulation of synaptic transmission by BRAG1/IQSEC2 and its requirement in long-term depression. Nat Commun 2016; 7:11080. [PMID: 27009485 PMCID: PMC4820844 DOI: 10.1038/ncomms11080] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/16/2016] [Indexed: 01/09/2023] Open
Abstract
Dysfunction of the proteins regulating synaptic function can cause synaptic plasticity imbalance that underlies neurological disorders such as intellectual disability. A study found that four distinct mutations within BRAG1, an Arf-GEF synaptic protein, each led to X-chromosome-linked intellectual disability (XLID). Although the physiological functions of BRAG1 are poorly understood, each of these mutations reduces BRAG1's Arf-GEF activity. Here we show that BRAG1 is required for the activity-dependent removal of AMPA receptors in rat hippocampal pyramidal neurons. Moreover, we show that BRAG1 bidirectionally regulates synaptic transmission. On one hand, BRAG1 is required for the maintenance of synaptic transmission. On the other hand, BRAG1 expression enhances synaptic transmission, independently of BRAG1 Arf-GEF activity or neuronal activity, but dependently on its C-terminus interactions. This study demonstrates a dual role of BRAG1 in synaptic function and highlights the functional relevance of reduced BRAG1 Arf-GEF activity as seen in the XLID-associated human mutations.
Collapse
Affiliation(s)
- Joshua C Brown
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Amber Petersen
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Ling Zhong
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Miranda L Himelright
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Jessica A Murphy
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Randall S Walikonis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| |
Collapse
|
72
|
Wellington H, Paterson RW, Portelius E, Törnqvist U, Magdalinou N, Fox NC, Blennow K, Schott JM, Zetterberg H. Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology 2016; 86:829-35. [PMID: 26826204 DOI: 10.1212/wnl.0000000000002423] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/02/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the specificity of the dendritic protein neurogranin (Ng) in CSF from patients with a broad range of neurodegenerative diseases including a variety of dementias, tauopathies, and synucleinopathies. METHOD An optimized immunoassay was used to analyze CSF Ng in a retrospective cohort of 331 participants with different neurodegenerative diseases, including healthy controls (n = 19), biomarker-proven Alzheimer disease (AD) (n = 100), genetic AD (n = 2), behavioral variant frontotemporal dementia (n = 20), speech variant frontotemporal dementia (n = 21), Lewy body dementia (n = 13), Parkinson disease (n = 31), progressive supranuclear palsy (n = 46), multiple system atrophy (n = 29), as well as a heterogeneous group with non-neurodegenerative cognitive impairment (n = 50). CSF Ng concentrations and correlations of CSF Ng with total tau, phosphorylated tau, and β-amyloid 42 concentrations, Mini-Mental State Examination score, and disease duration in the different groups were investigated. RESULTS Median CSF Ng concentration was higher in patients with AD compared to both controls (p < 0.001) and all other disease groups (all p < 0.001) except speech variant frontotemporal dementia. There were no significant differences in CSF Ng concentrations between any other neurodegenerative groups and controls. In addition, we found strong correlations between Ng and total tau (p < 0.001) and phosphorylated tau (p < 0.001). CONCLUSIONS These results confirm an increase in CSF Ng concentration in patients with AD as previously reported and show that this is specific to AD and not seen in a range of other neurodegenerative diseases.
Collapse
Affiliation(s)
- Henrietta Wellington
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Ross W Paterson
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erik Portelius
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Ulrika Törnqvist
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nadia Magdalinou
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nick C Fox
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan M Schott
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- From the Department of Molecular Neuroscience (H.W., H.Z.), UCL Institute of Neurology, Queen Square, London; Dementia Research Centre (H.W., R.W.P., N.M., N.C.F., J.M.S.), UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK; and Clinical Neurochemistry Lab (E.P., U.T., K.B., H.Z.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
73
|
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: 2.1] [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
|
74
|
Maruani A, Huguet G, Beggiato A, ElMaleh M, Toro R, Leblond CS, Mathieu A, Amsellem F, Lemière N, Verloes A, Leboyer M, Gillberg C, Bourgeron T, Delorme R. 11q24.2-25 micro-rearrangements in autism spectrum disorders: Relation to brain structures. Am J Med Genet A 2015; 167A:3019-30. [DOI: 10.1002/ajmg.a.37345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 08/12/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Anna Maruani
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Guillaume Huguet
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Anita Beggiato
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Monique ElMaleh
- Department of Radiology; Robert Debré Hospital, APHP; Paris France
| | - Roberto Toro
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Claire S. Leblond
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alexandre Mathieu
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | | | - Nathalie Lemière
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alain Verloes
- Department of Human Genetics; Robert Debré Hospital, APHP; Paris France
| | - Marion Leboyer
- INSERM U955, Team 15; Faculty of Medicine; Creteil France
- Department of Adult Psychiatry; Henri Mondor-Albert Chenevier Hospitals AP-HP; Creteil France
- Fondation FondaMental; French National Science Foundation; Creteil France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre; Gothenburg University; Göteborg Sweden
- Saint George's Hospital Medical School; London United Kingdom
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Diderot Paris 7; University Paris; Paris France
| | - Richard Delorme
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
- Fondation FondaMental; French National Science Foundation; Creteil France
| |
Collapse
|
75
|
Abstract
Increasing plasticity in neurons of the prefrontal cortex (PFC) has been proposed as a possible therapeutic tool to enhance extinction, a process that is impaired in post-traumatic stress disorder, schizophrenia, and addiction. To test this hypothesis, we generated transgenic mice that overexpress neurogranin (a calmodulin-binding protein that facilitates long-term potentiation) in the PFC. Neurogranin overexpression in the PFC enhanced long-term potentiation and increased the rates of extinction learning of both fear conditioning and sucrose self-administration. Our results indicate that elevated neurogranin function within the PFC can enhance local plasticity and increase the rate of extinction learning across different behavioral tasks. Thus, neurogranin can provide a molecular link between enhanced plasticity and enhanced extinction.
Collapse
|
76
|
Han X, Shao W, Liu Z, Fan S, Yu J, Chen J, Qiao R, Zhou J, Xie P. iTRAQ-based quantitative analysis of hippocampal postsynaptic density-associated proteins in a rat chronic mild stress model of depression. Neuroscience 2015; 298:220-92. [DOI: 10.1016/j.neuroscience.2015.04.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/20/2015] [Accepted: 04/02/2015] [Indexed: 01/26/2023]
|
77
|
Neurogranin regulates CaM dynamics at dendritic spines. Sci Rep 2015; 5:11135. [PMID: 26084473 PMCID: PMC4471661 DOI: 10.1038/srep11135] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
Calmodulin (CaM) plays a key role in synaptic function and plasticity due to its ability to mediate Ca2+ signaling. Therefore, it is essential to understand the dynamics of CaM at dendritic spines. In this study we have explored CaM dynamics using live-cell confocal microscopy and fluorescence recovery after photobleaching (FRAP) to study CaM diffusion. We find that only a small fraction of CaM in dendritic spines is immobile. Furthermore, the diffusion rate of CaM was regulated by neurogranin (Ng), a CaM-binding protein enriched at dendritic spines. Interestingly, Ng did not influence the immobile fraction of CaM at recovery plateau. We have previously shown that Ng enhances synaptic strength in a CaM-dependent manner. Taken together, these data indicate that Ng-mediated enhancement of synaptic strength is due to its ability to target, rather than sequester, CaM within dendritic spines.
Collapse
|
78
|
Martin MV, Mirnics K, Nisenbaum LK, Vawter MP. Olanzapine Reversed Brain Gene Expression Changes Induced by Phencyclidine Treatment in Non-Human Primates. MOLECULAR NEUROPSYCHIATRY 2015; 1:82-93. [PMID: 26405684 DOI: 10.1159/000430786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The NMDA receptor antagonist phencyclidine (PCP) creates schizophrenia-like symptoms in normal controls. The effect of PCP on non-human primate brain gene expression was examined and compared to changes induced by olanzapine treatment. Experimental studies of PCP and antipsychotic drugs have direct relevance to understanding the patho-physiology and treatment of schizophrenia. Genome-wide changes in prefrontal cortex gene expression revealed alterations of 146 transcripts in the PCP treatment group compared to vehicle controls. Dysregulated genes were enriched in identified classes implicated in neurological and genetic disorders, including schizophrenia genes from the Psychiatric Genomics Consortium 108 loci as well as cell death in PCP-treated primates. Canonical pathway analysis revealed a significant overrepresentation of several groups including synaptic long-term potentiation and calcium signaling. Olanzapine coadministered with PCP normalized 34% of the 146 PCP-induced probe set expression changes, and a network of 17 olanzapine-normalized genes was identified enriched in schizophrenia candidate genes containing RGS4, SYN1 and AKT as nodes. The results of this study support the use of PCP administration in non-human primates as a glutamatergic model of schizophrenia and suggest that a large number of PCP-induced expression differences can be reversed by olanzapine. The results of this study may be informative for identification of potential candidates for pharmacogenetics and biomarker research related to the treatment of schizophrenia.
Collapse
Affiliation(s)
- Maureen V Martin
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, Calif
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, Tenn
| | - Laura K Nisenbaum
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Ind., USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, Calif
| |
Collapse
|
79
|
Davis RVN, Lamont SJ, Rothschild MF, Persia ME, Ashwell CM, Schmidt CJ. Transcriptome analysis of post-hatch breast muscle in legacy and modern broiler chickens reveals enrichment of several regulators of myogenic growth. PLoS One 2015; 10:e0122525. [PMID: 25821972 PMCID: PMC4379050 DOI: 10.1371/journal.pone.0122525] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 02/21/2015] [Indexed: 11/19/2022] Open
Abstract
Agriculture provides excellent model systems for understanding how selective pressure, as applied by humans, can affect the genomes of plants and animals. One such system is modern poultry breeding in which intensive genetic selection has been applied for meat production in the domesticated chicken. As a result, modern meat-type chickens (broilers) exhibit enhanced growth, especially of the skeletal muscle, relative to their legacy counterparts. Comparative studies of modern and legacy broiler chickens provide an opportunity to identify genes and pathways affected by this human-directed evolution. This study used RNA-seq to compare the transcriptomes of a modern and a legacy broiler line to identify differentially enriched genes in the breast muscle at days 6 and 21 post-hatch. Among the 15,945 genes analyzed, 10,841 were expressed at greater than 0.1 RPKM. At day 6 post-hatch 189 genes, including several regulators of myogenic growth and development, were differentially enriched between the two lines. The transcriptional profiles between lines at day 21 post-hatch identify 193 genes differentially enriched and still include genes associated with myogenic growth. This study identified differentially enriched genes that regulate myogenic growth and differentiation between the modern and legacy broiler lines. Specifically, differences in the ratios of several positive (IGF1, IGF1R, WFIKKN2) and negative (MSTN, ACE) myogenic growth regulators may help explain the differences underlying the enhanced growth characteristics of the modern broilers.
Collapse
Affiliation(s)
- Richard V. N. Davis
- Dept. Biological Sciences, University of Delaware, Newark, Delaware, 19716, United States of America
| | - Susan J. Lamont
- Dept. of Animal Science, Iowa State University, Ames, Iowa, 50011, United States of America
| | - Max F. Rothschild
- Dept. of Animal Science, Iowa State University, Ames, Iowa, 50011, United States of America
| | - Michael E. Persia
- Dept. of Animal Science, Iowa State University, Ames, Iowa, 50011, United States of America
| | - Chris M. Ashwell
- Dept. of Poultry Science, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Carl J. Schmidt
- Dept. of Animal and Food Sciences, University of Delaware, Newark, Delaware, 19716, United States of America
- * E-mail:
| |
Collapse
|
80
|
Abstract
Schizophrenia is a common mental disorder, affecting 0.5-1% of the population. The mode of inheritance is complex and non-Mendelian with a high heritability of ca. 65-80%. Given this complexity, until most recently it was difficult to identify disease genes. But fortunately this has changed. Due to new technologies the last few years have brought highest interest in human genetics of complex diseases. The knowledge resulting from the availability of the complete sequence of the human genome, the systematic identification of single nucleotide polymorphisms (SNPs) throughout the genome, and the development of parallel genotyping technology (microarrays) established the conditions that brought about the current successful time in our ability to probe the genome for identifying disease genes. All these studies showed up new avenues for the biology of common complex diseases and yielded a multitude of genes showing strong association with complex diseases.
Collapse
|
81
|
Ryoo K, Hwang SG, Kim KJ, Choi EJ. RC3/neurogranin negatively regulates extracellular signal-regulated kinase pathway through its interaction with Ras. Mol Cell Biochem 2014; 402:33-40. [DOI: 10.1007/s11010-014-2311-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/20/2014] [Indexed: 12/19/2022]
|
82
|
Abstract
Today, psychiatrists are focusing on genetics aspects of various psychiatric disorders not only for a future classification of psychiatric disorders but also a notion that genetics would aid in the development of new medications to treat these disabling illnesses. This review therefore emphasizes on the basics of genetics in psychiatry as well as focuses on the emerging picture of genetics in psychiatry and their future implications.
Collapse
Affiliation(s)
- Shreekantiah Umesh
- Department of Psychiatry, K.S. Mani Centre for Cognitive Neurosciences, Kanke, Ranchi, Jharkhand, India
| | | |
Collapse
|
83
|
The Ever Changing Moods of Calmodulin: How Structural Plasticity Entails Transductional Adaptability. J Mol Biol 2014; 426:2717-35. [DOI: 10.1016/j.jmb.2014.05.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 11/20/2022]
|
84
|
Wang B, Zhao J, Yu M, Meng X, Cui X, Zhao Y, Zhu Y, Xing W, Guan Y. Disturbance of intracellular calcium homeostasis and CaMKII/CREB signaling is associated with learning and memory impairments induced by chronic aluminum exposure. Neurotox Res 2014; 26:52-63. [PMID: 24366850 DOI: 10.1007/s12640-013-9451-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/27/2013] [Accepted: 12/07/2013] [Indexed: 10/25/2022]
Abstract
Aluminum-induced neuronal injury has been implicated in various neurodegenerative disorders. However, the underlying mechanism involved in this pathogenesis still remains unknown. Our present findings demonstrated that chronic aluminum exposure resulted in spatial learning impairment and significantly increased intracellular calcium level in the hippocampus of rats. Examination of the associated protein molecules essential for induction and maintenance of long-term potentiation revealed that aluminum exposure could increase the expression level of calmodulin (CaM), but the expression levels of CaM-dependent protein kinase II (CaMKII), and phosphorylated cAMP-responsive element binding protein (CREB) were significantly reduced, whereas the total protein levels of CaMKII and CREB did not change in the aluminum-treated hippocampus. Thus, we provide a previously unrecognized mechanism whereby chronic aluminum exposure impairs hippocampal learning and memory, at least in part, through disruption of intracellular calcium homeostasis and CaM/CaMKII/CREB signaling pathway.
Collapse
Affiliation(s)
- Biao Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, No. 92 Bei'er Road, Heping District, Shenyang, 110001, People's Republic of China,
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Expression of neurogranin in hippocampus of rat offspring exposed to restraint stress and pulsed magnetic fields. Brain Res 2014; 1570:26-34. [DOI: 10.1016/j.brainres.2014.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/28/2014] [Accepted: 05/02/2014] [Indexed: 11/23/2022]
|
86
|
Hell JW. CaMKII: claiming center stage in postsynaptic function and organization. Neuron 2014; 81:249-65. [PMID: 24462093 DOI: 10.1016/j.neuron.2013.12.024] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 11/16/2022]
Abstract
While CaMKII has long been known to be essential for synaptic plasticity and learning, recent work points to new dimensions of CaMKII function in the nervous system, revealing that CaMKII also plays an important role in synaptic organization. Ca(2+)-triggered autophosphorylation of CaMKII not only provides molecular memory by prolonging CaMKII activity during long-term plasticity (LTP) and learning but also represents a mechanism for autoactivation of CaMKII's multifaceted protein-docking functions. New details are also emerging about the distinct roles of CaMKIIα and CaMKIIβ in synaptic homeostasis, further illustrating the multilayered and complex nature of CaMKII's involvement in synaptic regulation. Here, I review novel molecular and functional insight into how CaMKII supports synaptic function.
Collapse
Affiliation(s)
- Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA 95615, USA.
| |
Collapse
|
87
|
Chocyk A, Bobula B, Dudys D, Przyborowska A, Majcher-Maślanka I, Hess G, Wędzony K. Early-life stress affects the structural and functional plasticity of the medial prefrontal cortex in adolescent rats. Eur J Neurosci 2013; 38:2089-107. [PMID: 23581639 DOI: 10.1111/ejn.12208] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 03/03/2013] [Indexed: 02/06/2023]
Abstract
Early life experiences are crucial factors that shape brain development and function due to their ability to induce structural and functional plasticity. Among these experiences, early-life stress (ELS) is known to interfere with brain development and maturation, increasing the risk of future psychopathologies, including depression, anxiety, and personality disorders. Moreover, ELS may contribute to the emergence of these psychopathologies during adolescence. In this present study, we investigated the effects of ELS, in the form of maternal separation (MS), on the structural and functional plasticity of the medial prefrontal cortex (mPFC) and anxiety-like behavior in adolescent male rats. We found that the MS procedure resulted in disturbances in mother-pup interactions that lasted until weaning and were most strongly demonstrated by increases in nursing behavior. Moreover, MS caused atrophy of the basal dendritic tree and reduced spine density on both the apical and basal dendrites in layer II/III pyramidal neurons of the mPFC. The structural changes were accompanied by an impairment of long-term potentiation processes and increased expression of key proteins, specifically glutamate receptor 1, glutamate receptor 2, postsynaptic density protein 95, αCa(2+) /calmodulin-dependent protein kinase II and αCa(2+)/calmodulin-dependent protein kinase II phosphorylated at residue Thr305, that are engaged in long-term potentiation induction and maintenance in the mPFC. We also found that the MS animals were more anxious in the light/dark exploration test. The results of this study indicate that ELS has a significant impact on the structural and functional plasticity of the mPFC in adolescents. ELS-induced adaptive plasticity may underlie the pathomechanisms of some early-onset psychopathologies observed in adolescents.
Collapse
Affiliation(s)
- Agnieszka Chocyk
- Laboratory of Pharmacology and Brain Biostructure, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | | | | | | | | | | | | |
Collapse
|
88
|
Zhong L, Brown JC, Wells C, Gerges NZ. Post-embedding Immunogold labeling of synaptic proteins in hippocampal slice cultures. J Vis Exp 2013. [PMID: 23609099 DOI: 10.3791/50273] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Immunoelectron microscopy is a powerful tool to study biological molecules at the subcellular level. Antibodies coupled to electron-dense markers such as colloidal gold can reveal the localization and distribution of specific antigens in various tissues. The two most widely used techniques are pre-embedding and post-embedding techniques. In pre-embedding immunogold-electron microscopy (EM) techniques, the tissue must be permeabilized to allow antibody penetration before it is embedded. These techniques are ideal for preserving structures but poor penetration of the antibody (often only the first few micrometers) is a considerable drawback. The post-embedding labeling methods can avoid this problem because labeling takes place on sections of fixed tissues where antigens are more easily accessible. Over the years, a number of modifications have improved the post-embedding methods to enhance immunoreactivity and to preserve ultrastructure. Tissue fixation is a crucial part of EM studies. Fixatives chemically crosslink the macromolecules to lock the tissue structures in place. The choice of fixative affects not only structural preservation but also antigenicity and contrast. Osmium tetroxide (OsO4), formaldehyde, and glutaraldehyde have been the standard fixatives for decades, including for central nervous system (CNS) tissues that are especially prone to structural damage during chemical and physical processing. Unfortunately, OsO4 is highly reactive and has been shown to mask antigens, resulting in poor and insufficient labeling. Alternative approaches to avoid chemical fixation include freezing the tissues. But these techniques are difficult to perform and require expensive instrumentation. To address some of these problems and to improve CNS tissue labeling, Phend et al. replaced OsO4 with uranyl acetate (UA) and tannic acid (TA), and successfully introduced additional modifications to improve the sensitivity of antigen detection and structural preservation in brain and spinal cord tissues. We have adopted this osmium-free post-embedding method to rat brain tissue and optimized the immunogold labeling technique to detect and study synaptic proteins. We present here a method to determine the ultrastructural localization of synaptic proteins in rat hippocampal CA1 pyramidal neurons. We use organotypic hippocampal cultured slices. These slices maintain the trisynaptic circuitry of the hippocampus, and thus are especially useful for studying synaptic plasticity, a mechanism widely thought to underlie learning and memory. Organotypic hippocampal slices from postnatal day 5 and 6 mouse/rat pups can be prepared as described previously), and are especially useful to acutely knockdown or overexpress exogenous proteins. We have previously used this protocol to characterize neurogranin (Ng), a neuron-specific protein with a critical role in regulating synaptic function . We have also used it to characterize the ultrastructural localization of calmodulin (CaM) and Ca(2+)/CaM-dependent protein kinase II (CaMKII). As illustrated in the results, this protocol allows good ultrastructural preservation of dendritic spines and efficient labeling of Ng to help characterize its distribution in the spine. Furthermore, the procedure described here can have wide applicability in studying many other proteins involved in neuronal functions.
Collapse
Affiliation(s)
- Ling Zhong
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, USA
| | | | | | | |
Collapse
|
89
|
Gribaudo S, Bovetti S, Friard O, Denorme M, Oboti L, Fasolo A, De Marchis S. Transitory and activity-dependent expression of neurogranin in olfactory bulb tufted cells during mouse postnatal development. J Comp Neurol 2013; 520:3055-69. [PMID: 22592880 DOI: 10.1002/cne.23150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neurogranin (Ng) is a brain-specific postsynaptic calmodulin-binding protein involved in synaptic activity-dependent plasticity. In the adult olfactory bulb (OB), Ng is expressed by a large population of GABAergic interneurons in the granule cell layer. We show here that, during postnatal development, Ng is also expressed by OB neurons in the superficial external plexiform layer (sEPL) and glomerular layer (GL). These Ng-positive neurons display morphological and neurochemical features of superficial and external tufted cells. Ng expression in these cells is transient during OB development: few elements express Ng at postnatal day (P) 5, increasing in number and reaching a peak at P10, then progressively decreasing. At P30, Ng is rarely detectable in these neurons. Ng expression in developing tufted cells is also modulated at the cellular level: at earlier stages, Ng labeling is distributed throughout the cell body and dendritic arborization in the GL, but, at P20, when the glomerular circuits are fully matured, Ng becomes restricted to the soma and proximal portion of tufted cell apical dendrites. We show that olfactory deprivation at early postnatal stages induces a strong increase in Ng-positive tufted cells from P10 to P20, whereas no changes have been observed following olfactory deprivation in adult mice. These findings demonstrate that Ng expression in sEPL-GL is restricted to developmental stages and indicate its activity-dependent regulation in a time window critical for glomerular circuit development, suggesting a role for Ng in maturation and dendritic remodeling of tufted cells.
Collapse
Affiliation(s)
- S Gribaudo
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy.
| | | | | | | | | | | | | |
Collapse
|
90
|
Structural basis for the interaction of unstructured neuron specific substrates neuromodulin and neurogranin with Calmodulin. Sci Rep 2013; 3:1392. [PMID: 23462742 PMCID: PMC3589724 DOI: 10.1038/srep01392] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 02/21/2013] [Indexed: 01/07/2023] Open
Abstract
Neuromodulin (Nm) and neurogranin (Ng) are neuron-specific substrates of protein kinase C (PKC). Their interactions with Calmodulin (CaM) are crucial for learning and memory formation in neurons. Here, we report the structure of IQ peptides (24aa) of Nm/Ng complexed with CaM and their functional studies with full-length proteins. Nm/Ng and their respective IQ peptides are intrinsically unstructured; however, upon binding with CaM, IQ motifs adopt a helical conformation. Ser41 (Ser36) of Nm (Ng) is located in a negatively charged pocket in the apo CaM and, when phosphorylated, it will repel Nm/Ng from CaM. These observations explain the mechanism by which PKC-induced Ser phosphorylation blocks the association of Nm/Ng with CaM and interrupts several learning- and memory-associated functions. Moreover, the present study identified Arg as a key CaM interacting residue from Nm/Ng. This residue is crucial for CaM-mediated function, as evidenced by the inability of the Ng mutant (Arg-to-Ala) to potentiate synaptic transmission in CA1 hippocampal neurons.
Collapse
|
91
|
Krug A, Krach S, Jansen A, Nieratschker V, Witt SH, Shah NJ, Nöthen MM, Rietschel M, Kircher T. The effect of neurogranin on neural correlates of episodic memory encoding and retrieval. Schizophr Bull 2013; 39:141-50. [PMID: 21799211 PMCID: PMC3523918 DOI: 10.1093/schbul/sbr076] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neurogranin (NRGN) is the main postsynaptic protein regulating the availability of calmodulin-Ca(2+) in neurons. NRGN is expressed exclusively in the brain, particularly in dendritic spines and has been implicated in spatial learning and hippocampal plasticity. Genetic variation in rs12807809 in the NRGN gene has recently been confirmed to be associated with schizophrenia in a meta-analysis of genome-wide association studies: the T-allele was found to be genome-wide significantly associated with schizophrenia. Cognitive tests and personality questionnaires were administered in a large sample of healthy subjects. Brain activation was measured with functional magnetic resonance imaging (fMRI) during an episodic memory encoding and retrieval task in a subsample. All subjects were genotyped for NRGN rs12807809. There was no effect of genotype on personality or cognitive measures in the large sample. Homozygote carriers of the T-allele showed better performance in the retrieval task during fMRI. After controlling for memory performance, differential brain activation was evident in the anterior cingulate cortex for the encoding and posterior cingulate regions during retrieval. We could demonstrate that rs12807809 of NRGN is associated with differential neural functioning in the anterior and posterior cingulate. These areas are involved in episodic memory processes and have been implicated in the pathophysiology of schizophrenia in structural and functional imaging as well as postmortem studies.
Collapse
Affiliation(s)
- Axel Krug
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany.
| | - Sören Krach
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany,Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany
| | - Vanessa Nieratschker
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - N. Jon Shah
- Institute of Neuroscience and Biophysics 3—Medicine, Research Center Jülich, Jülich, Germany
| | - Markus M. Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany
| |
Collapse
|
92
|
Alaimo A, Alberdi A, Gomis-Perez C, Fernández-Orth J, Gómez-Posada JC, Areso P, Villarroel A. Cooperativity between calmodulin-binding sites in Kv7.2 channels. J Cell Sci 2012. [PMID: 23203804 DOI: 10.1242/jcs.114082] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Among the multiple roles assigned to calmodulin (CaM), controlling the surface expression of Kv7.2 channels by binding to two discontinuous sites is a unique property of this Ca(2+) binding protein. Mutations that interfere with CaM binding or the sequestering of CaM prevent this M-channel component from exiting the endoplasmic reticulum (ER), which reduces M-current density in hippocampal neurons, enhancing excitability and offering a rational mechanism to explain some forms of benign familial neonatal convulsions (BFNC). Previously, we identified a mutation (S511D) that impedes CaM binding while allowing the channel to exit the ER, hinting that CaM binding may not be strictly required for Kv7.2 channel trafficking to the plasma membrane. Alternatively, this interaction with CaM might escape detection and, indeed, we now show that the S511D mutant contains functional CaM-binding sites that are not detected by classical biochemical techniques. Surface expression and function is rescued by CaM, suggesting that free CaM in HEK293 cells is limiting and reinforcing the hypothesis that CaM binding is required for ER exit. Within the CaM-binding domain formed by two sites (helix A and helix B), we show that CaM binds to helix B with higher apparent affinity than helix A, both in the presence and absence of Ca(2+), and that the two sites cooperate. Hence, CaM can bridge two binding domains, anchoring helix A of one subunit to helix B of another subunit, in this way influencing the function of Kv7.2 channels.
Collapse
Affiliation(s)
- Alessandro Alaimo
- Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, Barrio Sarriena s/n, 48940 Leioa, Spain
| | | | | | | | | | | | | |
Collapse
|
93
|
Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, Lepack A, Majik MS, Jeong LS, Banasr M, Son H, Duman RS. Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med 2012; 18:1413-7. [PMID: 22885997 PMCID: PMC3491115 DOI: 10.1038/nm.2886] [Citation(s) in RCA: 556] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Accepted: 06/23/2012] [Indexed: 01/01/2023]
Abstract
Previous imaging and postmortem studies have reported a lower brain volume and a smaller size and density of neurons in the dorsolateral prefrontal cortex (dlPFC) of subjects with major depressive disorder (MDD). These findings suggest that synapse number and function are decreased in the dlPFC of patients with MDD. However, there has been no direct evidence reported for synapse loss in MDD, and the gene expression alterations underlying these effects have not been identified. Here we use microarray gene profiling and electron microscopic stereology to reveal lower expression of synaptic-function–related genes (CALM2, SYN1, RAB3A, RAB4B and TUBB4) in the dlPFC of subjects with MDD and a corresponding lower number of synapses. We also identify a transcriptional repressor, GATA1, expression of which is higher in MDD and that, when expressed in PFC neurons, is sufficient to decrease the expression of synapse-related genes, cause loss of dendritic spines and dendrites, and produce depressive behavior in rat models of depression.
Collapse
Affiliation(s)
- Hyo Jung Kang
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Zhong L, Gerges NZ. Neurogranin targets calmodulin and lowers the threshold for the induction of long-term potentiation. PLoS One 2012; 7:e41275. [PMID: 22848456 PMCID: PMC3405117 DOI: 10.1371/journal.pone.0041275] [Citation(s) in RCA: 35] [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: 05/29/2012] [Accepted: 06/19/2012] [Indexed: 11/19/2022] Open
Abstract
Calcium entry and the subsequent activation of CaMKII trigger synaptic plasticity in many brain regions. The induction of long-term potentiation (LTP) in the CA1 region of the hippocampus requires a relatively high amount of calcium-calmodulin. This requirement is usually explained, based on in vitro and theoretical studies, by the low affinity of CaMKII for calmodulin. An untested hypothesis, however, is that calmodulin is not randomly distributed within the spine and its targeting within the spine regulates LTP. We have previously shown that overexpression of neurogranin enhances synaptic strength in a calmodulin-dependent manner. Here, using post-embedding immunogold labeling, we show that calmodulin is not randomly distributed, but spatially organized in the spine. Moreover, neurogranin regulates calmodulin distribution such that its overexpression concentrates calmodulin closer to the plasma membrane, where a high level of CaMKII immunogold labeling is also found. Interestingly, the targeting of calmodulin by neurogranin results in lowering the threshold for LTP induction. These findings highlight the significance of calmodulin targeting within the spine in synaptic plasticity.
Collapse
Affiliation(s)
- Ling Zhong
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Nashaat Z. Gerges
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
95
|
Sullivan PF, Daly MJ, O'Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet 2012; 13:537-51. [PMID: 22777127 PMCID: PMC4110909 DOI: 10.1038/nrg3240] [Citation(s) in RCA: 830] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Psychiatric disorders are among the most intractable enigmas in medicine. In the past 5 years, there has been unprecedented progress on the genetics of many of these conditions. In this Review, we discuss the genetics of nine cardinal psychiatric disorders (namely, Alzheimer's disease, attention-deficit hyperactivity disorder, alcohol dependence, anorexia nervosa, autism spectrum disorder, bipolar disorder, major depressive disorder, nicotine dependence and schizophrenia). Empirical approaches have yielded new hypotheses about aetiology and now provide data on the often debated genetic architectures of these conditions, which have implications for future research strategies. Further study using a balanced portfolio of methods to assess multiple forms of genetic variation is likely to yield many additional new findings.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Departments of Genetics and Psychiatry, CB# 7264, 5097 Genomic Medicine, University of North Carolina at Chapel Hill, North Carolina 27599-27264, USA.
| | | | | |
Collapse
|
96
|
Morgan MAJ, Morgan JI. Pcp4l1 contains an auto-inhibitory element that prevents its IQ motif from binding to calmodulin. J Neurochem 2012; 121:843-51. [PMID: 22458599 DOI: 10.1111/j.1471-4159.2012.07745.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Purkinje cell protein 4-like 1 (Pcp4l1) is a small neuronal IQ motif protein closely related to the calmodulin-binding protein Pcp4/PEP-19. PEP-19 interacts with calmodulin via its IQ motif to inhibit calmodulin-dependent enzymes and we hypothesized Pcp4l1 would have similar properties. Surprisingly, full-length Pcp4l1 does not interact with calmodulin in yeast two-hybrid or pulldown experiments yet a synthetic peptide constituting only the IQ motif of Pcp4l1 binds calmodulin and inhibits calmodulin-dependent kinase II. A nine-residue glutamic acid-rich sequence in Pcp4l1 confers these unexpected properties. This element lies outside the IQ motif and its deletion or exchange with the homologous region of PEP-19 restores calmodulin binding. Conversion of a single isoleucine (Ile36) within this motif to phenylalanine, the residue present in PEP-19, imparts calmodulin binding onto Pcp4l1. Moreover, only aromatic amino acid substitutions at position 36 in Pcp4l1 allow binding. Thus, despite their sequence similarities PEP-19 and Pcp4l1 have distinct properties with the latter harboring an element that can functionally suppress an IQ motif. We speculate Pcp4l1 may be a latent calmodulin inhibitor regulated by post-translational modification and/or co-factor interactions.
Collapse
Affiliation(s)
- Marc A J Morgan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | | |
Collapse
|
97
|
Christian DT, Alexander NJ, Diaz MR, Robinson S, McCool BA. Chronic intermittent ethanol and withdrawal differentially modulate basolateral amygdala AMPA-type glutamate receptor function and trafficking. Neuropharmacology 2012; 62:2430-9. [PMID: 22387532 DOI: 10.1016/j.neuropharm.2012.02.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/15/2012] [Accepted: 02/16/2012] [Indexed: 12/19/2022]
Abstract
The amygdala plays a critical role in the generation and expression of anxiety-like behaviors including those expressed following withdrawal (WD) from chronic intermittent ethanol (CIE) exposure. In particular, the BLA glutamatergic system controls the expression of both innate and pathological anxiety. Recent data suggests that CIE and WD may functionally alter this system in a manner that closely parallels memory-related phenomena like long-term potentiation (LTP). We therefore specifically dissected CIE/WD-induced changes in glutamatergic signaling using electrophysiological and biochemical approaches with a particular focus on the plasticity-related components of this neurotransmitter system. Our results indicate that cortical glutamatergic inputs arriving at BLA principal via the external capsule undergo predominantly post-synaptic alterations in AMPA receptor function following CIE and WD. Biochemical analysis revealed treatment-dependent changes in AMPA receptor surface expression and subunit phosphorylation that are complemented by changes in total protein levels and/or phosphorylation status of several key, plasticity-associated protein kinases such as calcium/calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC). Together, these data show that CIE- and WD-induced changes in BLA glutamatergic function both functionally and biochemically mimic plasticity-related states. These mechanisms likely contribute to long-term increases in anxiety-like behavior following chronic ethanol exposure.
Collapse
Affiliation(s)
- Daniel T Christian
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | | |
Collapse
|
98
|
Kaleka KS, Petersen AN, Florence MA, Gerges NZ. Pull-down of calmodulin-binding proteins. J Vis Exp 2012:3502. [PMID: 22297704 DOI: 10.3791/3502] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Calcium (Ca(2+)) is an ion vital in regulating cellular function through a variety of mechanisms. Much of Ca(2+) signaling is mediated through the calcium-binding protein known as calmodulin (CaM). CaM is involved at multiple levels in almost all cellular processes, including apoptosis, metabolism, smooth muscle contraction, synaptic plasticity, nerve growth, inflammation and the immune response. A number of proteins help regulate these pathways through their interaction with CaM. Many of these interactions depend on the conformation of CaM, which is distinctly different when bound to Ca(2+) (Ca(2+)-CaM) as opposed to its Ca(2+)-free state (ApoCaM). While most target proteins bind Ca(2+)-CaM, certain proteins only bind to ApoCaM. Some bind CaM through their IQ-domain, including neuromodulin, neurogranin (Ng), and certain myosins. These proteins have been shown to play important roles in presynaptic function, postsynaptic function, and muscle contraction, respectively. Their ability to bind and release CaM in the absence or presence of Ca(2+) is pivotal in their function. In contrast, many proteins only bind Ca(2+)-CaM and require this binding for their activation. Examples include myosin light chain kinase, Ca(2+)/CaM-dependent kinases (CaMKs) and phosphatases (e.g. calcineurin), and spectrin kinase, which have a variety of direct and downstream effects. The effects of these proteins on cellular function are often dependent on their ability to bind to CaM in a Ca(2+)-dependent manner. For example, we tested the relevance of Ng-CaM binding in synaptic function and how different mutations affect this binding. We generated a GFP-tagged Ng construct with specific mutations in the IQ-domain that would change the ability of Ng to bind CaM in a Ca(2+)-dependent manner. The study of these different mutations gave us great insight into important processes involved in synaptic function. However, in such studies, it is essential to demonstrate that the mutated proteins have the expected altered binding to CaM. Here, we present a method for testing the ability of proteins to bind to CaM in the presence or absence of Ca(2+), using CaMKII and Ng as examples. This method is a form of affinity chromatography referred to as a CaM pull-down assay. It uses CaM-Sepharose beads to test proteins that bind to CaM and the influence of Ca(2+) on this binding. It is considerably more time efficient and requires less protein relative to column chromatography and other assays. Altogether, this provides a valuable tool to explore Ca(2+)/CaM signaling and proteins that interact with CaM.
Collapse
Affiliation(s)
- Kanwardeep S Kaleka
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, USA
| | | | | | | |
Collapse
|
99
|
Zhong L, Gerges NZ. Neurogranin and synaptic plasticity balance. Commun Integr Biol 2011; 3:340-2. [PMID: 20798820 DOI: 10.4161/cib.3.4.11763] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Accepted: 03/08/2010] [Indexed: 11/19/2022] Open
Abstract
Learning-related modifications of synaptic transmission at CA1 hippocampal excitatory synapses are activity- and NMDA receptor (NMDAR)-dependent. While a postsynaptic increase in Ca(2+) is absolutely required for synaptic plasticity induction, the molecular mechanisms underlying the transduction of synaptic signals to postsynaptic changes are not clearly understood. In our recent study, we found that the postsynaptic calmodulin (CaM)-binding protein neurogranin (Ng) enhances synaptic strength in an activity- and NMDAR-dependent manner. Furthermore we have shown that Ng is not only required for the induction of long-term potentiation (LTP), but its mediated synaptic potentiation also mimics and occludes LTP. Our results demonstrate that Ng plays an important role in the regulation of hippocampal synaptic plasticity and synaptic function. Here, we summarize our findings and further discuss their possible implications in aging-related synaptic plasticity deficits.
Collapse
Affiliation(s)
- Ling Zhong
- Department of Cell Biology, Neurobiology and Anatomy; Medical College of Wisconsin; Milwaukee, WI USA
| | | |
Collapse
|
100
|
Huang KP, Huang FL, Shetty PK. Stimulation-mediated translocation of calmodulin and neurogranin from soma to dendrites of mouse hippocampal CA1 pyramidal neurons. Neuroscience 2011; 178:1-12. [PMID: 21256930 DOI: 10.1016/j.neuroscience.2011.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 01/12/2011] [Accepted: 01/14/2011] [Indexed: 11/27/2022]
Abstract
Calmodulin (CaM) and neurogranin (Ng) are two abundant neuronal proteins in the forebrain whose interactions are implicated in the enhancement of synaptic plasticity. To gain further insight into the actions of these two proteins we investigated whether they co-localize in principle neurons and whether they respond to high frequency stimulation in a coordinated fashion. Immunohistochemical staining of CaM and Ng in mouse hippocampal slices revealed that CaM was highly concentrated in the nucleus of CA1 pyramidal neurons, whereas Ng was more broadly localized throughout the soma and dendrites. The asymmetrical localization of CaM in the nucleus of pyramidal neurons was in sharp contrast to the distribution observed in pyramidal cells of the neighboring subiculum, where CaM was uniformly localized throughout the soma and dendrites. The somatic concentrations of CaM and Ng in CA1 pyramidal neurons were approximately 10- and two-fold greater than observed in the dendrites, respectively. High frequency stimulation (HFS) of hippocampal slices promoted mobilization of CaM and Ng from soma to dendrites. These responses were spatially restricted to the area close to the site of stimulation and were inhibited by the N-methyl-D-asparate receptor antagonist 2-amino-5-phosphonopentanoic acid. Furthermore, HFS failed to promote translocation of CaM from soma to dendrites of slices from Ng knockout mice, which also exhibited deficits in HFS-induced long-term potentiation. Translocated CaM and Ng exhibited distinct puncta decorating the apical dendrites of pyramidal neurons and appeared to be concentrated in dendritic spines. These findings suggest that mobilization of CaM and Ng to stimulated dendritic spines may enhance synaptic efficacy by increasing and prolonging the Ca2+ transients and activation of Ca2+/CaM-dependent enzymes.
Collapse
Affiliation(s)
- K-P Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|