1
|
Bussmann B, Ayagama T, Liu K, Li D, Herring N. Bayliss Starling Prize Lecture 2023: Neuropeptide-Y being 'unsympathetic' to the broken hearted. J Physiol 2025; 603:1841-1864. [PMID: 38847435 PMCID: PMC11955873 DOI: 10.1113/jp285370] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 04/01/2025] Open
Abstract
William Bayliss and Ernest Starling are not only famous as pioneers in cardiovascular physiology, but also responsible for the discovery of the first hormone (from the Greek 'excite or arouse'), the intestinal signalling molecule and neuropeptide secretin in 1902. Our research group focuses on neuropeptides and neuromodulators that influence cardiovascular autonomic control as potential biomarkers in disease and tractable targets for therapeutic intervention. Acute myocardial infarction (AMI) and chronic heart failure (CHF) result in high levels of cardiac sympathetic stimulation, which is a poor prognostic indicator. Although beta-blockers improve mortality in these conditions by preventing the action of the neurotransmitter noradrenaline, a substantial residual risk remains. Recently, we have identified the sympathetic co-transmitter neuropeptide-Y (NPY) as being released during AMI, leading to larger infarcts and life-threatening arrhythmia in both animal models and patients. Here, we discuss recently published data demonstrating that peripheral venous NPY levels are associated with heart failure hospitalisation and mortality after AMI, and all cause cardiovascular mortality in CHF, even when adjusting for known risk factors (including brain natriuretic peptide). We have investigated the mechanistic basis for these observations in human and rat stellate ganglia and cardiac tissue, manipulating NPY neurochemistry at the same time as using state-of-the-art imaging techniques, to establish the receptor pathways responsible for NPY signalling. We propose NPY as a new mechanistic biomarker in AMI and CHF patients and aim to determine whether specific NPY receptor blockers can prevent arrhythmia and attenuate the development of heart failure.
Collapse
Affiliation(s)
- Benjamin Bussmann
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Thamali Ayagama
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Kun Liu
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Guo SS, Liu Z, Wang GM, Sun Z, Yu K, Fawcett JP, Buettner R, Gao B, Fässler R. KANK1 promotes breast cancer development by compromising Scribble-mediated Hippo activation. Nat Commun 2024; 15:10381. [PMID: 39613731 PMCID: PMC11607453 DOI: 10.1038/s41467-024-54645-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
KANK1 is expressed in epithelial cells and connects focal adhesions with the adjacent cortical microtubule stabilizing complex. Although KANK1 was shown to suppress cancer cell growth in vitro, TCGA database points to high KANK1 levels associated with poor prognosis in a wide spectrum of human malignancies. Here, we address this discrepancy and report that KANK1 promotes proliferation and survival of PyMT-transformed mammary tumor cells in vivo. Mechanistically, KANK1 localizes to the basal side of basement membrane (BM)-attached transformed luminal epithelial cells. When these cells lose the contact with the BM and disassemble integrin adhesions, KANK1 is found at cell-cell junctions where it competes with the polarity and tumor suppressor Scribble for NOS1AP binding, which curbs the ability of Scribble to promote Hippo pathway activity. The consequences are stabilization and nuclear accumulation of TAZ, growth and survival of tumor cells and elevated breast cancer development.
Collapse
Affiliation(s)
- Shiny Shengzhen Guo
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Zhiying Liu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Guan M Wang
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Zhiqi Sun
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Kaikai Yu
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - James P Fawcett
- Departments of Pharmacology and Surgery, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Reinhard Buettner
- Institute of Pathology, Medical Faculty, University Cologne, Cologne, Germany
| | - Bo Gao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
3
|
Mao LM, Thallapureddy K, Wang JQ. Effects of propofol on presynaptic synapsin phosphorylation in the mouse brain in vivo. Brain Res 2024; 1823:148671. [PMID: 37952872 PMCID: PMC10806815 DOI: 10.1016/j.brainres.2023.148671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The commonly used general anesthetic propofol can enhance the γ-aminobutyric acid-mediated inhibitory synaptic transmission and depress the glutamatergic excitatory synaptic transmission to achieve general anesthesia and other outcomes. In addition to the actions at postsynaptic sites, the modulation of presynaptic activity by propofol is thought to contribute to neurophysiological effects of the anesthetic, although potential targets of propofol within presynaptic nerve terminals are incompletely studied at present. In this study, we explored the possible linkage of propofol to synapsins, a family of neuron-specific phosphoproteins which are the most abundant proteins on presynaptic vesicles, in the adult mouse brain in vivo. We found that an intraperitoneal injection of propofol at a dose that caused loss of righting reflex increased basal levels of synapsin phosphorylation at the major representative phosphorylation sites (serine 9, serine 62/67, and serine 603) in the prefrontal cortex (PFC) of male and female mice. Propofol also elevated synapsin phosphorylation at these sites in the striatum and S9 and S62/67 phosphorylation in the hippocampus, while propofol had no effect on tyrosine hydroxylase phosphorylation in striatal nerve terminals. Total synapsin protein expression in the PFC, hippocampus, and striatum was not altered by propofol. These results reveal that synapsin could be a novel substrate of propofol in the presynaptic neurotransmitter release machinery. Propofol possesses the ability to upregulate synapsin phosphorylation in broad mouse brain regions.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Khyathi Thallapureddy
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
4
|
Xie W, Xing N, Qu J, Liu D, Pang Q. The Physiological Function of nNOS-Associated CAPON Proteins and the Roles of CAPON in Diseases. Int J Mol Sci 2023; 24:15808. [PMID: 37958792 PMCID: PMC10647562 DOI: 10.3390/ijms242115808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
In this review, the structure, isoform, and physiological role of the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) are summarized. There are three isoforms of CAPON in humans, including long CAPON protein (CAPON-L), short CAPON protein (CAPON-S), and CAPON-S' protein. CAPON-L includes three functional regions: a C-terminal PDZ-binding motif, carboxypeptidase (CPE)-binding region, and N-terminal phosphotyrosine (PTB) structural domain. Both CAPON-S and CAPON-S' only contain the C-terminal PDZ-binding motif. The C-terminal PDZ-binding motif of CAPON can bind with neuronal nitric oxide synthase (nNOS) and participates in regulating NO production and neuronal development. An overview is given on the relationship between CAPON and heart diseases, diabetes, psychiatric disorders, and tumors. This review will clarify future research directions on the signal pathways related to CAPON, which will be helpful for studying the regulatory mechanism of CAPON. CAPON may be used as a drug target, which will provide new ideas and solutions for treating human diseases.
Collapse
Affiliation(s)
| | | | | | - Dongwu Liu
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China; (W.X.); (N.X.)
| | - Qiuxiang Pang
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China; (W.X.); (N.X.)
| |
Collapse
|
5
|
Candemir E, Fattakhov N, Leary AO, Slattery DA, Courtney MJ, Reif A, Freudenberg F. Disrupting the nNOS/NOS1AP interaction in the medial prefrontal cortex impairs social recognition and spatial working memory in mice. Eur Neuropsychopharmacol 2023; 67:66-79. [PMID: 36513018 DOI: 10.1016/j.euroneuro.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/10/2022] [Accepted: 11/13/2022] [Indexed: 12/14/2022]
Abstract
The neuronal isoform of nitric oxide synthase (nNOS) and its interacting protein NOS1AP have been linked to several mental disorders including schizophrenia and depression. An increase in the interaction between nNOS and NOS1AP in the frontal cortex has been suggested to contribute to the emergence of these disorders. Here we aimed to uncover whether disruption of their interactions in the frontal cortex leads to mental disorder endophenotypes. Targeting the medial prefrontal cortex (mPFC), we stereotaxically injected wild-type C57BL/6J mice with recombinant adeno-associated virus (rAAV) expressing either full-length NOS1AP, the nNOS binding region of NOS1AP (i.e. NOS1AP396-503), or the nNOS amino-terminus (i.e. nNOS1-133), which was shown to disrupt the interaction of endogenous nNOS with PSD-95. We tested these mice in a comprehensive behavioural battery, assessing different endophenotypes related to mental disorders. We found no differences in anxiety-related and exploratory behaviours. Likewise, social interaction was comparable in all groups. However, social recognition was impaired in NOS1AP and NOS1AP396-503 mice. These mice, as well as mice overexpressing nNOS1-133 also displayed impaired spatial working memory (SWM) capacity, while spatial reference memory (SRM) remained intact. Finally, mice overexpressing NOS1AP and nNOS1-133, but not NOS1AP396-503, failed to habituate to the startling pulses in an acoustic startle response (ASR) paradigm, though we found no difference in overall startle intensity or prepulse inhibition (PPI) of the ASR. Our findings indicate a distinct role of NOS1AP/nNOS/PSD-95 interactions in the mPFC to contribute to specific endophenotypic changes observed in different mental disorders.
Collapse
Affiliation(s)
- Esin Candemir
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany; Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Nikolai Fattakhov
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Aet O Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Michael J Courtney
- Neuronal Signalling Laboratory, Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany.
| |
Collapse
|
6
|
Thom SR, Bhopale VM, Arya AK, Ruhela D, Bhat AR, Mitra N, Hoffstad O, Malay DS, Mirza ZK, Lantis JC, Lev-Tov HA, Kirsner RS, Hsia RC, Levinson SL, DiNubile MJ, Margolis DJ. Blood-Borne Microparticles Are an Inflammatory Stimulus in Type 2 Diabetes Mellitus. Immunohorizons 2023; 7:71-80. [PMID: 36645851 PMCID: PMC10563440 DOI: 10.4049/immunohorizons.2200099] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 01/18/2023] Open
Abstract
The proinflammatory state associated with diabetes mellitus (DM) remains poorly understood. We found patients with DM have 3- to 14-fold elevations of blood-borne microparticles (MPs) that bind phalloidin (Ph; Ph positive [+] MPs), indicating the presence of F-actin on their surface. We hypothesized that F-actin-coated MPs were an unrecognized cause for DM-associated proinflammatory status. Ph+MPs, but not Ph-negative MPs, activate human and murine (Mus musculus) neutrophils through biophysical attributes of F-actin and membrane expression of phosphatidylserine (PS). Neutrophils respond to Ph+MPs via a linked membrane array, including the receptor for advanced glycation end products and CD36, PS-binding membrane receptors. These proteins in conjunction with TLR4 are coupled to NO synthase 1 adaptor protein (NOS1AP). Neutrophil activation occurs because of Ph+MPs causing elevations of NF-κB and Src kinase (SrcK) via a concurrent increased association of NO synthase 2 and SrcK with NOS1AP, resulting in SrcK S-nitrosylation. We conclude that NOS1AP links PS-binding receptors with intracellular regulatory proteins. Ph+MPs are alarmins present in normal human plasma and are increased in those with DM and especially those with DM and a lower-extremity ulcer.
Collapse
Affiliation(s)
- Stephen R. Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Veena M. Bhopale
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Awadhesh K. Arya
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Deepa Ruhela
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Abid R. Bhat
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ole Hoffstad
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - D. Scot Malay
- Department of Surgery, Penn Presbyterian Medical Center, Philadelphia, PA
| | | | - John C. Lantis
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Hadar A. Lev-Tov
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, FL
| | - Robert S. Kirsner
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, FL
| | - Ru-Ching Hsia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD; and
| | | | | | - David J. Margolis
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
7
|
Cappelli S, Spalloni A, Feiguin F, Visani G, Šušnjar U, Brown AL, De Bardi M, Borsellino G, Secrier M, Phatnani H, Romano M, Fratta P, Longone P, Buratti E. NOS1AP is a novel molecular target and critical factor in TDP-43 pathology. Brain Commun 2022; 4:fcac242. [PMID: 36267332 PMCID: PMC9576154 DOI: 10.1093/braincomms/fcac242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/05/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Many lines of evidence have highlighted the role played by heterogeneous nuclear ribonucleoproteins in amyotrophic lateral sclerosis. In this study, we have aimed to identify transcripts co-regulated by TAR DNA-binding protein 43 kDa and highly conserved heterogeneous nuclear ribonucleoproteins which have been previously shown to regulate TAR DNA-binding protein 43 kDa toxicity (deleted in azoospermia-associated protein 1, heterogeneous nuclear ribonucleoprotein -Q, -D, -K and -U). Using the transcriptome analyses, we have uncovered that Nitric Oxide Synthase 1 Adaptor Protein mRNA is a direct TAR DNA-binding protein 43 kDa target, and in flies, its modulation alone can rescue TAR DNA-binding protein 43 kDa pathology. In primary mouse cortical neurons, we show that TAR DNA-binding protein 43 kDa mediated downregulation of Nitric Oxide Synthase 1 Adaptor Protein expression strongly affects the NMDA-receptor signalling pathway. In human patients, the downregulation of Nitric Oxide Synthase 1 Adaptor Protein mRNA strongly correlates with TAR DNA-binding protein 43 kDa proteinopathy as measured by cryptic Stathmin-2 and Unc-13 homolog A cryptic exon inclusion. Overall, our results demonstrate that Nitric Oxide Synthase 1 Adaptor Protein may represent a novel disease-relevant gene, potentially suitable for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Sara Cappelli
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Alida Spalloni
- Molecular Neurobiology, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Fabian Feiguin
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Giulia Visani
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Urša Šušnjar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Anna-Leigh Brown
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Marco De Bardi
- Neuroimmunology Unit, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Giovanna Borsellino
- Neuroimmunology Unit, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Maurizio Romano
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Patrizia Longone
- Molecular Neurobiology, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| |
Collapse
|
8
|
Qin C, Bian XL, Wu HY, Xian JY, Lin YH, Cai CY, Zhou Y, Kou XL, Li TY, Chang L, Luo CX, Zhu DY. Prevention of the return of extinguished fear by disrupting the interaction of neuronal nitric oxide synthase with its carboxy-terminal PDZ ligand. Mol Psychiatry 2021; 26:6506-6519. [PMID: 33931732 DOI: 10.1038/s41380-021-01118-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 04/13/2021] [Indexed: 02/03/2023]
Abstract
Exposure therapy based on the extinction of fear memory is first-line treatment for post-traumatic stress disorder (PTSD). However, fear extinction is relatively easy to learn but difficult to remember, extinguished fear often relapses under a number of circumstances. Here, we report that extinction learning-induced association of neuronal nitric oxide synthase (nNOS) with its carboxy-terminal PDZ ligand (CAPON) in the infralimbic (IL) subregion of medial prefrontal cortex negatively regulates extinction memory and dissociating nNOS-CAPON can prevent the return of extinguished fear in mice. Extinction training significantly increases nNOS-CAPON association in the IL. Disruptors of nNOS-CAPON increase extracellular signal-regulated kinase (ERK) phosphorylation and facilitate the retention of extinction memory in an ERK2-dependent manner. More importantly, dissociating nNOS-CAPON after extinction training enhances long-term potentiation and excitatory synaptic transmission, increases spine density in the IL, and prevents spontaneous recovery, renewal and reinstatement of remote fear of mice. Moreover, nNOS-CAPON disruptors do not affect other types of learning. Thus, nNOS-CAPON can serve as a new target for treating PTSD.
Collapse
Affiliation(s)
- Cheng Qin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xin-Lan Bian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jia-Yun Xian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Cheng-Yun Cai
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Ying Zhou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiao-Lin Kou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Ting-You Li
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China. .,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, China. .,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.
| |
Collapse
|
9
|
Szczurkowska J, Lee SI, Guo A, Cwetsch AW, Khan T, Rao S, Walz G, Huber TB, Cancedda L, Pautot S, Shelly M. A Localized Scaffold for cGMP Increase Is Required for Apical Dendrite Development. Cell Rep 2021; 31:107519. [PMID: 32294442 PMCID: PMC7293895 DOI: 10.1016/j.celrep.2020.03.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/18/2019] [Accepted: 03/24/2020] [Indexed: 10/26/2022] Open
Abstract
Studies in cultured neurons have established that axon specification instructs neuronal polarization and is necessary for dendrite development. However, dendrite formation in vivo occurs when axon formation is prevented. The mechanisms promoting dendrite development remain elusive. We find that apical dendrite development is directed by a localized cyclic guanosine monophosphate (cGMP)-synthesizing complex. We show that the scaffolding protein Scribble associates with cGMP-synthesizing enzymes soluble-guanylate-cyclase (sGC) and neuronal nitric oxide synthase (nNOS). The Scribble scaffold is preferentially localized to and mediates cGMP increase in dendrites. These events are regulated by kinesin KifC2. Knockdown of Scribble, sGC-β1, or KifC2 or disrupting their associations prevents cGMP increase in dendrites and causes severe defects in apical dendrite development. Local cGMP elevation or sGC expression rescues the effects of Scribble knockdown on dendrite development, indicating that Scribble is an upstream regulator of cGMP. During neuronal polarization, dendrite development is directed by the Scribble scaffold that might link extracellular cues to localized cGMP increase.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Seong-Il Lee
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Alan Guo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Università degli Studi di Genova, Genova, Italy
| | - Tanvir Khan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Sneha Rao
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Gerd Walz
- Department of Medicine IV, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Dulbecco Telethon Institute, Italy
| | | | - Maya Shelly
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA.
| |
Collapse
|
10
|
Dao VTV, Elbatreek MH, Fuchß T, Grädler U, Schmidt HHHW, Shah AM, Wallace A, Knowles R. Nitric Oxide Synthase Inhibitors into the Clinic at Last. Handb Exp Pharmacol 2021; 264:169-204. [PMID: 32797331 DOI: 10.1007/164_2020_382] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 1998 Nobel Prize in Medicine and Physiology for the discovery of nitric oxide, a nitrogen containing reactive oxygen species (also termed reactive nitrogen or reactive nitrogen/oxygen species) stirred great hopes. Clinical applications, however, have so far pertained exclusively to the downstream signaling of cGMP enhancing drugs such as phosphodiesterase inhibitors and soluble guanylate cyclase stimulators. All clinical attempts, so far, to inhibit NOS have failed even though preclinical models were strikingly positive and clinical biomarkers correlated perfectly. This rather casts doubt on our current way of target identification in drug discovery in general and our way of patient stratification based on correlating but not causal biomarkers or symptoms. The opposite, NO donors, nitrite and enhancing NO synthesis by eNOS/NOS3 recoupling in situations of NO deficiency, are rapidly declining in clinical relevance or hold promise but need yet to enter formal therapeutic guidelines, respectively. Nevertheless, NOS inhibition in situations of NO overproduction often jointly with enhanced superoxide (or hydrogen peroxide production) still holds promise, but most likely only in acute conditions such as neurotrauma (Stover et al., J Neurotrauma 31(19):1599-1606, 2014) and stroke (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019). Conversely, in chronic conditions, long-term inhibition of NOS might be too risky because of off-target effects on eNOS/NOS3 in particular for patients with cardiovascular risks or metabolic and renal diseases. Nitric oxide synthases (NOS) and their role in health (green) and disease (red). Only neuronal/type 1 NOS (NOS1) has a high degree of clinical validation and is in late stage development for traumatic brain injury, followed by a phase II safety/efficacy trial in ischemic stroke. The pathophysiology of NOS1 (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016) is likely to be related to parallel superoxide or hydrogen peroxide formation (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 114(46):12315-12320, 2017; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019) leading to peroxynitrite and protein nitration, etc. Endothelial/type 3 NOS (NOS3) is considered protective only and its inhibition should be avoided. The preclinical evidence for a role of high-output inducible/type 2 NOS (NOS2) isoform in sepsis, asthma, rheumatic arthritis, etc. was high, but all clinical development trials in these indications were neutral despite target engagement being validated. This casts doubt on the role of NOS2 in humans in health and disease (hence the neutral, black coloring).
Collapse
Affiliation(s)
- Vu Thao-Vi Dao
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Thomas Fuchß
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Ulrich Grädler
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Alan Wallace
- Health and Life Sciences, Coventry University, Coventry, UK
| | - Richard Knowles
- Knowles Consulting Ltd., The Stevenage Bioscience Catalyst, Stevenage, UK.
| |
Collapse
|
11
|
Saini R, Azam Z, Sapra L, Srivastava RK. Neuronal Nitric Oxide Synthase (nNOS) in Neutrophils: An Insight. Rev Physiol Biochem Pharmacol 2021; 180:49-83. [PMID: 34115206 DOI: 10.1007/112_2021_61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
NO (nitric oxide) is an important regulator of neutrophil functions and has a key role in diverse pathophysiological conditions. NO production by nitric oxide synthases (NOS) is under tight control at transcriptional, translational, and post-translational levels including interactions with heterologous proteins owing to its potent chemical reactivity and high diffusibility; this limits toxicity to other cellular components and promotes signaling specificity. The protein-protein interactions govern the activity and spatial distribution of NOS isoform to regulatory proteins and to their intended targets. In comparison with the vast literature available for endothelial, macrophages, and neuronal cells, demonstrating neuronal NOS (nNOS) interaction with other proteins through the PDZ domain, neutrophil nNOS, however, remains unexplored. Neutrophil's key role in both physiological and pathological conditions necessitates the need for further studies in delineating the NOS mediated NO modulations in signaling pathways operational in them. nNOS has been linked to depression, schizophrenia, and Parkinson's disease, suggesting the importance of exploring nNOS/NO-mediated neutrophil physiology in relation to such neuronal disorders. The review thus presents the scenario of neutrophil nNOS from the genetics to the functional level, including protein-protein interactions governing its intracellular sequestration in diverse cell types, besides speculating possible regulation in neutrophils and also addressing their clinical implications.
Collapse
Affiliation(s)
- Rashmi Saini
- Department of Zoology, Gargi College, University of Delhi, Delhi, India.
| | - Zaffar Azam
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, MP, India
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Leena Sapra
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rupesh K Srivastava
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
12
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
13
|
Ledo A, Lourenço CF, Cadenas E, Barbosa RM, Laranjinha J. The bioactivity of neuronal-derived nitric oxide in aging and neurodegeneration: Switching signaling to degeneration. Free Radic Biol Med 2021; 162:500-513. [PMID: 33186742 DOI: 10.1016/j.freeradbiomed.2020.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/22/2022]
Abstract
The small and diffusible free radical nitric oxide (•NO) has fascinated biological and medical scientists since it was promoted from atmospheric air pollutant to biological ubiquitous signaling molecule. Its unique physical chemical properties expand beyond its radical nature to include fast diffusion in aqueous and lipid environments and selective reactivity in a biological setting determined by bioavailability and reaction rate constants with biomolecules. In the brain, •NO is recognized as a key player in numerous physiological processes ranging from neurotransmission/neuromodulation to neurovascular coupling and immune response. Furthermore, changes in its bioactivity are central to the molecular pathways associated with brain aging and neurodegeneration. The understanding of •NO bioactivity in the brain, however, requires the knowledge of its concentration dynamics with high spatial and temporal resolution upon stimulation of its synthesis. Here we revise our current understanding of the role of neuronal-derived •NO in brain physiology, aging and degeneration, focused on changes in the extracellular concentration dynamics of this free radical and the regulation of bioenergetic metabolism and neurovascular coupling.
Collapse
Affiliation(s)
- A Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - C F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - E Cadenas
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, 90089, CA, USA
| | - R M Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - J Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| |
Collapse
|
14
|
Gao S, Zhang T, Jin L, Liang D, Fan G, Song Y, Lucassen PJ, Yu R, Swaab DF. CAPON Is a Critical Protein in Synaptic Molecular Networks in the Prefrontal Cortex of Mood Disorder Patients and Contributes to Depression-Like Behavior in a Mouse Model. Cereb Cortex 2020; 29:3752-3765. [PMID: 30307500 DOI: 10.1093/cercor/bhy254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 08/16/2018] [Indexed: 12/16/2022] Open
Abstract
Aberrant regulation and activity of synaptic proteins may cause synaptic pathology in the prefrontal cortex (PFC) of mood disorder patients. Carboxy-terminal PDZ ligand of NOS1 (CAPON) is a critical scaffold protein linked to synaptic proteins like nitric oxide synthase 1, synapsins. We hypothesized that CAPON is altered together with its interacting synaptic proteins in the PFC in mood disorder patients and may contribute to depression-like behaviors in mice subjected to chronic unpredictable mild stress (CUMS). Here, we found that CAPON-immunoreactivity (ir) was significantly increased in the dorsolateral PFC (DLPFC) and anterior cingulate cortex in major depressive disorder (MDD), which was accompanied by an upregulation of spinophilin-ir and a downregulation of synapsin-ir. The increases in CAPON and spinophilin and the decrease in synapsin in the DLPFC of MDD patients were also seen in the PFC of CUMS mice. CAPON-ir positively correlated with spinophilin-ir (but not with synapsin-ir) in mood disorder patients. CAPON colocalized with spinophilin in the DLPFC of MDD patients and interacted with spinophilin in human brain. Viral-mediated CAPON downregulation in the medial PFC notably reversed the depression-like behaviors in the CUMS mice. These data suggest that CAPON may contribute to aspects of depressive behavior, possibly as an interacting protein for spinophilin in the PFC.
Collapse
Affiliation(s)
- Shangfeng Gao
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Tong Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Lei Jin
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dong Liang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Guangwei Fan
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Yunnong Song
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, XH, Amsterdam, The Netherlands
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dick F Swaab
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Sharma NM, Haibara AS, Katsurada K, Liu X, Patel KP. Central angiotensin II-Protein inhibitor of neuronal nitric oxide synthase (PIN) axis contribute to neurogenic hypertension. Nitric Oxide 2019; 94:54-62. [PMID: 31654775 DOI: 10.1016/j.niox.2019.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/17/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Activation of renin-angiotensin- system, nitric oxide (NO•) bioavailability and subsequent sympathoexcitation plays a pivotal role in the pathogenesis of many cardiovascular diseases, including hypertension. Previously we have shown increased protein expression of PIN (a protein inhibitor of nNOS: neuronal nitric oxide synthase, known to dissociate nNOS dimers into monomers) with concomitantly reduced levels of catalytically active dimers of nNOS in the PVN of rats with heart failure. To elucidate the molecular mechanism by which Angiotensin II (Ang II) increases PIN expression, we used Sprague-Dawley rats (250-300 g) subjected to intracerebroventricular infusion of Ang II (20 ng/min, 0.5 μl/h) or saline as vehicle (Veh) for 14 days through osmotic mini-pumps and NG108-15 hybrid neuronal cell line treated with Ang II as an in vitro model. Ang II infusion significantly increased baseline renal sympathetic nerve activity and mean arterial pressure. Ang II infusion increased the expression of PIN (1.24 ± 0.04* Ang II vs. 0.65 ± 0.07 Veh) with a concomitant 50% decrease in dimeric nNOS and PIN-Ub conjugates (0.73 ± 0.04* Ang II vs. 1.00 ± 0.03 Veh) in the PVN. Substrate-dependent ligase assay in cells transfected with pCMV-(HA-Ub)8 vector revealed a reduction of HA-Ub-PIN conjugates after Ang II and a proteasome inhibitor, Lactacystin (LC), treatment (4.5 ± 0.7* LC Ang II vs. 9.2 ± 2.5 LC). TUBE (Tandem Ubiquitin-Binding Entities) assay showed decrease PIN-Ub conjugates in Ang II-treated cells (0.82 ± 0.12* LC Ang II vs. 1.21 ± 0.06 LC) while AT1R blocker, Losartan (Los) treatment diminished the Ang II-mediated stabilization of PIN (1.21 ± 0.07 LC Los vs. 1.16 ± 0.04* LC Ang II Los). Taken together, our studies suggest that increased central levels of Ang II contribute to the enhanced expression of PIN leading to reduced expression of the dimeric form of nNOS, thus diminishing the inhibitory action of NO• on pre-autonomic neurons in the PVN resulting in increased sympathetic outflow.
Collapse
Affiliation(s)
- Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA.
| | - Andrea S Haibara
- Department of Physiology and Biophysics, University of Minas Gerais, Belo Horizonte, MG, 31270-910, Brazil
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA
| | - Xuefei Liu
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198-5850, USA
| |
Collapse
|
16
|
Mu K, Sun Y, Zhao Y, Zhao T, Li Q, Zhang M, Li H, Zhang R, Hu C, Wang C, Jia W. Hepatic nitric oxide synthase 1 adaptor protein regulates glucose homeostasis and hepatic insulin sensitivity in obese mice depending on its PDZ binding domain. EBioMedicine 2019; 47:352-364. [PMID: 31473185 PMCID: PMC6796549 DOI: 10.1016/j.ebiom.2019.08.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/11/2019] [Accepted: 08/16/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND NOS1AP is an adaptor protein and its SNP rs12742393 was associated with type 2 diabetes (T2D). However, it remains uncertain whether NOS1AP plays a role in regulation of insulin sensitivity. Hepatic insulin resistance contributed to the development of T2D. Here, our investigation was focused on whether NOS1AP is involved in the regulation of hepatic insulin sensitivity and its underlying mechanisms. METHODS Liver specific NOS1AP condition knockout (CKO) and NOS1AP overexpression mice were generated and given a high fat diet. SNPs of NOS1AP gene were genotyped in 86 human subjects. FINDINGS NOS1AP protein is expressed in human and mouse liver. CKO mice exhibited impaired pyruvate, glucose and insulin tolerance, and increased lipid deposits in the liver. Conversely, NOS1AP overexpression in livers of obese mice improved pyruvate and/or glucose, and insulin tolerance, and attenuated liver lipid accumulation. Moreover, hepatocytes from CKO mice exhibited an elevated glucose production and mRNA expressions of Pc and Pck1. Overexpression of NOS1AP potentiated insulin-stimulated activation of IR/Akt in livers from obese mice. The insulin sensitizing effect of NOS1AP could be mimicked by overexpression of C-terminal domain of NOS1AP in ob/ob mice. Furthermore, NOS1AP overexpression in liver significantly inhibited p38 MAPK phosphorylation, and maintained ER homeostasis through p-eIF2a-ATF4-CHOP pathway. Subjects with rsl2742393 of NOS1AP have higher risk to develop hepatic steatosis. INTERPRETATION Our data demonstrate a novel role of NOS1AP in regulating hepatic insulin sensitivity and p38 MAPK inactivation in obese mice, which makes NOS1AP a potential therapeutic target for the prevention and treatment of T2D. FUND: This work was supported by the National Natural Science Foundation of China (81670707, 31340072) (to C. Wang), and National Basic Research Program of China (Nation 973 Program) (2011CB504001) (to W. Jia).
Collapse
Affiliation(s)
- Kaida Mu
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Yun Sun
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Yu Zhao
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Tianxue Zhao
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Qian Li
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Mingliang Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Huating Li
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Rong Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Cheng Hu
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| | - Chen Wang
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China.
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, People's Republic of China; Shanghai Diabetes Institute, Shanghai Jiao Tong University, People's Republic of China; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, People's Republic of China
| |
Collapse
|
17
|
Sifi M, Benabdesselam R, Souttou S, Annese T, Rendon A, Nico B, Dorbani-Mamine L. Dystrophin 71 and α1syntrophin in morpho-functional plasticity of rat supraoptic nuclei: Effect of saline surcharge and reversibly normal hydration. Acta Histochem 2018; 120:187-195. [PMID: 29395317 DOI: 10.1016/j.acthis.2018.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/25/2017] [Accepted: 01/09/2018] [Indexed: 02/02/2023]
Abstract
Dystrophin (Dp) is a multidomain protein that links the actin cytoskeleton to the extracellular matrix through the dystrophin associated proteins complex (DAPC). Dp of 71 kDa (Dp71), corresponding to the COOH-terminal domain of dystrophin, and α1-syntrophin (α1Syn) as the principal component of the DAPC, are strongly expressed in the brain. To clarify their involvement in the central control of osmotic homeostasis, we investigated the effect of 14 days of salt loading (with drinking water containing 2% NaCl) and then reversibly to 30 days of normal hydration (with drinking water without salt), first on the expression by western-blotting and the distribution by immunochemistry of Dp71 and α1Syn in the SON of the rat and, second, on the level of some physiological parameters, as the plasma osmolality, natremia and hematocrit. Dp71 is the most abundant form of dystrophin revealed in the supraoptic nucleu (SON) of control rat. Dp71 was localized in magnocellular neurons (MCNs) and astrocytes, when α1Syn was observed essentially in astrocytes end feet. After 14 days of salt-loading, Dp71 and α1Syn signals decreased and a dual signal for these two proteins was revealed in the astrocytes processes SON surrounding blood capillaries. In addition, salt loading leads to an increase in plasma osmolality, natremia and hematocrit. Reversibly, after 30 days of normal hydration, the intensity of the signal for the two proteins, Dp71 and α1Syn, increased and approached that of control. Furtheremore, the levels of the physiological parameters decreased and approximated those of control. This suggests that Dp71 and α1Syn may be involved in the functional activity of the SON. Their localization in astrocyte end feet emphasizes their importance in neuronal-vascular-astrocyte interactions for the central detection of osmolality. In the SON, Dp71 and α1Syn may be involved in osmosensitivity.
Collapse
Affiliation(s)
- Madina Sifi
- Equipe de Neurochimie, LBPO, Faculté des Sciences Biologiques, USTHB, Alger, Algeria
| | - Roza Benabdesselam
- Equipe de Neurochimie, LBPO, Faculté des Sciences Biologiques, USTHB, Alger, Algeria; Département de Biologie, Faculté des Sciences Biologiques et Agronomiques, UMMTO, Tizi Ouzou, Algeria.
| | - Sabrina Souttou
- Equipe de Neurochimie, LBPO, Faculté des Sciences Biologiques, USTHB, Alger, Algeria
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organes, University of Bari "Aldo Moro", Bari, Italy
| | - Alvaro Rendon
- Laboratoire de Physiopathologie Cellulaire et Moleculaire de la Retine, INSERM UMRS-592, Institut de la Vision, Paris, France
| | - Beatrice Nico
- Department of Basic Medical Sciences, Neurosciences and Sensory Organes, University of Bari "Aldo Moro", Bari, Italy
| | - Latifa Dorbani-Mamine
- Equipe de Neurochimie, LBPO, Faculté des Sciences Biologiques, USTHB, Alger, Algeria
| |
Collapse
|
18
|
Svane KC, Asis EK, Omelchenko A, Kunnath AJ, Brzustowicz LM, Silverstein SM, Firestein BL. d-Serine administration affects nitric oxide synthase 1 adaptor protein and DISC1 expression in sex-specific manner. Mol Cell Neurosci 2018; 89:20-32. [PMID: 29601869 DOI: 10.1016/j.mcn.2018.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/23/2018] [Accepted: 03/25/2018] [Indexed: 01/19/2023] Open
Abstract
Antipsychotic medications are inefficient at treating symptoms of schizophrenia (SCZ), and N-methyl d-aspartate receptor (NMDAR) agonists are potential therapeutic alternatives. As such, these agonists may act on different pathways and proteins altered in the brains of patients with SCZ than do antipsychotic medications. Here, we investigate the effects of administration of the antipsychotic haloperidol and NMDAR agonist d-serine on function and expression of three proteins that play significant roles in SCZ: nitric oxide synthase 1 adaptor protein (NOS1AP), dopamine D2 (D2) receptor, and disrupted in schizophrenia 1 (DISC1). We administered haloperidol or d-serine to male and female Sprague Dawley rats via intraperitoneal injection for 12 days and subsequently examined cortical expression of NOS1AP, D2 receptor, and DISC1. We found sex-specific effects of haloperidol and d-serine treatment on the expression of these proteins. Haloperidol significantly reduced expression of D2 receptor in male, but not female, rats. Conversely, d-serine reduced expression of NOS1AP in male rats and did not affect D2 receptor expression. d-serine treatment also reduced expression of DISC1 in male rats and increased DISC1 expression in female rats. As NOS1AP is overexpressed in the cortex of patients with SCZ and negatively regulates NMDAR signaling, we subsequently examined whether treatment with antipsychotics or NMDAR agonists can reverse the detrimental effects of NOS1AP overexpression in vitro as previously reported by our group. NOS1AP overexpression promotes reduced dendrite branching in vitro, and as such, we treated cortical neurons overexpressing NOS1AP with different antipsychotics (haloperidol, clozapine, fluphenazine) or d-serine for 24 h and determined the effects of these drugs on NOS1AP expression and dendrite branching. While antipsychotics did not affect NOS1AP protein expression or dendrite branching in vitro, d-serine reduced NOS1AP expression and rescued NOS1AP-mediated reductions in dendrite branching. Taken together, our data suggest that d-serine influences the function and expression of NOS1AP, D2 receptor, and DISC1 in a sex-specific manner and reverses the effects of NOS1AP overexpression on dendrite morphology.
Collapse
Affiliation(s)
- Kirsten C Svane
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA; Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ericka-Kate Asis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA; Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ansley J Kunnath
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Linda M Brzustowicz
- Department of Genetics, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Steven M Silverstein
- Division of Schizophrenia Research, Rutgers University Behavioral Health Care, 671 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
19
|
Abstract
SIGNIFICANCE The family of gasotransmitter molecules, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), has emerged as an important mediator of numerous cellular signal transduction and pathophysiological responses. As such, these molecules have been reported to influence a diverse array of biochemical, molecular, and cell biology events often impacting one another. Recent Advances: Discrete regulation of gasotransmitter molecule formation, movement, and reaction is critical to their biological function. Due to the chemical nature of these molecules, they can move rapidly throughout cells and tissues acting on targets through reactions with metal groups, reactive chemical species, and protein amino acids. CRITICAL ISSUES Given the breadth and complexity of gasotransmitter reactions, this field of research is expanding into exciting, yet sometimes confusing, areas of study with significant promise for understanding health and disease. The precise amounts of tissue and cellular gasotransmitter levels and where they are formed, as well as how they react with molecular targets or themselves, all remain poorly understood. FUTURE DIRECTIONS Elucidation of specific molecular targets, characteristics of gasotransmitter molecule heterotypic interactions, and spatiotemporal formation and metabolism are all important to better understand their true pathophysiological importance in various organ systems. Antioxid. Redox Signal. 26, 936-960.
Collapse
Affiliation(s)
- Gopi K Kolluru
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Xinggui Shen
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Shuai Yuan
- 2 Department of Cellular Biology and Anatomy, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Christopher G Kevil
- 1 Department of Pathology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana.,2 Department of Cellular Biology and Anatomy, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana.,3 Department of Molecular and Cellular Physiology, LSU Health Sciences Center-Shreveport , Shreveport, Louisiana
| |
Collapse
|
20
|
Sharma NM, Patel KP. Post-translational regulation of neuronal nitric oxide synthase: implications for sympathoexcitatory states. Expert Opin Ther Targets 2017; 21:11-22. [PMID: 27885874 PMCID: PMC5488701 DOI: 10.1080/14728222.2017.1265505] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 11/23/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Nitric oxide (NO) synthesized via neuronal nitric oxide synthase (nNOS) plays a significant role in regulation/modulation of autonomic control of circulation. Various pathological states are associated with diminished nNOS expression and blunted autonomic effects of NO in the central nervous system (CNS) including heart failure, hypertension, diabetes mellitus, chronic renal failure etc. Therefore, elucidation of the molecular mechanism/s involved in dysregulation of nNOS is essential to understand the pathogenesis of increased sympathoexcitation in these diseased states. Areas covered: nNOS is a highly regulated enzyme, being regulated at transcriptional and posttranslational levels via protein-protein interactions and modifications viz. phosphorylation, ubiquitination, and sumoylation. The enzyme activity of nNOS also depends on the optimal concentration of substrate, cofactors and association with regulatory proteins. This review focuses on the posttranslational regulation of nNOS in the context of normal and diseased states within the CNS. Expert opinion: Gaining insight into the mechanism/s involved in the regulation of nNOS would provide novel strategies for manipulating nNOS directed therapeutic modalities in the future, including catalytically active dimer stabilization and protein-protein interactions with intracellular protein effectors. Ultimately, this is expected to provide tools to improve autonomic dysregulation in various diseases such as heart failure, hypertension, and diabetes.
Collapse
Affiliation(s)
- Neeru M Sharma
- a Department of Cellular & Integrative Physiology , University of Nebraska Medical Center , Omaha , NE , USA
| | - Kaushik P Patel
- a Department of Cellular & Integrative Physiology , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
21
|
Wang J, Jin L, Zhu Y, Zhou X, Yu R, Gao S. Research progress in NOS1AP in neurological and psychiatric diseases. Brain Res Bull 2016; 125:99-105. [PMID: 27237129 DOI: 10.1016/j.brainresbull.2016.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 11/19/2022]
Abstract
Nitric Oxide Synthase 1 Adaptor Protein (NOS1AP, previously named CAPON) was firstly identified in rat brain in 1998. Structurally, NOS1AP consists of a phosphotyrosine-binding (PTB) domain at its N-terminal and a PDZ (PSD-95/discs-large/ZO-1) ligand motif at its C-terminal. The PTB domain of NOS1AP mediates the interactions with Dexras1, scribble, and synapsins. The PDZ ligand motif of NOS1AP binds to the PDZ domain of NOS1, the enzyme responsible for nitric oxide synthesis in the nervous system. NOS1AP is implicated in Dexras1 activation, neuronal nitric oxide production, Hippo pathway signaling, and dendritic development through the association with these important partners. An increasing body of evidence is pointing to the significant roles of NOS1AP in excitotoxic neuronal damage, traumatic nervous system injury, bipolar disorder, and schizophrenia. However, the study progress in NOS1AP in neurological or psychiatric diseases, has not been systematically reviewed. Here we introduce the expression, structure, and isoforms of NOS1AP, then summarize the physiological roles of NOS1AP, and discuss the relationships between NOS1AP alterations and the pathophysiology of some neurological and psychiatric disorders. The review will promote the further investigation of NOS1AP in brain disorders and the development of drugs targeting the NOS1AP PTB domain or PDZ-binding motif in the future.
Collapse
Affiliation(s)
- Jie Wang
- The Graduate School, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Lei Jin
- The Graduate School, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Yufu Zhu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-Hai Road, Xuzhou 221002, Jiangsu, People's Republic of China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-Hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China; Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-Hai Road, Xuzhou 221002, Jiangsu, People's Republic of China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-Hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China; Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-Hai Road, Xuzhou 221002, Jiangsu, People's Republic of China.
| | - Shangfeng Gao
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-Hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China; Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-Hai Road, Xuzhou 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Candemir E, Kollert L, Weißflog L, Geis M, Müller A, Post AM, O'Leary A, Harro J, Reif A, Freudenberg F. Interaction of NOS1AP with the NOS-I PDZ domain: Implications for schizophrenia-related alterations in dendritic morphology. Eur Neuropsychopharmacol 2016; 26:741-55. [PMID: 26861996 DOI: 10.1016/j.euroneuro.2016.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/23/2015] [Accepted: 01/23/2016] [Indexed: 12/19/2022]
Abstract
Schizophrenia involves morphological brain changes, including changes in synaptic plasticity and altered dendritic development. Amongst the most promising candidate molecules for schizophrenia are neuronal nitric oxide (NO) synthase (NOS-I, also known as nNOS) and its adapter protein NOS1AP (previously named CAPON). However, the precise molecular mechanisms by which NOS-I and NOS1AP affect disease pathology remain to be resolved. Interestingly, overexpression of NOS1AP affects dendritic morphology, possibly through increased association with the NOS-I PDZ domain. To investigate the effect of NOS1AP on dendritic morphology we overexpressed different NOS1AP isoforms, NOS1AP deletion mutants and the aminoterminal 133 amino acids of NOS-I (NOS-IN133) containing an extended PDZ domain. We examined the interaction of the overexpressed constructs with endogenous NOS-I by co-immunoprecipitation and the consequences of increased NOS-I/NOS1AP PDZ interaction in primary cultures of hippocampal and cortical neurons from C57BL/6J mice. Neurons overexpressing NOS1AP isoforms or deletion mutants showed highly altered spine morphology and excessive growth of filopodia-like protrusions. Sholl analysis of immunostained primary cultured neurons revealed that dendritic branching was mildly affected by NOS1AP overexpression. Our results hint towards an involvement of NOS-I/NOS1AP interaction in the regulation of dendritic spine plasticity. As altered dendritic spine development and filopodial outgrowth are important neuropathological features of schizophrenia, our findings may provide insight into part of the molecular mechanisms involved in brain morphology alterations observed in schizophrenia. As the NOS-I/NOS1AP interface can be targeted by small molecules, our findings ultimately might help to develop novel treatment strategies for schizophrenia patients.
Collapse
Affiliation(s)
- Esin Candemir
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt am Main, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany; Graduate School of Life Sciences, University of Würzburg, 97080 Würzburg, Germany
| | - Leonie Kollert
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Lena Weißflog
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt am Main, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Maria Geis
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Antje Müller
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Antonia M Post
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt am Main, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A, Tartu 50411 Estonia
| | - Jaanus Harro
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A, Tartu 50411 Estonia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt am Main, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt am Main, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
23
|
Hernandez K, Swiatkowski P, Patel MV, Liang C, Dudzinski NR, Brzustowicz LM, Firestein BL. Overexpression of Isoforms of Nitric Oxide Synthase 1 Adaptor Protein, Encoded by a Risk Gene for Schizophrenia, Alters Actin Dynamics and Synaptic Function. Front Cell Neurosci 2016; 10:6. [PMID: 26869880 PMCID: PMC4735351 DOI: 10.3389/fncel.2016.00006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
Abstract
Proper communication between neurons depends upon appropriate patterning of dendrites and correct distribution and structure of spines. Schizophrenia is a neuropsychiatric disorder characterized by alterations in dendrite branching and spine density. Nitric oxide synthase 1 adaptor protein (NOS1AP), a risk gene for schizophrenia, encodes proteins that are upregulated in the dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia. To elucidate the effects of NOS1AP overexpression observed in individuals with schizophrenia, we investigated changes in actin dynamics and spine development when a long (NOS1AP-L) or short (NOS1AP-S) isoform of NOS1AP is overexpressed. Increased NOS1AP-L protein promotes the formation of immature spines when overexpressed in rat cortical neurons from day in vitro (DIV) 14 to DIV 17 and reduces the amplitude of miniature excitatory postsynaptic currents (mEPSCs). In contrast, increased NOS1AP-S protein increases the rate of actin polymerization and the number of immature and mature spines, which may be attributed to a decrease in total Rac1 expression and a reduction in the levels of active cofilin. The increase in the number of mature spines by overexpression of NOS1AP-S is accompanied by an increase in the frequency of mEPSCs. Our findings show that overexpression of NOS1AP-L or NOS1AP-S alters the actin cytoskeleton and synaptic function. However, the mechanisms by which these isoforms induce these changes are distinct. These results are important for understanding how increased expression of NOS1AP isoforms can influence spine development and synaptic function.
Collapse
Affiliation(s)
- Kristina Hernandez
- Department of Cell Biology and Neuroscience, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Mihir V. Patel
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Chen Liang
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Natasha R. Dudzinski
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Linda M. Brzustowicz
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| |
Collapse
|
24
|
Bouvier G, Bidoret C, Casado M, Paoletti P. Presynaptic NMDA receptors: Roles and rules. Neuroscience 2015; 311:322-40. [DOI: 10.1016/j.neuroscience.2015.10.033] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/18/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
|
25
|
Freudenberg F, Alttoa A, Reif A. Neuronal nitric oxide synthase (NOS1) and its adaptor, NOS1AP, as a genetic risk factors for psychiatric disorders. GENES BRAIN AND BEHAVIOR 2015; 14:46-63. [PMID: 25612209 DOI: 10.1111/gbb.12193] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 12/15/2022]
Abstract
Nitric oxide (NO) is a gaseous transmitter produced by nitric oxide synthases (NOSs). The neuronal isoform (NOS-I, encoded by NOS1) is the main source of NO in the central nervous system (CNS). Animal studies suggest that nitrinergic dysregulation may lead to behavioral abnormalities. Unfortunately, the large number of animal studies is not adequately reflected by publications concerning humans. These include post-mortem studies, determination of biomarkers, and genetic association studies. Here, we review the evidence for the role of NO in psychiatric disorders by focusing on the human NOS1 gene as well as biomarker studies. Owing to the complex regulation of NOS1 and the varying function of NOS-I in different brain regions, no simple, unidirectional association is expected. Rather, the 'where, when and how much' of NO formation is decisive. Present data, although still preliminary and partially conflicting, suggest that genetically driven reduced NO signaling in the prefrontal cortex is associated with schizophrenia and cognition. Both NOS1 and its interaction partner NOS1AP have a role therein. Also, reduced NOS1 expression in the striatum determined by a length polymorphism in a NOS1 promoter (NOS1 ex1f-VNTR) goes along with a variety of impulsive behaviors. An association of NOS1 with mood disorders, suggested by animal models, is less clear on the genetic level; however, NO metabolites in blood may serve as biomarkers for major depression and bipolar disorder. As the nitrinergic system comprises a relevant target for pharmacological interventions, further studies are warranted not only to elucidate the pathophysiology of mental disorders, but also to evaluate NO function as a biomarker.
Collapse
Affiliation(s)
- F Freudenberg
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | | | | |
Collapse
|
26
|
Nacsa K, Elekes K, Serfőző Z. Ultrastructural localization of NADPH diaphorase and nitric oxide synthase in the neuropils of the snail CNS. Micron 2015; 75:58-66. [DOI: 10.1016/j.micron.2015.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022]
|
27
|
Carrel D, Hernandez K, Kwon M, Mau C, Trivedi MP, Brzustowicz LM, Firestein BL. Nitric oxide synthase 1 adaptor protein, a protein implicated in schizophrenia, controls radial migration of cortical neurons. Biol Psychiatry 2015; 77:969-78. [PMID: 25542305 PMCID: PMC4416077 DOI: 10.1016/j.biopsych.2014.10.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/08/2014] [Accepted: 10/22/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Where a neuron is positioned in the brain during development determines neuronal circuitry and information processing needed for normal brain function. When aberrations in this process occur, cognitive disorders may result. Patients diagnosed with schizophrenia have been reported to show altered neuronal connectivity and heterotopias. To elucidate pathways by which this process occurs and become aberrant, we have chosen to study the long isoform of nitric oxide synthase 1 adaptor protein (NOS1AP), a protein encoded by a susceptibility gene for schizophrenia. METHODS To determine whether NOS1AP plays a role in cortical patterning, we knocked down or co-overexpressed NOS1AP and a green fluorescent protein or red fluorescent protein (TagRFP) reporter in neuronal progenitor cells of the embryonic rat neocortex using in utero electroporation. We analyzed sections of cortex (ventricular zone, intermediate zone, and cortical plate [CP]) containing green fluorescent protein or red fluorescent protein TagRFP positive cells and counted the percentage of positive cells that migrated to each region from at least three rats for each condition. RESULTS NOS1AP overexpression disrupts neuronal migration, resulting in increased cells in intermediate zone and less cells in CP, and decreases dendritogenesis. Knockdown results in increased migration, with more cells reaching the CP. The phosphotyrosine binding region, but not the PDZ-binding motif, is necessary for NOS1AP function. Amino acids 181 to 307, which are sufficient for NOS1AP-mediated decreases in dendrite number, have no effect on migration. CONCLUSIONS Our studies show for the first time a critical role for the schizophrenia-associated gene NOS1AP in cortical patterning, which may contribute to underlying pathophysiology seen in schizophrenia.
Collapse
Affiliation(s)
- Damien Carrel
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey; Neurophotonics Laboratory, Paris Descartes University, Paris, France
| | - Kristina Hernandez
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey; Molecular Biosciences Graduate Program Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Munjin Kwon
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey; Molecular Biosciences Graduate Program Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Christine Mau
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Meera P Trivedi
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Linda M Brzustowicz
- Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey.
| |
Collapse
|
28
|
Lu CJ, Hao G, Nikiforova N, Larsen HE, Liu K, Crabtree MJ, Li D, Herring N, Paterson DJ. CAPON modulates neuronal calcium handling and cardiac sympathetic neurotransmission during dysautonomia in hypertension. Hypertension 2015; 65:1288-1297. [PMID: 25916729 DOI: 10.1161/hypertensionaha.115.05290] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/03/2015] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies implicate a variant in the neuronal nitric oxide synthase adaptor protein (CAPON) in electrocardiographic QT variation and sudden cardiac death. Interestingly, nitric oxide generated by neuronal NO synthase-1 reduces norepinephrine release; however, this pathway is downregulated in animal models of cardiovascular disease. Because sympathetic hyperactivity can trigger arrhythmia, is this neural phenotype linked to CAPON dysregulation? We hypothesized that CAPON resides in cardiac sympathetic neurons and is a part of the prediseased neuronal phenotype that modulates calcium handling and neurotransmission in dysautonomia. CAPON expression was significantly reduced in the stellate ganglia of spontaneously hypertensive rats before the development of hypertension compared with age-matched Wistar-Kyoto rats. The neuronal calcium current (ICa; n=8) and intracellular calcium transient ([Ca(2+)]i; n=16) were significantly larger in the spontaneously hypertensive rat than in Wistar-Kyoto rat (P<0.05). A novel noradrenergic specific vector (Ad.PRSx8-mCherry/CAPON) significantly upregulated CAPON expression, NO synthase-1 activity, and cGMP in spontaneously hypertensive rat neurons without altering NO synthase-1 levels. Neuronal ICa and [Ca(2+)]i were significantly reduced after CAPON transduction compared with the empty vector. In addition, Ad.PRSx8-mCherry/CAPON also reduced (3)H-norepinephrine release from spontaneously hypertensive rat atria (n=7). NO synthase-1 inhibition (AAAN, 10 μmol/L; n=6) reversed these effects compared with the empty virus alone. In conclusion, targeted upregulation of CAPON decreases cardiac sympathetic hyperactivity. Moreover, dysregulation of this adaptor protein in sympathetic neurons might further amplify the negative cardiac electrophysiological properties seen with CAPON mutations.
Collapse
Affiliation(s)
- Chieh-Ju Lu
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Guoliang Hao
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Natalia Nikiforova
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Hege E Larsen
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Kun Liu
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Mark J Crabtree
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Dan Li
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| | - David J Paterson
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre (C.-J.L., G.H., N.N., H.E.L., K.L., D.L., N.H., D.J.P.) and Radcliffe Department of Medicine, John Radcliffe Hospital (M.J.C.), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
NOS1AP Functionally Associates with YAP To Regulate Hippo Signaling. Mol Cell Biol 2015; 35:2265-77. [PMID: 25918243 DOI: 10.1128/mcb.00062-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022] Open
Abstract
Deregulation of cellular polarity proteins and their associated complexes leads to changes in cell migration and proliferation. The nitric oxide synthase 1 adaptor protein (NOS1AP) associates with the tumor suppressor protein Scribble to control cell migration and oncogenic transformation. However, how NOS1AP is linked to the cell signaling events that curb oncogenic progression has remained elusive. Here we identify several novel NOS1AP isoforms, NOS1APd, NOS1APe, and NOS1APf, with distinct cellular localizations. We show that isoforms with a membrane-interacting phosphotyrosine binding (PTB) domain can associate with Scribble and recognize acidic phospholipids. In a screen to identify novel binding proteins, we have discovered a complex consisting of NOS1AP and the transcriptional coactivator YAP linking NOS1AP to the Hippo signaling pathway. Silencing of NOS1AP reduces the phosphorylation of YAP and of the upstream kinase Lats1. Conversely, expression of NOS1AP promotes YAP and Lats1 phosphorylation, which correlates with reduced TEAD activity and restricted cell proliferation. Together, these data implicate a role for NOS1AP in the regulation of core Hippo signaling and are consistent with the idea that NOS1AP functions as a tumor suppressor.
Collapse
|
30
|
Altamirano F, Perez CF, Liu M, Widrick J, Barton ER, Allen PD, Adams JA, Lopez JR. Whole body periodic acceleration is an effective therapy to ameliorate muscular dystrophy in mdx mice. PLoS One 2014; 9:e106590. [PMID: 25181488 PMCID: PMC4152333 DOI: 10.1371/journal.pone.0106590] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/30/2014] [Indexed: 12/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder caused by the absence of dystrophin in both skeletal and cardiac muscles. This leads to severe muscle degeneration, and dilated cardiomyopathy that produces patient death, which in most cases occurs before the end of the second decade. Several lines of evidence have shown that modulators of nitric oxide (NO) pathway can improve skeletal muscle and cardiac function in the mdx mouse, a mouse model for DMD. Whole body periodic acceleration (pGz) is produced by applying sinusoidal motion to supine humans and in standing conscious rodents in a headward-footward direction using a motion platform. It adds small pulses as a function of movement frequency to the circulation thereby increasing pulsatile shear stress to the vascular endothelium, which in turn increases production of NO. In this study, we examined the potential therapeutic properties of pGz for the treatment of skeletal muscle pathology observed in the mdx mouse. We found that pGz (480 cpm, 8 days, 1 hr per day) decreased intracellular Ca2+ and Na+ overload, diminished serum levels of creatine kinase (CK) and reduced intracellular accumulation of Evans Blue. Furthermore, pGz increased muscle force generation and expression of both utrophin and the carboxy-terminal PDZ ligand of nNOS (CAPON). Likewise, pGz (120 cpm, 12 h) applied in vitro to skeletal muscle myotubes reduced Ca2+ and Na+ overload, diminished abnormal sarcolemmal Ca2+ entry and increased phosphorylation of endothelial NOS. Overall, this study provides new insights into the potential therapeutic efficacy of pGz as a non-invasive and non-pharmacological approach for the treatment of DMD patients through activation of the NO pathway.
Collapse
Affiliation(s)
- Francisco Altamirano
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claudio F. Perez
- Department of Anesthesiology Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Min Liu
- Department of Physiology, Perleman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jeffrey Widrick
- Division of Genetics and Program in Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elisabeth R. Barton
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Paul D. Allen
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
- Department of Anesthesiology Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami, Florida, United States of America
| | - Jose R. Lopez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
- Department of Anesthesiology Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
CAPON-nNOS coupling can serve as a target for developing new anxiolytics. Nat Med 2014; 20:1050-4. [PMID: 25129479 DOI: 10.1038/nm.3644] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023]
Abstract
Anxiety disorders are highly prevalent psychiatric diseases. There is need for a deeper understanding of anxiety control mechanisms in the mammalian brain and for development of new anxiolytic agents. Here we report that the coupling between neuronal nitric oxide synthase (nNOS) and its carboxy-terminal PDZ ligand (CAPON) can serve as a target for developing new anxiolytic agents. Augmenting nNOS-CAPON interaction in the hippocampus of mice by overexpressing full-length CAPON gave rise to anxiogenic-like behaviors, whereas dissociating CAPON from nNOS by overexpressing CAPON-125C or CAPON-20C (the C-terminal 125 or 20 amino acids of CAPON) or delivering Tat-CAPON-12C (a peptide comprising Tat and the 12 C-terminal amino acids of CAPON) in the hippocampus of mice produced anxiolytic-like effects. Mice subjected to chronic mild stress (CMS) displayed a substantial increase in nNOS-CAPON coupling in the hippocampus and a consequent anxiogenic-like phenotype. Disrupting nNOS-CAPON coupling reversed the CMS-induced anxiogenic-like behaviors. Moreover, small-molecule blockers of nNOS-CAPON binding rapidly produced anxiolytic-like effects. Dexamethasone-induced ras protein 1 (Dexras1)-extracellular signal-regulated kinase (ERK) signaling was involved in the behavioral effects of nNOS-CAPON association. Thus, nNOS-CAPON association contributes to the modulation of anxiety-related behaviors via regulating Dexras1-ERK signaling and can serve as a target for developing potential anxiolytics.
Collapse
|
32
|
Bouter Y, Kacprowski T, Weissmann R, Dietrich K, Borgers H, Brauß A, Sperling C, Wirths O, Albrecht M, Jensen LR, Kuss AW, Bayer TA. Deciphering the molecular profile of plaques, memory decline and neuron loss in two mouse models for Alzheimer's disease by deep sequencing. Front Aging Neurosci 2014; 6:75. [PMID: 24795628 PMCID: PMC3997018 DOI: 10.3389/fnagi.2014.00075] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/02/2014] [Indexed: 12/23/2022] Open
Abstract
One of the central research questions on the etiology of Alzheimer’s disease (AD) is the elucidation of the molecular signatures triggered by the amyloid cascade of pathological events. Next-generation sequencing allows the identification of genes involved in disease processes in an unbiased manner. We have combined this technique with the analysis of two AD mouse models: (1) The 5XFAD model develops early plaque formation, intraneuronal Aβ aggregation, neuron loss, and behavioral deficits. (2) The Tg4–42 model expresses N-truncated Aβ4–42 and develops neuron loss and behavioral deficits albeit without plaque formation. Our results show that learning and memory deficits in the Morris water maze and fear conditioning tasks in Tg4–42 mice at 12 months of age are similar to the deficits in 5XFAD animals. This suggested that comparative gene expression analysis between the models would allow the dissection of plaque-related and -unrelated disease relevant factors. Using deep sequencing differentially expressed genes (DEGs) were identified and subsequently verified by quantitative PCR. Nineteen DEGs were identified in pre-symptomatic young 5XFAD mice, and none in young Tg4–42 mice. In the aged cohort, 131 DEGs were found in 5XFAD and 56 DEGs in Tg4–42 mice. Many of the DEGs specific to the 5XFAD model belong to neuroinflammatory processes typically associated with plaques. Interestingly, 36 DEGs were identified in both mouse models indicating common disease pathways associated with behavioral deficits and neuron loss.
Collapse
Affiliation(s)
- Yvonne Bouter
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| | - Tim Kacprowski
- Department of Bioinformatics, Institute of Biometrics and Medical Informatics, University Medicine Greifswald , Greifswald , Germany ; Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald , Greifswald , Germany
| | - Robert Weissmann
- Human Molecular Genetics, Department for Human Genetics of the Institute for Genetics and Functional Genomics, Institute for Human Genetics, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald , Greifswald , Germany
| | - Katharina Dietrich
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| | - Henning Borgers
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| | - Andreas Brauß
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| | - Christian Sperling
- Human Molecular Genetics, Department for Human Genetics of the Institute for Genetics and Functional Genomics, Institute for Human Genetics, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald , Greifswald , Germany
| | - Oliver Wirths
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| | - Mario Albrecht
- Department of Bioinformatics, Institute of Biometrics and Medical Informatics, University Medicine Greifswald , Greifswald , Germany ; Institute for Knowledge Discovery, Graz University of Technology , Graz , Austria
| | - Lars R Jensen
- Human Molecular Genetics, Department for Human Genetics of the Institute for Genetics and Functional Genomics, Institute for Human Genetics, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald , Greifswald , Germany
| | - Andreas W Kuss
- Human Molecular Genetics, Department for Human Genetics of the Institute for Genetics and Functional Genomics, Institute for Human Genetics, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald , Greifswald , Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Georg-August-University Goettingen, University Medicine Goettingen , Goettingen , Germany
| |
Collapse
|
33
|
Auer DR, Sysa-Shah P, Bedja D, Simmers JL, Pak E, Dutra A, Cohn R, Gabrielson KL, Chakravarti A, Kapoor A. Generation of a cre recombinase-conditional Nos1ap over-expression transgenic mouse. Biotechnol Lett 2014; 36:1179-85. [PMID: 24563304 DOI: 10.1007/s10529-014-1473-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/09/2014] [Indexed: 10/25/2022]
Abstract
Polymorphic non-coding variants at the NOS1AP locus have been associated with the common cardiac, metabolic and neurological traits and diseases. Although, in vitro gene targeting-based cellular and biochemical studies have shed some light on NOS1AP function in cardiac and neuronal tissue, to enhance our understanding of NOS1AP function in mammalian physiology and disease, we report the generation of cre recombinase-conditional Nos1ap over-expression transgenic mice (Nos1ap (Tg)). Conditional transgenic mice were generated by the pronuclear injection method and three independent, single-site, multiple copies integration event-based founder lines were selected. For heart-restricted over-expression, Nos1ap (Tg) mice were crossed with Mlc2v-cre and Nos1ap transcript over-expression was observed in left ventricles from Nos1ap (Tg); Mlc2v-cre F1 mice. We believe that with the potential of conditional over-expression, Nos1ap (Tg) mice will be a useful resource in studying NOS1AP function in various tissues under physiological and disease states.
Collapse
Affiliation(s)
- Dallas R Auer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD, 21205, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Weber H, Klamer D, Freudenberg F, Kittel-Schneider S, Rivero O, Scholz CJ, Volkert J, Kopf J, Heupel J, Herterich S, Adolfsson R, Alttoa A, Post A, Grußendorf H, Kramer A, Gessner A, Schmidt B, Hempel S, Jacob CP, Sanjuán J, Moltó MD, Lesch KP, Freitag CM, Kent L, Reif A. The genetic contribution of the NO system at the glutamatergic post-synapse to schizophrenia: further evidence and meta-analysis. Eur Neuropsychopharmacol 2014; 24:65-85. [PMID: 24220657 DOI: 10.1016/j.euroneuro.2013.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/09/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
NO is a pleiotropic signaling molecule and has an important role in cognition and emotion. In the brain, NO is produced by neuronal nitric oxide synthase (NOS-I, encoded by NOS1) coupled to the NMDA receptor via PDZ interactions; this protein-protein interaction is disrupted upon binding of NOS1 adapter protein (encoded by NOS1AP) to NOS-I. As both NOS1 and NOS1AP were associated with schizophrenia, we here investigated these genes in greater detail by genotyping new samples and conducting a meta-analysis of our own and published data. In doing so, we confirmed association of both genes with schizophrenia and found evidence for their interaction in increasing risk towards disease. Our strongest finding was the NOS1 promoter SNP rs41279104, yielding an odds ratio of 1.29 in the meta-analysis. As findings from heterologous cell systems have suggested that the risk allele decreases gene expression, we studied the effect of the variant on NOS1 expression in human post-mortem brain samples and found that the risk allele significantly decreases expression of NOS1 in the prefrontal cortex. Bioinformatic analyses suggest that this might be due the replacement of six transcription factor binding sites by two new binding sites as a consequence of proxy SNPs. Taken together, our data argue that genetic variance in NOS1 resulting in lower prefrontal brain expression of this gene contributes to schizophrenia liability, and that NOS1 interacts with NOS1AP in doing so. The NOS1-NOS1AP PDZ interface may thus well constitute a novel target for small molecules in at least some forms of schizophrenia.
Collapse
Affiliation(s)
- H Weber
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany; Microarray Core Unit, IZKF Würzburg, University Hospital of Würzburg, Germany
| | - D Klamer
- Department of Pharmacology, The Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Sweden
| | - F Freudenberg
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - S Kittel-Schneider
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - O Rivero
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany; CIBERSAM, Universitat de Valencia, Valencia, Spain
| | - C-J Scholz
- Microarray Core Unit, IZKF Würzburg, University Hospital of Würzburg, Germany
| | - J Volkert
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - J Kopf
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - J Heupel
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - S Herterich
- Comprehensive Heart Failure Center, University of Würzburg, Germany
| | - R Adolfsson
- Department of Clinivcal Sciences, Psychiatry, Umeå University, Sweden
| | - A Alttoa
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - A Post
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - H Grußendorf
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - A Kramer
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - A Gessner
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - B Schmidt
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - S Hempel
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - C P Jacob
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
| | - J Sanjuán
- CIBERSAM, Universitat de Valencia, Valencia, Spain
| | - M D Moltó
- CIBERSAM, Universitat de Valencia, Valencia, Spain
| | - K-P Lesch
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany; Comprehensive Heart Failure Center, University of Würzburg, Germany
| | - C M Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University of Frankfurt, Germany
| | - L Kent
- School of Medicine, University of St Andrews, Scotland, UK
| | - A Reif
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany; Comprehensive Heart Failure Center, University of Würzburg, Germany.
| |
Collapse
|
35
|
Cossenza M, Socodato R, Portugal CC, Domith ICL, Gladulich LFH, Encarnação TG, Calaza KC, Mendonça HR, Campello-Costa P, Paes-de-Carvalho R. Nitric oxide in the nervous system: biochemical, developmental, and neurobiological aspects. VITAMINS AND HORMONES 2014; 96:79-125. [PMID: 25189385 DOI: 10.1016/b978-0-12-800254-4.00005-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is a very reactive molecule, and its short half-life would make it virtually invisible until its discovery. NO activates soluble guanylyl cyclase (sGC), increasing 3',5'-cyclic guanosine monophosphate levels to activate PKGs. Although NO triggers several phosphorylation cascades due to its ability to react with Fe II in heme-containing proteins such as sGC, it also promotes a selective posttranslational modification in cysteine residues by S-nitrosylation, impacting on protein function, stability, and allocation. In the central nervous system (CNS), NO synthesis usually requires a functional coupling of nitric oxide synthase I (NOS I) and proteins such as NMDA receptors or carboxyl-terminal PDZ ligand of NOS (CAPON), which is critical for specificity and triggering of selected pathways. NO also modulates CREB (cAMP-responsive element-binding protein), ERK, AKT, and Src, with important implications for nerve cell survival and differentiation. Differences in the regulation of neuronal death or survival by NO may be explained by several mechanisms involving localization of NOS isoforms, amount of NO being produced or protein sets being modulated. A number of studies show that NO regulates neurotransmitter release and different aspects of synaptic dynamics, such as differentiation of synaptic specializations, microtubule dynamics, architecture of synaptic protein organization, and modulation of synaptic efficacy. NO has also been associated with synaptogenesis or synapse elimination, and it is required for long-term synaptic modifications taking place in axons or dendrites. In spite of tremendous advances in the knowledge of NO biological effects, a full description of its role in the CNS is far from being completely elucidated.
Collapse
Affiliation(s)
- Marcelo Cossenza
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Fisiologia e Farmacologia, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Renato Socodato
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Camila C Portugal
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Ivan C L Domith
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Luis F H Gladulich
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Thaísa G Encarnação
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Karin C Calaza
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Henrique R Mendonça
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Paula Campello-Costa
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Roberto Paes-de-Carvalho
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
36
|
Sharma NM, Llewellyn TL, Zheng H, Patel KP. Angiotensin II-mediated posttranslational modification of nNOS in the PVN of rats with CHF: role for PIN. Am J Physiol Heart Circ Physiol 2013; 305:H843-55. [PMID: 23832698 DOI: 10.1152/ajpheart.00170.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An increased sympathetic drive is an adverse characteristic in chronic heart failure (CHF). The protein expression of neuronal nitric oxide synthase (nNOS)- and hence nitric oxide (NO)-mediated sympathoinhibition is reduced in the paraventricular nucleus (PVN) of rats with CHF. However, the molecular mechanism(s) of nNOS downregulation remain(s) unclear. The aim of the study was to reveal the underlying molecular mechanism for the downregulation of nNOS in the PVN of CHF rats. Sprague-Dawley rats with CHF (6-8 wk after coronary artery ligation) demonstrated decreased nNOS dimer/monomer ratio (42%), with a concomitant increase in the expression of PIN (a protein inhibitor of nNOS known to dissociate nNOS dimers into monomers) by 47% in the PVN. Similarly, PIN expression is increased in a neuronal cell line (NG108) treated with angiotensin II (ANG II). Furthermore, there is an increased accumulation of high-molecular-weight nNOS-ubiquitin (nNOS-Ub) conjugates in the PVN of CHF rats (29%). ANG II treatment in NG108 cells in the presence of a proteasome inhibitor, lactacystin, also leads to a 69% increase in accumulation of nNOS-Ub conjugates immunoprecipitated by an antiubiquitin antibody. There is an ANG II-driven, PIN-mediated decrease in the dimeric catalytically active nNOS in the PVN, due to ubiquitin-dependent proteolytic degradation in CHF. Our results show a novel intermediary mechanism that leads to decreased levels of active nNOS in the PVN, involved in subsequent reduction in sympathoinhibition during CHF, offering a new target for the treatment of CHF and other cardiovascular diseases.
Collapse
Affiliation(s)
- Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | |
Collapse
|
37
|
Chang KC, Sasano T, Wang YC, Huang SKS. Nitric Oxide Synthase 1 Adaptor Protein, an Emerging New Genetic Marker for QT Prolongation and Sudden Cardiac Death. ACTA CARDIOLOGICA SINICA 2013; 29:217-225. [PMID: 27122710 PMCID: PMC4804833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 04/26/2013] [Indexed: 06/05/2023]
Abstract
UNLABELLED Sudden cardiac death (SCD) is defined as sudden unexplained death due to cardiac causes with an acute change in cardiovascular status within 1 hour of onset of symptoms. Alternatively, in unwitnessed cases, SCD can also be defined as a person last seen functionally normal 24 hours before being found dead. Despite significant advances in understanding the pathophysiology of cardiovascular diseases and the resultant improvement in resuscitation science, SCD remains a major healthcare challenge worldwide. Although the most pronounced risk factor for SCD is the presence of coronary artery disease in the setting of a depressed left ventricular function, most deaths occur in the larger, lower-risk subgroups where genetic variations and other conditions may be the precipitating factors in triggering SCD. Recently, a common genetic variation in a neuronal nitric oxide synthase regulator, nitric oxide synthase 1 adaptor protein (NOS1AP) also known as carboxyl-terminal PDZ ligand of neuronal nitric oxide synthase protein (CAPON) gene, has been identified as a new genetic marker in modulating QT interval prolongation and SCD in general populations. Animal study revealed that NOS1AP is expressed in the heart and interacts with NOS1-NO pathways to modulate cardiac repolarization via suppressing the sarcolemmal L-type calcium current and enhancing the IKr current. This important genetic implication was soon replicated in other racial/ethnic populations and extended to a variety of clinical settings including diabetes mellitus, coronary artery disease, myocardial infarction, and congenital or drug-induced long QT syndrome. The purpose of this review aims to provide up-to-date information about the emerging new genetic marker, NOS1AP, in relation to QT prolongation and SCD. KEY WORDS NOS1AP; QT interval; Sudden cardiac death.
Collapse
Affiliation(s)
- Kuan-Cheng Chang
- Division of Cardiology, Department of Medicine, China Medical University Hospital
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Tetsuo Sasano
- Department of Biofunctional Informatics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yu-Chen Wang
- Division of Cardiology, Department of Medicine, China Medical University Hospital
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Shoei K. Stephen Huang
- Section of Cardiac Electrophysiology and Pacing, Scott & White Healthcare, Texas A & M University College of Medicine, Temple, TX, USA
| |
Collapse
|
38
|
Markwardt ML, Nkobena A, Ding SY, Rizzo MA. Association with nitric oxide synthase on insulin secretory granules regulates glucokinase protein levels. Mol Endocrinol 2012; 26:1617-29. [PMID: 22771492 PMCID: PMC3434526 DOI: 10.1210/me.2012-1183] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 06/19/2012] [Indexed: 11/19/2022] Open
Abstract
Glucokinase (GCK) association with insulin-secretory granules is controlled by interaction with nitric oxide synthase (NOS) and is reversed by GCK S-nitrosylation. Nonetheless, the function of GCK sequestration on secretory granules is unknown. Here we report that the S-nitrosylation blocking V367M mutation prevents GCK accumulation on secretory granules by inhibiting association with NOS. Expression of this mutant is reduced compared with a second S-nitrosylation blocking GCK mutant (C371S) that accumulates to secretory granules and is expressed at levels greater than wild type. Even so, the rate of degradation for wild type and mutant GCK proteins were not significantly different from one another, and neither mutation disrupted the ability of GCK to be ubiquitinated. Furthermore, gene silencing of NOS reduced endogenous GCK content but did not affect β-actin content. Treatment of GCK(C371S) expressing cells with short interfering RNA specific for NOS also blocked accumulation of this protein to secretory granules and reduced expression levels to that of GCK(V367M). Conversely, cotransfection of catalytically inactive NOS increased GCK-mCherry levels. Expression of GCK(C371S) in βTC3 cells enhanced glucose metabolism compared with untransfected cells and cells expressing wild type GCK, even though this mutant has slightly reduced enzymatic activity in vitro. Finally, molecular dynamics simulations revealed that V367M induces conformational changes in GCK that are similar to S-nitrosylated GCK, thereby suggesting a mechanism for V367M-inhibition of NOS association. Our findings suggest that sequestration of GCK on secretory granules regulates cellular GCK protein content, and thus cellular GCK activity, by acting as a storage pool for GCK proteins.
Collapse
Affiliation(s)
- Michele L Markwardt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
39
|
Benabdesselam R, Dorbani-Mamine L, Benmessaoud-Mesbah O, Rendon A, Mhaouty-Kodja S, Hardin-Pouzet H. Dp71 gene disruption alters the composition of the dystrophin-associated protein complex and neuronal nitric oxide synthase expression in the hypothalamic supraoptic and paraventricular nuclei. J Endocrinol 2012; 213:239-49. [PMID: 22493004 DOI: 10.1530/joe-12-0066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
DP71 is the major cerebral dystrophin isoform and exerts its multiple functions via the dystrophin-associated protein complex (DAPC), also comprised of β-dystroglycan (β-DG) and α1-syntrophin (α1-Syn). Since DP71 disruption leads to impairment in the central control of the osmoregulatory axis, we investigated: 1) the DAPC composition in the hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) of Dp71-null mice; and 2) the expression and activity of neuronal nitric oxide synthase (nNOS), because it is a potential partner of the DAPC and a functional index of osmoregulatory axis activity. In wild-type mice, dystrophins and their autosomal homologs the utrophins, β-DG, and α1-Syn were localized in astrocyte end feet. In Dp71-null mice, the levels of β-DG and α1-Syn were lower and utrophin expression did not change. The location of the DAPC in astrocytic end feet suggests that it could be involved in hypothalamic osmosensitivity, which adapts the osmotic response. The altered composition of the DAPC in Dp71-null mice could thus explain why these mice manifest an hypo-osmolar status. In the SON and PVN neurons of Dp71-null mice, nNOS expression and activity were increased. Although we previously established that DP140 is expressed de novo in these neurons, the DAPC remained incomplete due to the low levels of β-DG and α1-Syn produced in these cells. Our data reveal the importance of DP71 for the constitution of a functional DAPC in the hypothalamus. Such DAPC disorganization may lead to modification of the microenvironment of the SON and PVN neurons and thus may result in a perturbed osmoregulation.
Collapse
Affiliation(s)
- Roza Benabdesselam
- Unité de Recherches, Faculté des Sciences Biologiques/UMMTO, BP 17, Tizi-Ouzou, Algeria
| | | | | | | | | | | |
Collapse
|
40
|
Anastas JN, Biechele TL, Robitaille M, Muster J, Allison KH, Angers S, Moon RT. A protein complex of SCRIB, NOS1AP and VANGL1 regulates cell polarity and migration, and is associated with breast cancer progression. Oncogene 2011; 31:3696-708. [PMID: 22179838 PMCID: PMC3419983 DOI: 10.1038/onc.2011.528] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
By analyzing public data sets of gene expression in human breast cancers we observed that increased levels of transcripts encoding the planar cell polarity (PCP) proteins SCRIB and VANGL1 correlate with increased risk of patient relapse. Experimentally, we found that reducing expression of SCRIB by short-hairpin RNAs (shRNAs) reduces the growth of human breast cancer cells in xenograft assays. To investigate SCRIB-associated proteins that might participate in the responses of breast cancer cells to altered levels of SCRIB, we used mass spectrometry and confocal microscopy. These studies reveal that SCRIB is present in at least two unique protein complexes: (1) a complex of SCRIB, ARHGEF, GIT and PAK (p21-activated kinase), and (2) a complex of SCRIB, NOS1AP and VANGL. Focusing on NOS1AP, we observed that NOS1AP colocalizes with both SCRIB and VANGL1 along cellular protrusions in metastatic breast cancer cells, but does not colocalize with either SCRIB or VANGL1 at cell junctions in normal breast cells. We investigated the effects of shRNA-mediated knockdown of NOS1AP and SCRIB in vitro, and found that reducing NOS1AP and SCRIB slows breast cancer cell migration and prevents the establishment of leading-trailing polarity. We also find that reduction of NOS1AP enhances anchorage-independent growth. Collectively these data point to the relevance of NOS1AP and SCRIB protein complexes in breast cancer.
Collapse
Affiliation(s)
- J N Anastas
- Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Sharma NM, Zheng H, Mehta PP, Li YF, Patel KP. Decreased nNOS in the PVN leads to increased sympathoexcitation in chronic heart failure: role for CAPON and Ang II. Cardiovasc Res 2011; 92:348-57. [PMID: 21831995 PMCID: PMC3193834 DOI: 10.1093/cvr/cvr217] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 07/01/2011] [Accepted: 08/05/2011] [Indexed: 12/29/2022] Open
Abstract
AIMS Previously, we showed an enhanced excitatory (N-methyl d-aspartate receptor-NR(1)) and decreased inhibitory neuronal nitric oxide (NO) synthase (nNOS) influence within the paraventricular nucleus (PVN) of rats with chronic heart failure (CHF). Although NR(1) and nNOS are normally linked, they can be disconnected by nNOS sequestering with nNOS-associated protein (CAPON). The aim of this study was to elucidate the underlying mechanism for the disconnection between increased expression of NR(1) and decreased nNOS in the PVN of rats with CHF which leads to enhanced sympathoexcitation. METHODS AND RESULTS CAPON expression was augmented while nNOS expression was decreased in the PVN of rats with CHF (6-8 weeks after left coronary artery ligation). Angiotensin II (Ang II) type I receptor (AT(1)) antagonist losartan (Los) treatment in rats with CHF reduced renal sympathetic nerve activity with concomitant normalization of protein expression of CAPON and nNOS in the PVN. Los treatment also reversed the blunting of endogenous NO-mediated sympatho-inhibition in rats with CHF. Moreover, Ang II-induced increase in CAPON expression in NG108 neuronal cells was also ameliorated by Los. CONCLUSION Blocking AT(1) receptors prevents the overexpression of CAPON and concomitant decrease in nNOS in the PVN, resulting in attenuation of sympathoexcitation commonly observed in CHF. Taken together, our data highlight the importance of altered expression and subsequent interaction of nNOS and CAPON within the PVN, leading to increased sympathoexcitation in CHF. Identifying this crucial nNOS/CAPON interaction regulated by AT(1) receptors may provide an important potential therapeutic target in CHF.
Collapse
Affiliation(s)
- Neeru M. Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Hong Zheng
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Parmender P. Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Yi-Fan Li
- Division of Basic Biomedical Science, College of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| |
Collapse
|
42
|
Mulvihill M, Guttman M, Komives EA. Protein interactions among Fe65, the low-density lipoprotein receptor-related protein, and the amyloid precursor protein. Biochemistry 2011; 50:6208-16. [PMID: 21650223 PMCID: PMC3139566 DOI: 10.1021/bi200508f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The adapter protein Fe65 has been proposed to be the link between the intracellular domains of the amyloid precursor protein, APP (AICD), and the low-density lipoprotein receptor-related protein (LRP-CT). Functional linkage between these two proteins has been established, and mutations within LRP-CT affect the amount of Aβ produced from APP. Previous work showed that AICD binds to protein interaction domain 2 (PID2) of Fe65. Although the structure of PID1 was determined recently, all attempts to demonstrate LRP-CT binding to this domain failed. We used biophysical experiments and binding studies to investigate the binding among these three proteins. Full-length Fe65 bound more weakly to AICD than did N-terminally truncated forms; however, the intramolecular domain-domain interactions that had been proposed to inhibit binding could not be observed using amide H-D exchange. Surprisingly, when LRP-CT is phosphorylated at Tyr4507, it bound to Fe65 PID1 despite the fact that this domain belongs to the Dab-like subclass of PIDs that are not supposed to be phosphorylation-dependent. Mutation of a critical arginine abolished binding, providing further proof of the phosphorylation dependence. Fe65 PID1 thus provides a link between the Dab-like class and the IRS-like class of PIDs and is the first Dab-like family member to show phosphorylation-dependent binding.
Collapse
Affiliation(s)
- Melinda Mulvihill
- Department of Chemistry and Biochemistry, University of California, San Diego La Jolla, CA 92093-0378
| | | | - Elizabeth A. Komives
- Department of Chemistry and Biochemistry, University of California, San Diego La Jolla, CA 92093-0378
| |
Collapse
|
43
|
Sandoval R, González A, Caviedes A, Pancetti F, Smalla KH, Kaehne T, Michea L, Gundelfinger ED, Wyneken U. Homeostatic NMDA receptor down-regulation via brain derived neurotrophic factor and nitric oxide-dependent signalling in cortical but not in hippocampal neurons. J Neurochem 2011; 118:760-72. [PMID: 21699542 DOI: 10.1111/j.1471-4159.2011.07365.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nitric oxide (NO) has been proposed to down-regulate NMDA receptors (NMDA-Rs) in a homeostatic manner. However, NMDA-R-dependent NO synthesis also can cause excitotoxic cell death. Using bicuculline-stimulated hippocampal and cortical cell cultures, we have addressed the role of the brain-derived neurotrophic factor-NO pathway in NMDA-R down-regulation. This pathway protected cortical cells from NMDA-induced death and led to NMDA-R inhibition. In contrast, no evidence was gained for the presence of this protective pathway in hippocampal neurons, in which NMDA-induced NO synthesis was confirmed to be toxic. Therefore, opposing effects of NO depended on the activation of different signalling pathways. The pathophysiological relevance of this observation was investigated in synaptosomes and post-synaptic densities isolated from rat hippocampi and cerebral cortices following kainic acid-induced status epilepticus. In cortical, but not in hippocampal synaptosomes, brain-derived neurotrophic factor induced NO synthesis and inhibited NMDA-R currents present in isolated post-synaptic densities. In conclusion, we identified a NO-dependent homeostatic response in the rat cerebral cortex induced by elevated activity. A low performance of this pathway in brain areas including the hippocampus may be related to their selective vulnerability in pathologies such as temporal lobe epilepsy.
Collapse
Affiliation(s)
- Rodrigo Sandoval
- Laboratorio de Neurotoxicología Ambiental, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Subramaniam S, Snyder SH. Huntington’s Disease is a disorder of the corpus striatum: Focus on Rhes (Ras homologue enriched in the striatum). Neuropharmacology 2011; 60:1187-92. [DOI: 10.1016/j.neuropharm.2010.10.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 10/20/2010] [Accepted: 10/26/2010] [Indexed: 11/30/2022]
|
45
|
|
46
|
Spanier KI, Leese F, Mayer C, Colbourne JK, Gilbert D, Pfrender ME, Tollrian R. Predator-induced defences in Daphnia pulex: selection and evaluation of internal reference genes for gene expression studies with real-time PCR. BMC Mol Biol 2010; 11:50. [PMID: 20587017 PMCID: PMC3148505 DOI: 10.1186/1471-2199-11-50] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 06/29/2010] [Indexed: 11/10/2022] Open
Abstract
Background The planktonic microcrustacean Daphnia pulex is among the best-studied animals in ecological, toxicological and evolutionary research. One aspect that has sustained interest in the study system is the ability of D. pulex to develop inducible defence structures when exposed to predators, such as the phantom midge larvae Chaoborus. The available draft genome sequence for D. pulex is accelerating research to identify genes that confer plastic phenotypes that are regularly cued by environmental stimuli. Yet for quantifying gene expression levels, no experimentally validated set of internal control genes exists for the accurate normalization of qRT-PCR data. Results In this study, we tested six candidate reference genes for normalizing transcription levels of D. pulex genes; alpha tubulin (aTub), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), TATA box binding protein (Tbp) syntaxin 16 (Stx16), X-box binding protein 1 (Xbp1) and CAPON, a protein associated with the neuronal nitric oxide synthase, were selected on the basis of an earlier study and from microarray studies. One additional gene, a matrix metalloproteinase (MMP), was tested to validate its transcriptional response to Chaoborus, which was earlier observed in a microarray study. The transcription profiles of these seven genes were assessed by qRT-PCR from RNA of juvenile D. pulex that showed induced defences in comparison to untreated control animals. We tested the individual suitability of genes for expression normalization using the programs geNorm, NormFinder and BestKeeper. Intriguingly, Xbp1, Tbp, CAPON and Stx16 were selected as ideal reference genes. Analyses on the relative expression level using the software REST showed that both classical housekeeping candidate genes (aTub and GAPDH) were significantly downregulated, whereas the MMP gene was shown to be significantly upregulated, as predicted. aTub is a particularly ill suited reference gene because five copies are found in the D. pulex genome sequence. When applying aTub for expression normalization Xbp1 and Tbp are falsely reported as significantly upregulated. Conclusions Our results suggest that the genes Xbp1, Tbp, CAPON and Stx16 are suitable reference genes for accurate normalization in qRT-PCR studies using Chaoborus-induced D. pulex specimens. Furthermore, our study underscores the importance of verifying the expression stability of putative reference genes for normalization of expression levels.
Collapse
Affiliation(s)
- Katina I Spanier
- Department of Animal Ecology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Corso-Díaz X, Krukoff TL. nNOS alpha and nNOS beta localization to aggresome-like inclusions is dependent on HSP90 activity. J Neurochem 2010; 114:864-72. [PMID: 20492351 DOI: 10.1111/j.1471-4159.2010.06813.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Nitric oxide (NO) is a highly reactive gas that participates in many physiological processes including neuroplasticity and neuronal survival. In brain neurons, NO is produced by two variants of neuronal nitric oxide synthase (nNOS), nNOSalpha and nNOSbeta. The activity of nNOSalpha is tightly regulated at the transcriptional and post-transcriptional levels. Heat shock protein 90 (HSP90) regulates nNOSalpha activity by facilitating heme insertion into the nNOSalpha monomer, resulting in increased NO production. HSP90 also regulates nNOSalpha degradation through the proteasome pathway. Here, we show in vitro that inhibition of HSP90 with geldanamycin increases nNOS mobility and induces formation of aggresome-like inclusions containing both nNOSalpha and nNOSbeta in primary cortical neurons. We also report the formation of endogenous nNOS-containing aggresome-like inclusions in healthy, untreated, mature primary cortical neurons. We propose that nNOS aggregation may be an additional mechanism for regulating nNOS activity, as has been proposed for inducible nitric oxide synthase. These findings reveal a new role for HSP90 in regulating nNOS sub-cellular localization and underscore the complexity of nNOS regulatory mechanisms.
Collapse
Affiliation(s)
- Ximena Corso-Díaz
- Center for Neuroscience, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
48
|
Richier L, Williton K, Clattenburg L, Colwill K, O'Brien M, Tsang C, Kolar A, Zinck N, Metalnikov P, Trimble WS, Krueger SR, Pawson T, Fawcett JP. NOS1AP associates with Scribble and regulates dendritic spine development. J Neurosci 2010; 30:4796-805. [PMID: 20357130 PMCID: PMC5123869 DOI: 10.1523/jneurosci.3726-09.2010] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 02/19/2010] [Accepted: 03/01/2010] [Indexed: 12/22/2022] Open
Abstract
The formation and function of the neuronal synapse is dependent on the asymmetric distribution of proteins both presynaptically and postsynaptically. Recently, proteins important in establishing cellular polarity have been implicated in the synapse. We therefore performed a proteomic screen with known polarity proteins and identified novel complexes involved in synaptic function. Specifically, we show that the tumor suppressor protein, Scribble, associates with neuronal nitric oxide synthase (nNOS) adaptor protein (NOS1AP) [also known as C-terminal PDZ ligand of nNOS (CAPON)] and is found both presynaptically and postsynaptically. The Scribble-NOS1AP association is direct and is mediated through the phosphotyrosine-binding (PTB) domain of NOS1AP and the fourth PDZ domain of Scribble. Further, we show that Scribble bridges NOS1AP to a beta-Pix [beta-p21-activated kinase (PAK)-interacting exchange factor]/Git1 (G-protein-coupled receptor kinase-interacting protein)/PAK complex. The overexpression of NOS1AP leads to an increase in dendritic protrusions, in a fashion that depends on the NOS1AP PTB domain. Consistent with these observations, both full-length NOS1AP and the NOS1AP PTB domain influence Rac activity. Together these data suggest that NOS1AP plays an important role in the mammalian synapse.
Collapse
Affiliation(s)
| | - Kelly Williton
- Samuel Lunenfeld Research Institute, Toronto, Ontario M5G 1X5, Canada
| | | | - Karen Colwill
- Samuel Lunenfeld Research Institute, Toronto, Ontario M5G 1X5, Canada
| | | | - Christopher Tsang
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Annette Kolar
- Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | | | - Pavel Metalnikov
- Samuel Lunenfeld Research Institute, Toronto, Ontario M5G 1X5, Canada
| | - William S. Trimble
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Stefan R. Krueger
- Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Tony Pawson
- Samuel Lunenfeld Research Institute, Toronto, Ontario M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada, and
| | | |
Collapse
|
49
|
Cui Z, Lv Q, Yan M, Cheng C, Guo Z, Yang J, Chen M, Xia Y, Zhang L, Shen A. Elevated expression of CAPON and neuronal nitric oxide synthase in the sciatic nerve of rats following constriction injury. Vet J 2010; 187:374-80. [PMID: 20202870 DOI: 10.1016/j.tvjl.2010.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 01/23/2010] [Accepted: 01/25/2010] [Indexed: 02/07/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) has been implicated in peripheral nerve lesions and regeneration. The CAPON adaptor protein interacts with the PDZ domain of nNOS, helping to regulate nNOS activity at post-synaptic sites in neurones, but it is not known whether its expression is altered in sciatic nerves after chronic nerve constriction injury. In the present study, the spatiotemporal expression of CAPON was determined in chronically constricted rat sciatic nerves. Similar to the level of protein expression, CAPON mRNA was significantly up-regulated for almost 5weeks following sciatic nerve injury. Immunohistochemistry demonstrated that increased CAPON was found mainly in S-100-positive Schwann cells. In addition, co-immunoprecipitation demonstrated an interaction between CAPON and nNOS in Schwann cells and the interaction was enhanced in injured sciatic nerves. CAPON may be involved in peripheral nerve regeneration through regulation of nNOS activity.
Collapse
Affiliation(s)
- Zhiming Cui
- The Second Hospital of Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jiang J, Yan M, Lv Q, Cheng C, Li X, Guo Z, Tao T, Shen A. Inhibition of nitric oxide-induced nuclear localization of CAPON by NMDA receptor antagonist in cultured rat primary astrocytes. Neurochem Int 2010; 56:561-568. [PMID: 20064573 DOI: 10.1016/j.neuint.2009.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 12/19/2009] [Accepted: 12/31/2009] [Indexed: 10/20/2022]
Abstract
Astrocytes play a key role in regulating aspects of inflammation in the central nervous system. It was observed that nNOS had located in the nucleus of cultured cerebral cortical astrocytes of 7 days. In the present study, we found that carboxy-terminal PDZ ligand of nNOS (CAPON) mainly located in the nucleus of astrocytes stimulated with NO donor sodium nitroprusside (SNP) or GSNO or N-methyl-d-aspartate (NMDA) receptor agonist-NMDA. However, originally, it was localized mostly in the cytoplasm of normal astrocytes. Immunocytochemistry showed that nNOS was co-localized with CAPON in the nucleus of astrocytes stimulated with SNP. In addition to the nuclear localization, treatment with SNP increased the mRNA and protein expression of CAPON. When SNP was removed from media, CAPON accumulated in nucleus transported back to cytoplasm. MK801, an inhibitor of NMDA receptor, was able to reverse the nuclear localization of CAPON resulted from SNP, suggesting that there is a functional relationship of NO with NMDA receptor in the regulation of the nuclear localization of CAPON. These findings provide a new insight in the understanding of the physical and pathological significances of CAPON/nNOS/NMDA receptor.
Collapse
Affiliation(s)
- Jing Jiang
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | | | | | | | | | | | | | | |
Collapse
|