1
|
Moreno-Fernández M, Luján V, Baliyan S, Poza C, Capellán R, de Las Heras-Martínez N, Morcillo MÁ, Oteo M, Ambrosio E, Ucha M, Higuera-Matas A. A Hidden Mark of a Troubled Past: Neuroimaging and Transcriptomic Analyses Reveal Interactive Effects of Maternal Immune Activation and Adolescent THC Exposure Suggestive of Increased Neuropsychiatric Risk. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100452. [PMID: 40115746 PMCID: PMC11925510 DOI: 10.1016/j.bpsgos.2025.100452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/04/2024] [Accepted: 01/12/2025] [Indexed: 03/23/2025] Open
Abstract
Background Maternal exposure to infections during gestation has been shown to predispose individuals to neuropsychiatric disorders. Additionally, clinical data suggest that cannabis use may trigger the onset of schizophrenia in vulnerable individuals. However, the direction of causality remains unclear. Methods To elucidate this issue, we utilized a rat model of maternal immune activation combined with exposure to increasing doses of Δ9-tetrahydrocannabinol during adolescence in both male and female rats. We investigated several behaviors in adulthood relevant for neuropsychiatric disorders, including impairments in working memory, deficits in sensorimotor gating, alterations in social behavior, anhedonia, and potential changes in implicit learning (conditioned taste aversion). Furthermore, we conducted a longitudinal positron emission tomography study to target affected brain regions and, subsequently, collected brain samples of one such region (the orbitofrontal cortex) for RNA sequencing analyses, which were also performed on peripheral blood mononuclear cells to identify peripheral biomarkers. Results While adolescent Δ9-tetrahydrocannabinol did not unmask latent behavioral disruptions, positron emission tomography scans revealed several brain alterations dependent on the combination of both hits. Additionally, the transcriptomic studies demonstrated that maternal immune activation affected dopaminergic, glutamatergic, and serotoninergic genes, with the combination of both exposures in most cases shifting the expression from downregulation to upregulation. In peripheral cells, interactive effects were observed on inflammatory pathways, and some genes were proposed as biomarkers. Conclusions These results suggest that the combination of these 2 vulnerability factors leaves a lasting mark on the body, potentially predisposing individuals to neuropsychiatric disorders even before behavioral alterations manifest.
Collapse
Affiliation(s)
- Mario Moreno-Fernández
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | - Víctor Luján
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
- National University of Distance Education International Graduate School (EIDUNED), Madrid, Spain
- Medical Application of Ionising Radiations Unit, Centre for Energy, Environmental and Technological Research (CIEMAT), Madrid, Spain
| | - Shishir Baliyan
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | - Celia Poza
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | - Roberto Capellán
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | | | - Miguel Ángel Morcillo
- Medical Application of Ionising Radiations Unit, Centre for Energy, Environmental and Technological Research (CIEMAT), Madrid, Spain
| | - Marta Oteo
- Medical Application of Ionising Radiations Unit, Centre for Energy, Environmental and Technological Research (CIEMAT), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | - Marcos Ucha
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, Faculty of Psychology, National University of Distance Education (UNED), Madrid, Spain
| |
Collapse
|
2
|
Turner-Ivey B, Jenkins DP, Carroll SL. Multiple Roles for Neuregulins and Their ERBB Receptors in Neurodegenerative Disease Pathogenesis and Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00119-1. [PMID: 40254133 DOI: 10.1016/j.ajpath.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
The role that neurotrophins, such as nerve growth factor, play in the pathogenesis of neurodegenerative diseases has long been appreciated. However, the neuregulin (NRG) family of growth factors and/or their v-erb-B2 avian erythroblastic leukemia viral oncogene homolog (ERBB) receptors have also been implicated in the pathogenesis of conditions, such as Alzheimer disease (AD), frontotemporal lobar degeneration (FTLD), and amyotrophic lateral sclerosis (ALS). In this review, we consider i) the structural variability of NRG isoforms generated by alternative RNA splicing, the use of multiple promoters and proteolysis, and the impact that this structural variability has on neuronal and glial physiology during development and adulthood. We discuss ii) the NRG receptors ERBB2, ERBB3, and ERBB4, how activation of each of these receptors further diversifies NRG actions in the central nervous system, and how dementia-related proteins, such as γ-secretase modulate the action of NRGs and their ERBB receptors. We then iii) turn to the abnormalities in NRG and ERBB expression and function evident in human AD and mouse AD models, how these abnormalities affect brain function, and attempts to use NRGs to treat AD. Finally, iv) we discuss NRG effects on the survival and function of neurons relevant to FTLD and ALS, alterations in NRG/ERBB signaling identified in these conditions, and the recent discovery of multiple human pedigrees in which autosomal dominant FTLD/ALS potentially results from point mutations in ERBB4.
Collapse
Affiliation(s)
- Brittany Turner-Ivey
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dorea P Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
3
|
Maciąg F, Chhikara A, Heine M. Calcium channel signalling at neuronal endoplasmic reticulum-plasma membrane junctions. Biochem Soc Trans 2024; 52:1617-1629. [PMID: 38934485 PMCID: PMC11668288 DOI: 10.1042/bst20230819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Neurons are highly specialised cells that need to relay information over long distances and integrate signals from thousands of synaptic inputs. The complexity of neuronal function is evident in the morphology of their plasma membrane (PM), by far the most intricate of all cell types. Yet, within the neuron lies an organelle whose architecture adds another level to this morphological sophistication - the endoplasmic reticulum (ER). Neuronal ER is abundant in the cell body and extends to distant axonal terminals and postsynaptic dendritic spines. It also adopts specialised structures like the spine apparatus in the postsynapse and the cisternal organelle in the axon initial segment. At membrane contact sites (MCSs) between the ER and the PM, the two membranes come in close proximity to create hubs of lipid exchange and Ca2+ signalling called ER-PM junctions. The development of electron and light microscopy techniques extended our knowledge on the physiological relevance of ER-PM MCSs. Equally important was the identification of ER and PM partners that interact in these junctions, most notably the STIM-ORAI and VAP-Kv2.1 pairs. The physiological functions of ER-PM junctions in neurons are being increasingly explored, but their molecular composition and the role in the dynamics of Ca2+ signalling are less clear. This review aims to outline the current state of research on the topic of neuronal ER-PM contacts. Specifically, we will summarise the involvement of different classes of Ca2+ channels in these junctions, discuss their role in neuronal development and neuropathology and propose directions for further research.
Collapse
Affiliation(s)
- Filip Maciąg
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Arun Chhikara
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| |
Collapse
|
4
|
Chien WH, Chen CH, Cheng MC, Wu YY, Gau SSF. Neuregulin 2 Is a Candidate Gene for Autism Spectrum Disorder. Int J Mol Sci 2024; 25:5547. [PMID: 38791584 PMCID: PMC11121989 DOI: 10.3390/ijms25105547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with heterogeneous and complex genetic underpinnings. Our previous microarray gene expression profiling identified significantly different neuregulin-2 gene (NRG2) expression between ASD patients and controls. Thus, we aimed to clarify whether NRG2 is a candidate gene associated with ASD. The study consisted of two stages. First, we used real-time quantitative PCR in 20 ASDs and 20 controls to confirm the microarray gene expression profiling results. The average NRG2 gene expression level in patients with ASD (3.23 ± 2.80) was significantly lower than that in the controls (9.27 ± 4.78, p < 0.001). Next, we conducted resequencing of all the exons of NRG2 in a sample of 349 individuals with ASD, aiming to identify variants of the NRG2 associated with ASD. We identified three variants, including two single nucleotide variants (SNVs), IVS3 + 13A > G (rs889022) and IVS10 + 32T > A (rs182642591), and one small deletion at exon 11 of NRG2 (delGCCCGG, rs933769137). Using data from the Taiwan Biobank as the controls, we found no significant differences in allele frequencies of rs889022 and rs182642591 between two groups. However, there is a significant difference in the genotype and allele frequency distribution of rs933769137 between ASDs and controls (p < 0.0001). The small deletion is located in the EGF-like domain at the C-terminal of the NRG2 precursor protein. Our findings suggest that NRG2 might be a susceptibility gene for ASD.
Collapse
Affiliation(s)
- Wei-Hsien Chien
- Department of Occupational Therapy, College of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-Y.W.)
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien 981, Taiwan;
| | - Yu-Yu Wu
- Department of Psychiatry, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-Y.W.)
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital, Taipei 10002, Taiwan
- Graduate Institute of Brain and Mind Sciences and Graduate of Clinical Medicine, National Taiwan University, Taipei 10002, Taiwan
| |
Collapse
|
5
|
Jiang A, Han K, Wei J, Su X, Wang R, Zhang W, Liu X, Qiao J, Liu P, Liu Q, Zhang J, Zhang N, Ge Y, Zhuang Y, Yu H, Wang S, Chen K, Lu W, Xu X, Yang H, Fan G, Dong B. Spatially resolved single-cell atlas of ascidian endostyle provides insight into the origin of vertebrate pharyngeal organs. SCIENCE ADVANCES 2024; 10:eadi9035. [PMID: 38552007 PMCID: PMC10980280 DOI: 10.1126/sciadv.adi9035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
The pharyngeal endoderm, an innovation of deuterostome ancestors, contributes to pharyngeal development by influencing the patterning and differentiation of pharyngeal structures in vertebrates; however, the evolutionary origin of the pharyngeal organs in vertebrates is largely unknown. The endostyle, a distinct pharyngeal organ exclusively present in basal chordates, represents a good model for understanding pharyngeal organ origins. Using Stereo-seq and single-cell RNA sequencing, we constructed aspatially resolved single-cell atlas for the endostyle of the ascidian Styela clava. We determined the cell composition of the hemolymphoid region, which illuminates a mixed ancestral structure for the blood and lymphoid system. In addition, we discovered a cluster of hair cell-like cells in zone 3, which has transcriptomic similarity with the hair cells of the vertebrate acoustico-lateralis system. These findings reshape our understanding of the pharynx of the basal chordate and provide insights into the evolutionary origin of multiplexed pharyngeal organs.
Collapse
Affiliation(s)
- An Jiang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Kai Han
- BGI Research, Qingdao 266555, China
| | - Jiankai Wei
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | | | - Rui Wang
- BGI Research, Qingdao 266555, China
| | - Wei Zhang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | | | - Jinghan Qiao
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Penghui Liu
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Qun Liu
- BGI Research, Qingdao 266555, China
| | - Jin Zhang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | | | - Yonghang Ge
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yuan Zhuang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Haiyan Yu
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shi Wang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Kai Chen
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Wange Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xun Xu
- BGI Research, Shenzhen 518083, China
| | | | - Guangyi Fan
- BGI Research, Qingdao 266555, China
- BGI Research, Shenzhen 518083, China
- Qingdao Key Laboratory of Marine Genomics BGI Research, Qingdao 266555, China
| | - Bo Dong
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
6
|
Stewart RG, Camacena M, Copits BA, Sack JT. Distinct cellular expression and subcellular localization of Kv2 voltage-gated K + channel subtypes in dorsal root ganglion neurons conserved between mice and humans. J Comp Neurol 2024; 532:e25575. [PMID: 38335058 PMCID: PMC10861167 DOI: 10.1002/cne.25575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/07/2023] [Accepted: 10/03/2023] [Indexed: 02/12/2024]
Abstract
The distinct organization of Kv2 voltage-gated potassium channels on and near the cell body of brain neurons enables their regulation of action potentials and specialized membrane contact sites. Somatosensory neurons have a pseudounipolar morphology and transmit action potentials from peripheral nerve endings through axons that bifurcate to the spinal cord and the cell body within ganglia including the dorsal root ganglia (DRG). Kv2 channels regulate action potentials in somatosensory neurons, yet little is known about where Kv2 channels are located. Here, we define the cellular and subcellular localization of the Kv2 paralogs, Kv2.1 and Kv2.2, in DRG somatosensory neurons with a panel of antibodies, cell markers, and genetically modified mice. We find that relative to spinal cord neurons, DRG neurons have similar levels of detectable Kv2.1 and higher levels of Kv2.2. In older mice, detectable Kv2.2 remains similar, while detectable Kv2.1 decreases. Both Kv2 subtypes adopt clustered subcellular patterns that are distinct from central neurons. Most DRG neurons co-express Kv2.1 and Kv2.2, although neuron subpopulations show preferential expression of Kv2.1 or Kv2.2. We find that Kv2 protein expression and subcellular localization are similar between mouse and human DRG neurons. We conclude that the organization of both Kv2 channels is consistent with physiological roles in the somata and stem axons of DRG neurons. The general prevalence of Kv2.2 in DRG as compared to central neurons and the enrichment of Kv2.2 relative to detectable Kv2.1 in older mice, proprioceptors, and axons suggest more widespread roles for Kv2.2 in DRG neurons.
Collapse
Affiliation(s)
- Robert G Stewart
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
| | - Miriam Camacena
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
| | - Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
7
|
Stewart RG, Camacena M, Copits BA, Sack JT. Distinct cellular expression and subcellular localization of Kv2 voltage-gated K + channel subtypes in dorsal root ganglion neurons conserved between mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530679. [PMID: 38187582 PMCID: PMC10769185 DOI: 10.1101/2023.03.01.530679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The distinct organization of Kv2 voltage-gated potassium channels on and near the cell body of brain neurons enables their regulation of action potentials and specialized membrane contact sites. Somatosensory neurons have a pseudounipolar morphology and transmit action potentials from peripheral nerve endings through axons that bifurcate to the spinal cord and the cell body within ganglia including the dorsal root ganglia (DRG). Kv2 channels regulate action potentials in somatosensory neurons, yet little is known about where Kv2 channels are located. Here we define the cellular and subcellular localization of the Kv2 paralogs, Kv2.1 and Kv2.2, in DRG somatosensory neurons with a panel of antibodies, cell markers, and genetically modified mice. We find that relative to spinal cord neurons, DRG neurons have similar levels of detectable Kv2.1, and higher levels of Kv2.2. In older mice, detectable Kv2.2 remains similar while detectable Kv2.1 decreases. Both Kv2 subtypes adopt clustered subcellular patterns that are distinct from central neurons. Most DRG neurons co-express Kv2.1 and Kv2.2, although neuron subpopulations show preferential expression of Kv2.1 or Kv2.2. We find that Kv2 protein expression and subcellular localization is similar between mouse and human DRG neurons. We conclude that the organization of both Kv2 channels is consistent with physiological roles in the somata and stem axons of DRG neurons. The general prevalence of Kv2.2 in DRG as compared to central neurons and the enrichment of Kv2.2 relative to detectable Kv2.1, in older mice, proprioceptors, and axons suggest more widespread roles for Kv2.2 in DRG neurons.
Collapse
Affiliation(s)
- Robert G Stewart
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Miriam Camacena
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
8
|
Dong B, Yue Y, Dong H, Wang Y. N-methyl-D-aspartate receptor hypofunction as a potential contributor to the progression and manifestation of many neurological disorders. Front Mol Neurosci 2023; 16:1174738. [PMID: 37396784 PMCID: PMC10308130 DOI: 10.3389/fnmol.2023.1174738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDA) are glutamate-gated ion channels critical for synaptic transmission and plasticity. A slight variation of NMDAR expression and function can result in devastating consequences, and both hyperactivation and hypoactivation of NMDARs are detrimental to neural function. Compared to NMDAR hyperfunction, NMDAR hypofunction is widely implicated in many neurological disorders, such as intellectual disability, autism, schizophrenia, and age-related cognitive decline. Additionally, NMDAR hypofunction is associated with the progression and manifestation of these diseases. Here, we review the underlying mechanisms of NMDAR hypofunction in the progression of these neurological disorders and highlight that targeting NMDAR hypofunction is a promising therapeutic intervention in some neurological disorders.
Collapse
Affiliation(s)
- Bin Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Yue
- School of Psychology, Northeast Normal University, Changchun, China
| | - Han Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Zhang Y, Zhu Y, Wang J, Jin L, Guo M, Chen L, Zhang L, Li Y, Wan B, Zhang R, Jia W, Hu C. Neuregulin4 Acts on Hypothalamic ErBb4 to Excite Oxytocin Neurons and Preserve Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204824. [PMID: 37060105 PMCID: PMC10238187 DOI: 10.1002/advs.202204824] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Neuregulin 4 (Nrg4) is an adipose tissue-enriched secreted factor that modulates glucose and lipid metabolism. Nrg4 is closely associated with obesity and preserves diet-induced metabolic disorders. However, the specific mechanisms via which Nrg4 regulates metabolic homeostasis remain incompletely understood. Here, this work finds that the Nrg4 receptor, ErbB4, is highly expressed in the hypothalamus, and the phosphorylation of hypothalamic ErbB4 is reduced in diet-induced obesity (DIO) mice. Peripheral Nrg4 can act on ErbB4 via blood circulation and excite neurons in the paraventricular nucleus of hypothalamus (PVN). Central administration of recombinant Nrg4 protein (rNrg4) reduces obesity and related metabolic disorders by influencing energy expenditure and intake. Overexpression of ErbB4 in the PVN protects against obesity, whereas its knock down in oxytocin (Oxt) neuron accelerates obesity. Furthermore, Nrg4-ErbB4 signaling excites Oxt release, and ablation of Oxt neuron considerably attenuates the effect of Nrg4 on energy balance. These data suggest that the hypothalamus is a key target of Nrg4, which partially explains the multifaceted roles of Nrg4 in metabolism.
Collapse
Affiliation(s)
- Yi Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Zhu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| | - Jinghui Wang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of EndocrinologyAffiliated Hospital of Nantong UniversityNantong226001P. R. China
| | - Li Jin
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Liwei Chen
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Lina Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Li
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Baocheng Wan
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Rong Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Weiping Jia
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Cheng Hu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| |
Collapse
|
10
|
Vullhorst D, Bloom MS, Akella N, Buonanno A. ER-PM Junctions on GABAergic Interneurons Are Organized by Neuregulin 2/VAP Interactions and Regulated by NMDA Receptors. Int J Mol Sci 2023; 24:2908. [PMID: 36769244 PMCID: PMC9917868 DOI: 10.3390/ijms24032908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Neuregulins (NRGs) signal via ErbB receptors to regulate neural development, excitability, synaptic and network activity, and behaviors relevant to psychiatric disorders. Bidirectional signaling between NRG2/ErbB4 and NMDA receptors is thought to homeostatically regulate GABAergic interneurons in response to increased excitatory neurotransmission or elevated extracellular glutamate levels. Unprocessed proNRG2 forms discrete clusters on cell bodies and proximal dendrites that colocalize with the potassium channel Kv2.1 at specialized endoplasmic reticulum-plasma membrane (ER-PM) junctions, and NMDA receptor activation triggers rapid dissociation from ER-PM junctions and ectodomain shedding by ADAM10. Here, we elucidate the mechanistic basis of proNRG2 clustering at ER-PM junctions and its regulation by NMDA receptors. Importantly, we demonstrate that proNRG2 promotes the formation of ER-PM junctions by directly binding the ER-resident membrane tether VAP, like Kv2.1. The proNRG2 intracellular domain harbors two non-canonical, low-affinity sites that cooperatively mediate VAP binding. One of these is a cryptic and phosphorylation-dependent VAP binding motif that is dephosphorylated following NMDA receptor activation, thus revealing how excitatory neurotransmission promotes the dissociation of proNRG2 from ER-PM junctions. Therefore, proNRG2 and Kv2.1 can independently function as VAP-dependent organizers of neuronal ER-PM junctions. Based on these and prior studies, we propose that proNRG2 and Kv2.1 serve as co-regulated downstream effectors of NMDA receptors to homeostatically regulate GABAergic interneurons.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
11
|
Watanabe K, Nakagawasai O, Kanno SI, Mitazaki S, Onogi H, Takahashi K, Watanabe KI, Tan-No K, Ishikawa M, Srivastava LK, Quirion R, Tadano T. Alterations in prefrontal cortical neuregulin-1 levels in post-pubertal rats with neonatal ventral hippocampal lesions. Front Behav Neurosci 2022; 16:1008623. [PMID: 36620856 PMCID: PMC9813588 DOI: 10.3389/fnbeh.2022.1008623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Genetic studies in humans have implicated the gene encoding neuregulin-1 (NRG-1) as a candidate susceptibility gene for schizophrenia. Furthermore, it has been suggested that NRG-1 is involved in regulating the expression and function of the N-methyl-D-aspartate receptor and the GABAA receptor in several brain areas, including the prefrontal cortex (PFC), the hippocampus, and the cerebellum. Neonatal ventral hippocampal lesioned (NVHL) rats have been considered as a putative model for schizophrenia with characteristic post-pubertal alteration in response to stress and neuroleptics. In this study, we examined NRG-1, erb-b2 receptor tyrosine kinase 4 (erbB4), and phospho-erbB4 (p-erbB4) levels in the PFC and the distribution of NRG-1 in the NVHL rats by using immunoblotting and immunohistochemical analyses. Neonatal lesions were induced by bilateral injection of ibotenic acid in the ventral hippocampus of postnatal day 7 Sprague-Dawley (SD)-rats. NVHL rats showed significantly decreased levels of NRG-1 and p-erbB4 in the PFC compared to sham controls at post-pubertal period, while the level of erbB4 did not differ between sham and NVHL rats. Moreover, microinjection of NRG-1 into the mPFC improved NVHL-induced prepulse inhibition deficits. Our study suggests PFC NRG-1 alteration as a potential mechanism in schizophrenia-like behaviors in the NVHL model.
Collapse
Affiliation(s)
- Kenya Watanabe
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,Department of Pharmacy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,*Correspondence: Osamu Nakagawasai,
| | - Syu-ichi Kanno
- Division of Clinical Pharmaceutical Therapy, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Satoru Mitazaki
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,Laboratory of Forensic Toxicology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Japan
| | - Hiroshi Onogi
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,Faculty of Health Science, Tohoku Fukushi University, Sendai, Japan
| | - Kohei Takahashi
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, Otawara, Tochigi, Japan
| | - Kei-ichiro Watanabe
- Center for Research on Counseling and Support Services, The University of Tokyo, Bunkyō-ku, Tokyo, Japan
| | - Koichi Tan-No
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Masaaki Ishikawa
- Division of Clinical Pharmaceutical Therapy, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | | | - Remi Quirion
- Douglas Hospital Research Centre, McGill University, Montreal, QC, Canada
| | - Takeshi Tadano
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan,Complementary and Alternative Medicine Clinical Research and Development, Graduate School of Medicine Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
12
|
Ahmad T, Vullhorst D, Chaudhuri R, Guardia CM, Chaudhary N, Karavanova I, Bonifacino JS, Buonanno A. Transcytosis and trans-synaptic retention by postsynaptic ErbB4 underlie axonal accumulation of NRG3. J Cell Biol 2022; 221:213222. [PMID: 35579602 PMCID: PMC9118086 DOI: 10.1083/jcb.202110167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 01/07/2023] Open
Abstract
Neuregulins (NRGs) are EGF-like ligands associated with cognitive disorders. Unprocessed proNRG3 is cleaved by BACE1 to generate the mature membrane-bound NRG3 ligand, but the subcellular site of proNRG3 cleavage, mechanisms underlying its transport into axons, and presynaptic accumulation remain unknown. Using an optogenetic proNRG3 cleavage reporter (LA143-NRG3), we investigate the spatial-temporal dynamics of NRG3 processing and sorting in neurons. In dark conditions, unprocessed LA143-NRG3 is retained in the trans-Golgi network but, upon photoactivation, is cleaved by BACE1 and released from the TGN. Mature NRG3 then emerges on the somatodendritic plasma membrane from where it is re-endocytosed and anterogradely transported on Rab4+ vesicles into axons via transcytosis. By contrast, the BACE1 substrate APP is sorted into axons on Rab11+ vesicles. Lastly, by a mechanism we denote "trans-synaptic retention," NRG3 accumulates at presynaptic terminals by stable interaction with its receptor ErbB4 on postsynaptic GABAergic interneurons. We propose that trans-synaptic retention may account for polarized expression of other neuronal transmembrane ligands and receptors.
Collapse
Affiliation(s)
- Tanveer Ahmad
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD,Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Rituparna Chaudhuri
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Haryana, India
| | - Carlos M. Guardia
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Nisha Chaudhary
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Irina Karavanova
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Juan S. Bonifacino
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD,Correspondence to Andres Buonanno:
| |
Collapse
|
13
|
Erben L, Welday JP, Cronin ME, Murphy R, Skirzewski M, Vullhorst D, Carroll SL, Buonanno A. Developmental, neurochemical, and behavioral analyses of ErbB4 Cyt-1 knockout mice. J Neurochem 2022; 161:435-452. [PMID: 35523590 PMCID: PMC9149141 DOI: 10.1111/jnc.15612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 01/26/2023]
Abstract
Neuregulins (NRGs) and their cognate neuronal receptor ERBB4, which is expressed in GABAergic and dopaminergic neurons, regulate numerous behaviors in rodents and have been identified as schizophrenia at-risk genes. ErbB4 transcripts are alternatively spliced to generate isoforms that either include (Cyt-1) or exclude (Cyt-2) exon 26, which encodes a cytoplasmic domain that imparts ErbB4 receptors the ability to signal via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. Although ErbB4 Cyt-1/2 isoforms have been studied in transfected cultured cells, their functions in vivo remain unknown. Here, we generated ErbB4-floxed (ErbB4-Cyt1fl/fl ) mice to investigate the effects of germline (constitutive) and conditional (acute) deletions of the Cyt-1 exon. Overall receptor mRNA levels remain unchanged in germline ErbB4 Cyt-1 knockouts (Cyt-1 KOs), with all transcripts encoding Cyt-2 variants. In contrast to mice lacking all ErbB4 receptor function, GABAergic interneuron migration and number are unaltered in Cyt-1 KOs. However, basal extracellular dopamine (DA) levels in the medial prefrontal cortex are increased in Cyt-1 heterozygotes. Despite these neurochemical changes, Cyt-1 heterozygous and homozygous mice do not manifest behavioral abnormalities previously reported to be altered in ErbB4 null mice. To address the possibility that Cyt-2 variants compensate for the lack of Cyt-1 during development, we microinjected an adeno-associated virus expressing Cre-recombinase (AAV-Cre) into the DA-rich ventral tegmental area of adult ErbB4-Cyt1fl/fl mice to acutely target exon 26. These conditional Cyt-1 KOs were found to exhibit behavioral abnormalities in the elevated plus maze and startle response, consistent with the idea that late exon 26 ablations may circumvent compensation by Cyt-2 variants. Taken together, our observations indicate that ErbB4 Cyt-1 function in vivo is important for DA balance and behaviors in adults.
Collapse
Affiliation(s)
- Larissa Erben
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Jacqueline P Welday
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Marie E Cronin
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Ricardo Murphy
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Miguel Skirzewski
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Pathway-specific contribution of parvalbumin interneuron NMDARs to synaptic currents and thalamocortical feedforward inhibition. Mol Psychiatry 2022; 27:5124-5134. [PMID: 36075962 PMCID: PMC9763122 DOI: 10.1038/s41380-022-01747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023]
Abstract
Prefrontal cortex (PFC) is a site of information convergence important for behaviors relevant to psychiatric disorders. Despite the importance of inhibitory GABAergic parvalbumin-expressing (PV+) interneurons to PFC circuit function and decades of interest in N-methyl-D-aspartate receptors (NMDARs) in these neurons, examples of defined circuit functions that depend on PV+ interneuron NMDARs have been elusive. Indeed, it remains controversial whether all PV+ interneurons contain functional NMDARs in adult PFC, which has major consequences for hypotheses of the pathogenesis of psychiatric disorders. Using a combination of fluorescent in situ hybridization, pathway-specific optogenetics, cell-type-specific gene ablation, and electrophysiological recordings from PV+ interneurons, here we resolve this controversy. We found that nearly 100% of PV+ interneurons in adult medial PFC (mPFC) express transcripts encoding GluN1 and GluN2B, and they have functional NMDARs. By optogenetically stimulating corticocortical and thalamocortical inputs to mPFC, we show that synaptic NMDAR contribution to PV+ interneuron EPSCs is pathway-specific, which likely explains earlier reports of PV+ interneurons without synaptic NMDAR currents. Lastly, we report a major contribution of NMDARs in PV+ interneurons to thalamus-mediated feedforward inhibition in adult mPFC circuits, suggesting molecular and circuit-based mechanisms for cognitive impairment under conditions of reduced NMDAR function. These findings represent an important conceptual advance that has major implications for hypotheses of the pathogenesis of psychiatric disorders.
Collapse
|
15
|
Ithal D, Sukumaran SK, Bhattacharjee D, Vemula A, Nadella R, Mahadevan J, Sud R, Viswanath B, Purushottam M, Jain S. Exome hits demystified: The next frontier. Asian J Psychiatr 2021; 59:102640. [PMID: 33892377 DOI: 10.1016/j.ajp.2021.102640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Severe mental illnesses such as schizophrenia and bipolar disorder have complex inheritance patterns, involving both common and rare variants. Whole exome sequencing is a promising approach to find out the rare genetic variants. We had previously reported several rare variants in multiplex families with severe mental illnesses. The current article tries to summarise the biological processes and pattern of expression of genes harbouring the aforementioned variants, linking them to known clinical manifestations through a methodical narrative review. Of the 28 genes considered for this review from 7 families with multiple affected individuals, 6 genes are implicated in various neuropsychiatric manifestations including some variations in the brain morphology assessed by magnetic resonance imaging. Another 15 genes, though associated with neuropsychiatric manifestations, did not have established brain morphological changes whereas the remaining 7 genes did not have any previously recorded neuropsychiatric manifestations at all. Wnt/b-catenin signaling pathway was associated with 6 of these genes and PI3K/AKT, calcium signaling, ERK, RhoA and notch signaling pathways had at least 2 gene associations. We present a comprehensive review of biological and clinical knowledge about the genes previously reported in multiplex families with severe mental illness. A 'disease in dish approach' can be helpful to further explore the fundamental mechanisms.
Collapse
Affiliation(s)
- Dhruva Ithal
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Salil K Sukumaran
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Debanjan Bhattacharjee
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Alekhya Vemula
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Ravi Nadella
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Jayant Mahadevan
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Reeteka Sud
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Biju Viswanath
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Meera Purushottam
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India.
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| |
Collapse
|
16
|
Neuregulin-4 Is Required for Maintaining Soma Size of Pyramidal Neurons in the Motor Cortex. eNeuro 2021; 8:ENEURO.0288-20.2021. [PMID: 33495243 PMCID: PMC8114872 DOI: 10.1523/eneuro.0288-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 11/21/2022] Open
Abstract
The regulation of neuronal soma size is essential for appropriate brain circuit function and its dysregulation is associated with several neurodevelopmental disorders. A defect in the dendritic growth and elaboration of motor neocortical pyramidal neurons in neonates lacking neuregulin-4 (NRG4) has previously been reported. In this study, we investigated whether the loss of NRG4 causes further morphologic defects that are specific to these neurons. We analyzed the soma size of pyramidal neurons of layer (L)2/3 and L5 of the motor cortex and a subpopulation of multipolar interneurons in this neocortical region in Nrg4+/+ and Nrg4−/− mice. There were significant decreases in pyramidal neuron soma size in Nrg4−/− mice compared with Nrg4+/+ littermates at all stages studied [postnatal day (P)10, P30, and P60]. The reduction was especially marked at P10 and in L5 pyramidal neurons. Soma size was not significantly different for multipolar interneurons at any age. This in vivo phenotype was replicated in pyramidal neurons cultured from Nrg4−/− mice and was rescued by NRG treatment. Analysis of a public single-cell RNA sequencing repository revealed discrete Nrg4 and Erbb4 expression in subpopulations of L5 pyramidal neurons, suggesting that the observed defects were due in part to loss of autocrine Nrg4/ErbB4 signaling. The pyramidal phenotype in the motor cortex of Nrg4−/− mice was associated with a lack of Rotarod test improvement in P60 mice, suggesting that absence of NRG4 causes alterations in motor performance.
Collapse
|
17
|
Deardorff AS, Romer SH, Fyffe RE. Location, location, location: the organization and roles of potassium channels in mammalian motoneurons. J Physiol 2021; 599:1391-1420. [DOI: 10.1113/jp278675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Adam S. Deardorff
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
- Department of Neurology and Internal Medicine, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| | - Shannon H. Romer
- Odyssey Systems Environmental Health Effects Laboratory, Navy Medical Research Unit‐Dayton Wright‐Patterson Air Force Base OH 45433 USA
| | - Robert E.W. Fyffe
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| |
Collapse
|
18
|
Ou GY, Lin WW, Zhao WJ. Neuregulins in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:662474. [PMID: 33897409 PMCID: PMC8064692 DOI: 10.3389/fnagi.2021.662474] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), are typically characterized by progressive neuronal loss and neurological dysfunctions in the nervous system, affecting both memory and motor functions. Neuregulins (NRGs) belong to the epidermal growth factor (EGF)-like family of extracellular ligands and they play an important role in the development, maintenance, and repair of both the central nervous system (CNS) and peripheral nervous system (PNS) through the ErbB signaling pathway. They also regulate multiple intercellular signal transduction and participate in a wide range of biological processes, such as differentiation, migration, and myelination. In this review article, we summarized research on the changes and roles of NRGs in neurodegenerative diseases, especially in AD. We elaborated on the structural features of each NRG subtype and roles of NRG/ErbB signaling networks in neurodegenerative diseases. We also discussed the therapeutic potential of NRGs in the symptom remission of neurodegenerative diseases, which may offer hope for advancing related treatment.
Collapse
Affiliation(s)
- Guan-yong Ou
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wen-wen Lin
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wei-jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Wei-jiang Zhao
| |
Collapse
|
19
|
Shi L, Bergson CM. Neuregulin 1: an intriguing therapeutic target for neurodevelopmental disorders. Transl Psychiatry 2020; 10:190. [PMID: 32546684 PMCID: PMC7297728 DOI: 10.1038/s41398-020-00868-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/14/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodevelopmental psychiatric disorders including schizophrenia (Sz) and attention deficit hyperactivity disorder (ADHD) are chronic mental illnesses, which place costly and painful burdens on patients, their families and society. In recent years, the epidermal growth factor (EGF) family member Neuregulin 1 (NRG1) and one of its receptors, ErbB4, have received considerable attention due to their regulation of inhibitory local neural circuit mechanisms important for information processing, attention, and cognitive flexibility. Here we examine an emerging body of work indicating that either decreasing NRG1-ErbB4 signaling in fast-spiking parvalbumin positive (PV+) interneurons or increasing it in vasoactive intestinal peptide positive (VIP+) interneurons could reactivate cortical plasticity, potentially making it a future target for gene therapy in adults with neurodevelopmental disorders. We propose preclinical studies to explore this model in prefrontal cortex (PFC), but also review the many challenges in pursuing cell type and brain-region-specific therapeutic approaches for the NRG1 system.
Collapse
Affiliation(s)
- Liang Shi
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Boulevard, Augusta, GA 30912 USA ,grid.189967.80000 0001 0941 6502Present Address: Department of Cell Biology, Emory University School of Medicine, Atlanta, GA USA
| | - Clare M. Bergson
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Boulevard, Augusta, GA 30912 USA
| |
Collapse
|
20
|
Neuregulins 1, 2, and 3 Promote Early Neurite Outgrowth in ErbB4-Expressing Cortical GABAergic Interneurons. Mol Neurobiol 2020; 57:3568-3588. [PMID: 32542595 DOI: 10.1007/s12035-020-01966-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022]
Abstract
The neuregulins (Nrgs 1-4) are a family of signaling molecules that play diverse roles in the nervous system. Nrg1 has been implicated in the formation of synapses and in synaptic plasticity. Previous studies have shown Nrg1 can affect neurite outgrowth in several neuronal populations, while the role of Nrg2 and Nrg3 in this process has remained understudied. The Nrgs can bind and activate the ErbB4 receptor tyrosine kinase which is preferentially expressed in GABAergic interneurons in the rodent hippocampus and cerebral cortex. In the present study, we evaluated the effects of Nrgs 1, 2, and 3 on neurite outgrowth of dissociated rat cortical ErbB4-positive (+)/GABA+ interneurons in vitro. All three Nrgs were able to promote neurite outgrowth during the first 2 days in vitro, with increases detected for both the axon (116-120%) and other neurites (100-120%). Increases in the average number of primary and secondary neurites were also observed. Treatment with the Nrgs for an additional 3 days promoted an increase in axonal length (86-96%), with only minimal effects on the remaining neurites (8-13%). ErbB4 expression persisted throughout the dendritic arbor and cell soma at all stages examined, while its expression in the axon was transient and declined with cell maturation. ErbB4 overexpression in GABAergic neurons promoted neurite outgrowth, an effect that was potentiated by Nrg treatment. These results show that Nrgs 1, 2, and 3 are each capable of influencing dendritic and axonal growth at early developmental stages in GABAergic neurons grown in vitro.
Collapse
|
21
|
ErbB4 Null Mice Display Altered Mesocorticolimbic and Nigrostriatal Dopamine Levels as well as Deficits in Cognitive and Motivational Behaviors. eNeuro 2020; 7:ENEURO.0395-19.2020. [PMID: 32354758 PMCID: PMC7242816 DOI: 10.1523/eneuro.0395-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Natural genetic variants of Neuregulin1 (NRG1) and its cognate receptor ErbB4 are associated with a risk for schizophrenia. Whereas most studies on NRG1-ErbB4 signaling have focused on GABAergic interneurons, ErbB4 is also expressed by midbrain dopaminergic neurons where it modulates extracellular dopamine (DA) levels. Here, we report that extracellular steady-state levels of DA are reduced in the medial prefrontal cortex (mPFC; −65%), hippocampus (−53%) and nucleus accumbens (NAc; −35%), but are elevated in the dorsal striatum (+25%) of ErbB4 knock-out mice (ErbB4 KOs) relative to wild-type controls. This pattern of DA imbalance recapitulates the reported prefrontal cortical reduction and striatal increase of DA levels in schizophrenia patients. Next, we report on a battery of behavioral tasks used to evaluate locomotor, cognitive and motivational behaviors in ErbB4 KOs relative to controls. We found that ErbB4 KOs are hyperactive in a novel open field but not in their familiar home cage, are more sensitive to amphetamine, perform poorly in the T-maze and novel object recognition (NOR) tasks, exhibit reduced spatial learning and memory on the Barnes maze, and perform markedly worse in conditioned place preference (CPP) tasks when associating cued-reward palatable food with location. However, we found that the poor performance of ErbB4 KOs in CPP are likely due to deficits in spatial memory, instead of reward seeking, as ErbB4 KOs are more motivated to work for palatable food rewards. Our findings indicate that ErbB4 signaling affects tonic DA levels and modulates a wide array of behavioral deficits relevant to psychiatric disorders, including schizophrenia.
Collapse
|
22
|
Paterson C, Cumming B, Law AJ. Temporal Dynamics of the Neuregulin-ErbB Network in the Murine Prefrontal Cortex across the Lifespan. Cereb Cortex 2020; 30:3325-3339. [PMID: 31897479 DOI: 10.1093/cercor/bhz312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Neuregulin-ErbB signaling is essential for numerous functions in the developing, adult, and aging brain, particularly in the prefrontal cortex (PFC). Mouse models with disrupted Nrg and/or ErbB genes are relevant to psychiatric, developmental, and age-related disorders, displaying a range of abnormalities stemming from cortical circuitry impairment. Many of these models display nonoverlapping phenotypes dependent upon the gene target and timing of perturbation, suggesting that cortical expression of the Nrg-ErbB network undergoes temporal regulation across the lifespan. Here, we report a comprehensive temporal expression mapping study of the Nrg-ErbB signaling network in the mouse PFC across postnatal development through aging. We find that Nrg and ErbB genes display distinct expression profiles; moreover, splice isoforms of these genes are differentially expressed across the murine lifespan. We additionally find a developmental switch in ErbB4 splice isoform expression potentially mediated through coregulation of the lncRNA Miat expression. Our results are the first to comprehensively and quantitatively map the expression patterns of the Nrg-ErbB network in the mouse PFC across the postnatal lifespan and may help disentangle the pathway's involvement in normal cortical sequences of events across the lifespan, as well as shedding light on the pathophysiological mechanisms of abnormal Nrg-ErbB signaling in neurological disease.
Collapse
Affiliation(s)
- Clare Paterson
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooke Cumming
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amanda J Law
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Cell and Developmental Biology, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Medicine, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
23
|
Czarnek M, Bereta J. Proteolytic Processing of Neuregulin 2. Mol Neurobiol 2019; 57:1799-1813. [PMID: 31838721 PMCID: PMC7118043 DOI: 10.1007/s12035-019-01846-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 11/25/2019] [Indexed: 01/17/2023]
Abstract
Neuregulin 2 (NRG2) belongs to the EGF family of growth factors. Most of this family members require proteolytic cleavage to liberate their ectodomains capable of binding and activating their cognate ErbB receptors. To date, most of the studies investigating proteolytic processing of neuregulins focused on NRG1, which was shown to undergo ectodomain shedding by several ADAM proteases and BACE1 and the remaining fragment was further cleaved by γ-secretase. Recently, NRG2 attracted more attention due to its role in the neurogenesis and modulation of behaviors associated with psychiatric disorders. In this study, we used genetic engineering methods to identify proteases involved in proteolytic processing of murine NRG2. Using non-neuronal cell lines as well as cultures of primary hippocampal neurons, we demonstrated that the major proteases responsible for releasing NRG2 ectodomain are ADAM10 and BACE2. Co-expression of NRG2 and BACE2 in neurons of certain brain structures including medulla oblongata and cerebellar deep nuclei was confirmed via immunohistochemical staining. The cleavage of NRG2 by ADAM10 or BACE2 generates a C-terminal fragment that serves as a substrate for γ-secretase. We also showed that murine NRG2 is subject to post-translational modifications, substantial glycosylation of its extracellular part, and phosphorylation of the cytoplasmic tail.
Collapse
Affiliation(s)
- Maria Czarnek
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
24
|
Nakazawa K, Sapkota K. The origin of NMDA receptor hypofunction in schizophrenia. Pharmacol Ther 2019; 205:107426. [PMID: 31629007 DOI: 10.1016/j.pharmthera.2019.107426] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptor (NMDAR) hypofunction plays a key role in pathophysiology of schizophrenia. Since NMDAR hypofunction has also been reported in autism, Alzheimer's disease and cognitive dementia, it is crucial to identify the location, timing, and mechanism of NMDAR hypofunction for schizophrenia for better understanding of disease etiology and for novel therapeutic intervention. In this review, we first discuss the shared underlying mechanisms of NMDAR hypofunction in NMDAR antagonist models and the anti-NMDAR autoantibody model of schizophrenia and suggest that NMDAR hypofunction could occur in GABAergic neurons in both models. Preclinical models using transgenic mice have shown that NMDAR hypofunction in cortical GABAergic neurons, in particular parvalbumin-positive fast-spiking interneurons, in the early postnatal period confers schizophrenia-related phenotypes. Recent studies suggest that NMDAR hypofunction can also occur in PV-positive GABAergic neurons with alterations of NMDAR-associated proteins, such as neuregulin/ErbB4, α7nAChR, and serine racemase. Furthermore, several environmental factors, such as oxidative stress, kynurenic acid and hypoxia, may also potentially elicit NMDAR hypofunction in GABAergic neurons in early postnatal period. Altogether, the studies discussed here support a central role for GABAergic abnormalities in the context of NMDAR hypofunction. We conclude by suggesting potential therapeutic strategies to improve the function of fast-spiking neurons.
Collapse
|
25
|
Johnson B, Leek AN, Tamkun MM. Kv2 channels create endoplasmic reticulum / plasma membrane junctions: a brief history of Kv2 channel subcellular localization. Channels (Austin) 2019; 13:88-101. [PMID: 30712450 PMCID: PMC6380216 DOI: 10.1080/19336950.2019.1568824] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The potassium channels Kv2.1 and Kv2.2 are widely expressed throughout the mammalian brain. Kv2.1 provides the majority of delayed rectifying current in rat hippocampus while both channels are differentially expressed in cortex. Particularly unusual is their neuronal surface localization pattern: while half the channel population is freely-diffusive on the plasma membrane as expected from the generalized Singer & Nicolson fluid mosaic model, the other half localizes into micron-sized clusters on the soma, dendrites, and axon initial segment. These clusters contain hundreds of channels, which for Kv2.1, are largely non-conducting. Competing theories of the mechanism underlying Kv2.1 clustering have included static tethering to being corralled by an actin fence. Now, recent work has demonstrated channel clustering is due to formation of endoplasmic reticulum/plasma membrane (ER/PM) junctions through interaction with ER-resident VAMP-associated proteins (VAPs). Interaction between surface Kv2 channels and ER VAPs groups channels together in clusters. ER/PM junctions play important roles in inter-organelle communication: they regulate ion flux, are involved in lipid transfer, and are sites of endo- and exocytosis. Kv2-induced ER/PM junctions are regulated through phosphorylation of the channel C-terminus which in turn regulates VAP binding, providing a rapid means to create or dismantle these microdomains. In addition, insults such as hypoxia or ischemia disrupt this interaction resulting in ER/PM junction disassembly. Kv2 channels are the only known plasma membrane protein to form regulated, injury sensitive junctions in this manner. Furthermore, it is likely that concentrated VAPs at these microdomains sequester additional interactors whose functions are not yet fully understood.
Collapse
Affiliation(s)
- Ben Johnson
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Ashley N Leek
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Michael M Tamkun
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA.,c Department of Biochemistry and Molecular Biology , Colorado State University , Fort Collins , CO , USA
| |
Collapse
|
26
|
Grieco SF, Wang G, Mahapatra A, Lai C, Holmes TC, Xu X. Neuregulin and ErbB expression is regulated by development and sensory experience in mouse visual cortex. J Comp Neurol 2019; 528:419-432. [PMID: 31454079 PMCID: PMC6901715 DOI: 10.1002/cne.24762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/22/2019] [Accepted: 08/14/2019] [Indexed: 01/14/2023]
Abstract
Neuregulins (NRGs) are protein ligands that impact neural development and circuit function. NRGs signal through the ErbB receptor tyrosine kinase family. NRG1/ErbB4 signaling in parvalbumin-expressing (PV) inhibitory interneurons is critical for visual cortical plasticity. There are multiple types of NRGs and ErbBs that can potentially contribute to visual cortical plasticity at different developmental stages. Thus, it is important to understand the normal developmental expression profiles of NRGs and ErbBs in specific neuron types in the visual cortex, and to study whether and how their expression changes in PV inhibitory neurons and excitatory neurons track with sensory perturbation. Cell type-specific translating ribosome affinity purification and qPCR was used to compare mRNA expression of nrg1,2,3,4 and erbB1,2,3,4 in PV and excitatory neurons in mouse visual cortex. We show that the expression of nrg1 and nrg3 decreases in PV neurons at the critical period peak, postnatal day 28 (P28) after monocular deprivation and dark rearing, and in the adult cortex (at P104) after 2-week long dark exposure. In contrast, nrg1 expression by excitatory neurons is unchanged at P28 and P104 following sensory deprivation, whereas nrg3 expression by excitatory neurons shows changes depending on the age and the mode of sensory deprivation. ErbB4 expression in PV neurons remains consistently high and does not appear to change in response to sensory deprivation. These data provide new important details of cell type-specific NRG/ErbB expression in the visual cortex and support that NRG1/ErbB4 signaling is implicated in both critical period and adult visual cortical plasticity.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
| | - Gina Wang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
| | - Ananya Mahapatra
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
| | - Cary Lai
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California.,Department of Biomedical Engineering, University of California, Irvine, California
| |
Collapse
|
27
|
Paramo B, Wyatt S, Davies AM. Neuregulin-4 Is Required for the Growth and Elaboration of Striatal Medium Spiny Neuron Dendrites. J Neuropathol Exp Neurol 2019; 78:725-734. [PMID: 31225596 PMCID: PMC6640913 DOI: 10.1093/jnen/nlz046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Medium spiny neurons (MSNs) comprise the vast majority of neurons in the striatum. Changes in the exuberant dendrites of these widely connected neurons are associated with a multitude of neurological conditions and are caused by a variety of recreational and medicinal drugs. However, we have a poor understanding of the physiological regulators of dendrite growth and elaboration of this clinically important population of neurons. Here, we show that MSN dendrites are markedly smaller and less branched in neonatal mice that possess a homozygous null mutation in the neuregulin-4 gene (Nrg4-/-) compared with wild type (Nrg4+/+) littermates. Nrg4-/- mice also had a highly significant reduction in MSN dendrite spine number in neonates and adults. The striking stunted dendrite arbor phenotype of MSNs observed in Nrg4-/- neonates was replicated in MSNs cultured from Nrg4-/- embryos and was completely rescued by soluble recombinant neuregulin-4. MSNs cultured from wild type mice coexpressed NRG4 and its receptor ErbB4. Our findings show that NRG4 is a major novel regulator of dendritic growth and arborization and spine formation in the striatum and suggest that it exerts its effects by an autocrine/paracrine mechanism.
Collapse
Affiliation(s)
- Blanca Paramo
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
28
|
Vullhorst D, Buonanno A. NMDA Receptors Regulate Neuregulin 2 Binding to ER-PM Junctions and Ectodomain Release by ADAM10 [corrected]. Mol Neurobiol 2019; 56:8345-8363. [PMID: 31240601 DOI: 10.1007/s12035-019-01659-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
Unprocessed pro-neuregulin 2 (pro-NRG2) accumulates on neuronal cell bodies at junctions between the endoplasmic reticulum and plasma membrane (ER-PM junctions). NMDA receptors (NMDARs) trigger NRG2 ectodomain shedding from these sites followed by activation of ErbB4 receptor tyrosine kinases, and ErbB4 signaling cell-autonomously downregulates intrinsic excitability of GABAergic interneurons by reducing voltage-gated sodium channel currents. NMDARs also promote dispersal of Kv2.1 clusters from ER-PM junctions and cause a hyperpolarizing shift in its voltage-dependent channel activation, suggesting that NRG2/ErbB4 and Kv2.1 work together to regulate intrinsic interneuron excitability in an activity-dependent manner. Here we explored the cellular processes underlying NMDAR-dependent NRG2 shedding in cultured rat hippocampal neurons. We report that NMDARs control shedding by two separate but converging mechanisms. First, NMDA treatment disrupts binding of pro-NRG2 to ER-PM junctions by post-translationally modifying conserved Ser/Thr residues in its intracellular domain. Second, using a mutant NRG2 protein that cannot be modified at these residues and that fails to accumulate at ER-PM junctions, we demonstrate that NMDARs also directly promote NRG2 shedding by ADAM-type metalloproteinases. Using pharmacological and shRNA-mediated knockdown, and metalloproteinase overexpression, we unexpectedly find that ADAM10, but not ADAM17/TACE, is the major NRG2 sheddase acting downstream of NMDAR activation. Together, these findings reveal how NMDARs exert tight control over the NRG2/ErbB4 signaling pathway, and suggest that NRG2 and Kv2.1 are co-regulated components of a shared pathway that responds to elevated extracellular glutamate levels.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
Salvany S, Casanovas A, Tarabal O, Piedrafita L, Hernández S, Santafé M, Soto-Bernardini MC, Calderó J, Schwab MH, Esquerda JE. Localization and dynamic changes of neuregulin-1 at C-type synaptic boutons in association with motor neuron injury and repair. FASEB J 2019; 33:7833-7851. [PMID: 30912977 DOI: 10.1096/fj.201802329r] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
C-type synaptic boutons (C-boutons) provide cholinergic afferent input to spinal cord motor neurons (MNs), which display an endoplasmic reticulum (ER)-related subsurface cistern (SSC) adjacent to their postsynaptic membrane. A constellation of postsynaptic proteins is clustered at C-boutons, including M2 muscarinic receptors, potassium channels, and σ-1 receptors. In addition, we previously found that neuregulin (NRG)1 is associated with C-boutons at postsynaptic SSCs, whereas its ErbB receptors are located in the presynaptic compartment. C-bouton-mediated regulation of MN excitability has been implicated in MN disease, but NRG1-mediated functions and the impact of various pathologic conditions on C-bouton integrity have not been studied in detail. Here, we investigated changes in C-boutons after electrical stimulation, pharmacological treatment, and peripheral nerve axotomy. SSC-linked NRG1 clusters were severely disrupted in acutely stressed MNs and after tunicamycin-induced ER stress. In axotomized MNs, C-bouton loss occurred in concomitance with microglial recruitment and was prevented by the ER stress inhibitor salubrinal. Activated microglia displayed a positive chemotaxis to C-boutons. Analysis of transgenic mice overexpressing NRG1 type I and type III isoforms in MNs indicated that NRG1 type III acts as an organizer of SSC-like structures, whereas NRG1 type I promotes synaptogenesis of presynaptic cholinergic terminals. Moreover, MN-derived NRG1 signals may regulate the activity of perineuronal microglial cells. Together, these data provide new insights into the molecular and cellular pathology of C-boutons in MN injury and suggest that distinct NRG1 isoform-mediated signaling functions regulate the complex matching between pre- and postsynaptic C-bouton elements.-Salvany, S., Casanovas, A., Tarabal, O., Piedrafita, L., Hernández, S., Santafé, M., Soto-Bernardini, M. C., Calderó, J., Schwab, M. H., Esquerda, J. E. Localization and dynamic changes of neuregulin-1 at C-type synaptic boutons in association with motor neuron injury and repair.
Collapse
Affiliation(s)
- Sara Salvany
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Sara Hernández
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Manuel Santafé
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Catalonia, Spain
| | - María Clara Soto-Bernardini
- Instituto Tecnológico de Costa Rica (TEC), Centro de Investigación en Biotecnología (CIB), Escuela de Biología, Cartago, Costa Rica
| | - Jordi Calderó
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Markus H Schwab
- Institute of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Josep E Esquerda
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| |
Collapse
|
30
|
Bygrave AM, Kilonzo K, Kullmann DM, Bannerman DM, Kätzel D. Can N-Methyl-D-Aspartate Receptor Hypofunction in Schizophrenia Be Localized to an Individual Cell Type? Front Psychiatry 2019; 10:835. [PMID: 31824347 PMCID: PMC6881463 DOI: 10.3389/fpsyt.2019.00835] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/21/2019] [Indexed: 01/07/2023] Open
Abstract
Hypofunction of N-methyl-D-aspartate glutamate receptors (NMDARs), whether caused by endogenous factors like auto-antibodies or mutations, or by pharmacological or genetic manipulations, produces a wide variety of deficits which overlap with-but do not precisely match-the symptom spectrum of schizophrenia. In order to understand how NMDAR hypofunction leads to different components of the syndrome, it is necessary to take into account which neuronal subtypes are particularly affected by it in terms of detrimental functional alterations. We provide a comprehensive overview detailing findings in rodent models with cell type-specific knockout of NMDARs. Regarding inhibitory cortical cells, an emerging model suggests that NMDAR hypofunction in parvalbumin (PV) positive interneurons is a potential risk factor for this disease. PV interneurons display a selective vulnerability resulting from a combination of genetic, cellular, and environmental factors that produce pathological multi-level positive feedback loops. Central to this are two antioxidant mechanisms-NMDAR activity and perineuronal nets-which are themselves impaired by oxidative stress, amplifying disinhibition. However, NMDAR hypofunction in excitatory pyramidal cells also produces a range of schizophrenia-related deficits, in particular maladaptive learning and memory recall. Furthermore, NMDAR blockade in the thalamus disturbs thalamocortical communication, and NMDAR ablation in dopaminergic neurons may provoke over-generalization in associative learning, which could relate to the positive symptom domain. Therefore, NMDAR hypofunction can produce schizophrenia-related effects through an action on various different circuits and cell types.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Kasyoka Kilonzo
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| | - Dimitri M Kullmann
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Dennis Kätzel
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
31
|
Konte B, Leicht G, Giegling I, Pogarell O, Karch S, Hartmann AM, Friedl M, Hegerl U, Rujescu D, Mulert C. A genome-wide association study of early gamma-band response in a schizophrenia case-control sample. World J Biol Psychiatry 2018; 19:602-609. [PMID: 28922980 DOI: 10.1080/15622975.2017.1366054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Disturbances in the gamma-frequency band of electroencephalography (EEG) measures are among the most consistently observed intermediate phenotypes in schizophrenia. We assessed whether genetic variations are associated with gamma-band activity. METHODS We performed a genome-wide association analysis of the early auditory evoked gamma-band response in schizophrenia affected subjects and healthy control individuals (in total N = 315). RESULTS No marker surpassed the threshold for genome-wide significant association. Several of the markers that were closest to significance mapped to genes involved in neuronal development and the Neuregulin-ErbB signalling network, such as NRG2 and KALRN. Using a gene-set enrichment analysis, we found suggestive evidence for association with genes involved in EEG abnormality (P = .048). CONCLUSIONS We identified no marker genome-wide significantly associating with gamma response; independent replication of the gene-set analysis result and larger sample sizes will be required to provide leads to cellular pathways involved in gamma-band activity.
Collapse
Affiliation(s)
- Bettina Konte
- a Department of Psychiatry, Psychotherapy and Psychosomatics , Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | - Gregor Leicht
- b Psychiatry Neuroimaging Branch, Imaging Center NeuroImage Nord and Department of Psychiatry and Psychotherapy , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Ina Giegling
- a Department of Psychiatry, Psychotherapy and Psychosomatics , Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | - Oliver Pogarell
- c Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University , Munich , Germany
| | - Susanne Karch
- c Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University , Munich , Germany
| | - Annette M Hartmann
- a Department of Psychiatry, Psychotherapy and Psychosomatics , Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | - Marion Friedl
- a Department of Psychiatry, Psychotherapy and Psychosomatics , Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | - Ulrich Hegerl
- d Department of Psychiatry and Psychotherapy , University of Leipzig , Leipzig , Germany
| | - Dan Rujescu
- a Department of Psychiatry, Psychotherapy and Psychosomatics , Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | - Christoph Mulert
- b Psychiatry Neuroimaging Branch, Imaging Center NeuroImage Nord and Department of Psychiatry and Psychotherapy , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
32
|
Rahman A, Weber J, Labin E, Lai C, Prieto AL. Developmental expression of Neuregulin‐3 in the rat central nervous system. J Comp Neurol 2018; 527:797-817. [DOI: 10.1002/cne.24559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/24/2018] [Accepted: 10/11/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Afrida Rahman
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| | - Janet Weber
- Department NeuroscienceUniversity of California San Diego San Diego California
| | - Edward Labin
- Department of NeurologyUniversity of Minnesota Minneapolis
| | - Cary Lai
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| | - Anne L Prieto
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| |
Collapse
|
33
|
Pafundo DE, Miyamae T, Lewis DA, Gonzalez-Burgos G. Presynaptic Effects of N-Methyl-D-Aspartate Receptors Enhance Parvalbumin Cell-Mediated Inhibition of Pyramidal Cells in Mouse Prefrontal Cortex. Biol Psychiatry 2018; 84:460-470. [PMID: 29523414 PMCID: PMC6068001 DOI: 10.1016/j.biopsych.2018.01.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 01/12/2018] [Accepted: 01/13/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Testing hypotheses regarding the role of N-methyl-D-aspartate receptor (NMDAR) hypofunction in schizophrenia requires understanding the mechanisms of NMDAR regulation of prefrontal cortex (PFC) circuit function. NMDAR antagonists are thought to produce pyramidal cell (PC) disinhibition. However, inhibitory parvalbumin-positive basket cells (PVBCs) have modest NMDAR-mediated excitatory drive and thus are unlikely to participate in NMDAR antagonist-mediated disinhibition. Interestingly, recent studies demonstrated that presynaptic NMDARs enhance transmitter release at central synapses. Thus, if presynaptic NMDARs enhance gamma-aminobutyric acid release at PVBC-to-PC synapses, they could participate in NMDAR-dependent PC disinhibition. Here, we examined whether presynaptic NMDAR effects could modulate gamma-aminobutyric acid release at PVBC-to-PC synapses in mouse PFC. METHODS Using whole-cell recordings from synaptically connected pairs in mouse PFC, we determined whether NMDA or NMDAR antagonist application affects PVBC-to-PC inhibition in a manner consistent with a presynaptic mechanism. RESULTS NMDAR activation enhanced by ∼40% the synaptic current at PVBC-to-PC pairs. This effect was consistent with a presynaptic mechanism given that it was 1) observed with postsynaptic NMDARs blocked by intracellular MK801, 2) associated with a lower rate of transmission failures and a higher transmitter release probability, and 3) blocked by intracellular MK801 in the PVBC. NMDAR antagonist application did not affect the synaptic currents in PVBC-to-PC pairs, but it reduced the inhibitory currents elicited in PCs with simultaneous glutamate release by extracellular stimulation. CONCLUSIONS We demonstrate that NMDAR activation enhances PVBC-to-PC inhibition in a manner consistent with presynaptic mechanisms, and we suggest that the functional impact of this presynaptic effect depends on the activity state of the PFC network.
Collapse
Affiliation(s)
- Diego E Pafundo
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Takeaki Miyamae
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Guillermo Gonzalez-Burgos
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
34
|
Chandrasekar A, Olde Heuvel F, Wepler M, Rehman R, Palmer A, Catanese A, Linkus B, Ludolph A, Boeckers T, Huber-Lang M, Radermacher P, Roselli F. The Neuroprotective Effect of Ethanol Intoxication in Traumatic Brain Injury Is Associated with the Suppression of ErbB Signaling in Parvalbumin-Positive Interneurons. J Neurotrauma 2018; 35:2718-2735. [PMID: 29774782 DOI: 10.1089/neu.2017.5270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.
Collapse
Affiliation(s)
| | | | - Martin Wepler
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Rida Rehman
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Annette Palmer
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Alberto Catanese
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Birgit Linkus
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Albert Ludolph
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Tobias Boeckers
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Peter Radermacher
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Francesco Roselli
- 1 Department of Neurology, Ulm University , Ulm, Germany .,4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| |
Collapse
|
35
|
Wehr MC, Hinrichs W, Brzózka MM, Unterbarnscheidt T, Herholt A, Wintgens JP, Papiol S, Soto-Bernardini MC, Kravchenko M, Zhang M, Nave KA, Wichert SP, Falkai P, Zhang W, Schwab MH, Rossner MJ. Spironolactone is an antagonist of NRG1-ERBB4 signaling and schizophrenia-relevant endophenotypes in mice. EMBO Mol Med 2018; 9:1448-1462. [PMID: 28743784 PMCID: PMC5653977 DOI: 10.15252/emmm.201707691] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Enhanced NRG1‐ERBB4 signaling is a risk pathway in schizophrenia, and corresponding mouse models display several endophenotypes of the disease. Nonetheless, pathway‐directed treatment strategies with clinically applicable compounds have not been identified. Here, we applied a cell‐based assay using the split TEV technology to screen a library of clinically applicable compounds to identify modulators of NRG1‐ERBB4 signaling for repurposing. We recovered spironolactone, known as antagonist of corticosteroids, as an inhibitor of the ERBB4 receptor and tested it in pharmacological and biochemical assays to assess secondary compound actions. Transgenic mice overexpressing Nrg1 type III display cortical Erbb4 hyperphosphorylation, a condition observed in postmortem brains from schizophrenia patients. Spironolactone treatment reverted hyperphosphorylation of activated Erbb4 in these mice. In behavioral tests, spironolactone treatment of Nrg1 type III transgenic mice ameliorated schizophrenia‐relevant behavioral endophenotypes, such as reduced sensorimotor gating, hyperactivity, and impaired working memory. Moreover, spironolactone increases spontaneous inhibitory postsynaptic currents in cortical slices supporting an ERBB4‐mediated mode‐of‐action. Our findings suggest that spironolactone, a clinically safe drug, provides an opportunity for new treatment options for schizophrenia.
Collapse
Affiliation(s)
- Michael C Wehr
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Wilko Hinrichs
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Magdalena M Brzózka
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Tilmann Unterbarnscheidt
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Hannover, Germany
| | | | | | - Sergi Papiol
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany.,Institute of Psychiatric Phenomics and Genomics (IPPG), Medical Center of the University of Munich, Munich, Germany
| | | | - Mykola Kravchenko
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Münster, Germany
| | - Mingyue Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Münster, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sven P Wichert
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter Falkai
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Münster, Germany
| | - Markus H Schwab
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hanover, Germany
| | - Moritz J Rossner
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilian University of Munich, Munich, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
36
|
Zhou L, Fisher ML, Cole RD, Gould TJ, Parikh V, Ortinski PI, Turner JR. Neuregulin 3 Signaling Mediates Nicotine-Dependent Synaptic Plasticity in the Orbitofrontal Cortex and Cognition. Neuropsychopharmacology 2018; 43:1343-1354. [PMID: 29114105 PMCID: PMC5916355 DOI: 10.1038/npp.2017.278] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/02/2017] [Accepted: 10/24/2017] [Indexed: 11/09/2022]
Abstract
Neuregulin 3 (NRG3) and ErbB4 have been linked to nicotine addiction; however, the neuronal mechanisms and behavioral consequences of NRG3-ErbB4 sensitivity to nicotine remain elusive. Recent literature suggests that relapse to smoking is due to a lack of impulsive control, which is thought to be due to altered functioning within the orbitofrontal cortex (OFC). Therefore, we examined circuitry changes within this structure following nicotine application. We report that nicotine controls synaptic plasticity in the OFC through NRG3/ErbB4-dependent regulation of GABAergic inhibition. We observed that both nicotine and NRG3 facilitated the conversion of long-term potentiation into long-term depression at cortical layer 3/5 synapses. Induction of long-term depression by nicotine relied on nicotinic receptor activation and key regulators of NRG3 signaling: (1) release of intracellular calcium, (2) activation of the BACE1 beta-secretase, and (3) ErbB4 receptor activation. Nicotine-induced synaptic plasticity was also associated with accumulation of intracellular GABA and was completely blocked by GABAA/GABAB antagonists. To test whether these mechanisms underlie OFC-dependent behavior, we evaluated the effects of nicotine in the go/no-go task. Nicotine-impaired stimulus discrimination in this task was rescued by pharmacologic disruption of the NRG3 receptor, ErbB4. Altogether, our data indicate that nicotine-induced synaptic plasticity in the OFC and cognitive changes depend on NRG3-ErbB4 signaling. We propose that nicotine activation of this pathway may contribute to nicotine addiction, particularly in individuals with genetic variation in NRG3.
Collapse
Affiliation(s)
- Luyi Zhou
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Miranda L Fisher
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Robert D Cole
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, USA
| | - Vinay Parikh
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Pavel I Ortinski
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Jill R Turner
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
37
|
Neuregulin-2 ablation results in dopamine dysregulation and severe behavioral phenotypes relevant to psychiatric disorders. Mol Psychiatry 2018; 23:1233-1243. [PMID: 28322273 PMCID: PMC5608621 DOI: 10.1038/mp.2017.22] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/16/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022]
Abstract
Numerous genetic and functional studies implicate variants of Neuregulin-1 (NRG1) and its neuronal receptor ErbB4 in schizophrenia and many of its endophenotypes. Although the neurophysiological and behavioral phenotypes of NRG1 mutant mice have been investigated extensively, practically nothing is known about the function of NRG2, the closest NRG1 homolog. We found that NRG2 expression in the adult rodent brain does not overlap with NRG1 and is more extensive than originally reported, including expression in the striatum and medial prefrontal cortex (mPFC), and therefore generated NRG2 knockout mice (KO) to study its function. NRG2 KOs have higher extracellular dopamine levels in the dorsal striatum but lower levels in the mPFC; a pattern with similarities to dopamine dysbalance in schizophrenia. Like ErbB4 KO mice, NRG2 KOs performed abnormally in a battery of behavioral tasks relevant to psychiatric disorders. NRG2 KOs exhibit hyperactivity in a novelty-induced open field, deficits in prepulse inhibition, hypersensitivity to amphetamine, antisocial behaviors, reduced anxiety-like behavior in the elevated plus maze and deficits in the T-maze alteration reward test-a task dependent on hippocampal and mPFC function. Acute administration of clozapine rapidly increased extracellular dopamine levels in the mPFC and improved alternation T-maze performance. Similar to mice treated chronically with N-methyl-d-aspartate receptor (NMDAR) antagonists, we demonstrate that NMDAR synaptic currents in NRG2 KOs are augmented at hippocampal glutamatergic synapses and are more sensitive to ifenprodil, indicating an increased contribution of GluN2B-containing NMDARs. Our findings reveal a novel role for NRG2 in the modulation of behaviors with relevance to psychiatric disorders.
Collapse
|
38
|
Kotzadimitriou D, Nissen W, Paizs M, Newton K, Harrison PJ, Paulsen O, Lamsa K. Neuregulin 1 Type I Overexpression Is Associated with Reduced NMDA Receptor-Mediated Synaptic Signaling in Hippocampal Interneurons Expressing PV or CCK. eNeuro 2018; 5:ENEURO.0418-17.2018. [PMID: 29740596 PMCID: PMC5938717 DOI: 10.1523/eneuro.0418-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/25/2018] [Accepted: 02/28/2018] [Indexed: 11/21/2022] Open
Abstract
Hypofunction of N-methyl-d-aspartate receptors (NMDARs) in inhibitory GABAergic interneurons is implicated in the pathophysiology of schizophrenia (SZ), a heritable disorder with many susceptibility genes. However, it is still unclear how SZ risk genes interfere with NMDAR-mediated synaptic transmission in diverse inhibitory interneuron populations. One putative risk gene is neuregulin 1 (NRG1), which signals via the receptor tyrosine kinase ErbB4, itself a schizophrenia risk gene. The type I isoform of NRG1 shows increased expression in the brain of SZ patients, and ErbB4 is enriched in GABAergic interneurons expressing parvalbumin (PV) or cholecystokinin (CCK). Here, we investigated ErbB4 expression and synaptic transmission in interneuronal populations of the hippocampus of transgenic mice overexpressing NRG1 type I (NRG1tg-type-I mice). Immunohistochemical analyses confirmed that ErbB4 was coexpressed with either PV or CCK in hippocampal interneurons, but we observed a reduced number of ErbB4-immunopositive interneurons in the NRG1tg-type-I mice. NMDAR-mediated currents in interneurons expressing PV (including PV+ basket cells) or CCK were reduced in NRG1tg-type-I mice compared to their littermate controls. We found no difference in AMPA receptor-mediated currents. Optogenetic activation (5 pulses at 20 Hz) of local glutamatergic fibers revealed a decreased NMDAR-mediated contribution to disynaptic GABAergic inhibition of pyramidal cells in the NRG1tg-type-I mice. GABAergic synaptic transmission from either PV+ or CCK+ interneurons, and glutamatergic transmission onto pyramidal cells, did not significantly differ between genotypes. The results indicate that synaptic NMDAR-mediated signaling in hippocampal interneurons is sensitive to chronically elevated NGR1 type I levels. This may contribute to the pathophysiological consequences of increased NRG1 expression in SZ.
Collapse
Affiliation(s)
| | - Wiebke Nissen
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Melinda Paizs
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| | - Kathryn Newton
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Karri Lamsa
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| |
Collapse
|
39
|
An essential role for neuregulin-4 in the growth and elaboration of developing neocortical pyramidal dendrites. Exp Neurol 2018; 302:85-92. [PMID: 29317193 PMCID: PMC5866123 DOI: 10.1016/j.expneurol.2018.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/12/2017] [Accepted: 01/02/2018] [Indexed: 01/05/2023]
Abstract
Neuregulins, with the exception of neuregulin-4 (NRG4), have been shown to be extensively involved in many aspects of neural development and function and are implicated in several neurological disorders, including schizophrenia, depression and bipolar disorder. Here we provide the first evidence that NRG4 has a crucial function in the developing brain. We show that both the apical and basal dendrites of neocortical pyramidal neurons are markedly stunted in Nrg4-/- neonates in vivo compared with Nrg4+/+ littermates. Neocortical pyramidal neurons cultured from Nrg4-/- embryos had significantly shorter and less branched neurites than those cultured from Nrg4+/+ littermates. Recombinant NRG4 rescued the stunted phenotype of embryonic neocortical pyramidal neurons cultured from Nrg4-/- mice. The majority of cultured wild type embryonic cortical pyramidal neurons co-expressed NRG4 and its receptor ErbB4. The difference between neocortical pyramidal dendrites of Nrg4-/- and Nrg4+/+ mice was less pronounced, though still significant, in juvenile mice. However, by adult stages, the pyramidal dendrite arbors of Nrg4-/- and Nrg4+/+ mice were similar, suggesting that compensatory changes in Nrg4-/- mice occur with age. Our findings show that NRG4 is a major novel regulator of dendritic arborisation in the developing cerebral cortex and suggest that it exerts its effects by an autocrine/paracrine mechanism.
Collapse
|
40
|
Skirzewski M, Karavanova I, Shamir A, Erben L, Garcia-Olivares J, Shin JH, Vullhorst D, Alvarez VA, Amara SG, Buonanno A. ErbB4 signaling in dopaminergic axonal projections increases extracellular dopamine levels and regulates spatial/working memory behaviors. Mol Psychiatry 2018; 23:2227-2237. [PMID: 28727685 PMCID: PMC5775946 DOI: 10.1038/mp.2017.132] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/13/2017] [Accepted: 04/04/2017] [Indexed: 02/07/2023]
Abstract
Genetic variants of Neuregulin 1 (NRG1) and its neuronal tyrosine kinase receptor ErbB4 are associated with risk for schizophrenia, a neurodevelopmental disorder characterized by excitatory/inhibitory imbalance and dopamine (DA) dysfunction. To date, most ErbB4 studies have focused on GABAergic interneurons in the hippocampus and neocortex, particularly fast-spiking parvalbumin-positive (PV+) basket cells. However, NRG has also been shown to modulate DA levels, suggesting a role for ErbB4 signaling in dopaminergic neuron function. Here we report that ErbB4 in midbrain DAergic axonal projections regulates extracellular DA levels and relevant behaviors. Mice lacking ErbB4 in tyrosine hydroxylase-positive (TH+) neurons, but not in PV+ GABAergic interneurons, exhibit different regional imbalances of basal DA levels and fail to increase DA in response to local NRG1 infusion into the dorsal hippocampus, medial prefrontal cortex and dorsal striatum measured by reverse microdialysis. Using Lund Human Mesencephalic (LUHMES) cells, we show that NRG/ErbB signaling increases extracellular DA levels, at least in part, by reducing DA transporter (DAT)-dependent uptake. Interestingly, TH-Cre;ErbB4f/f mice manifest deficits in learning, spatial and working memory-related behaviors, but not in numerous other behaviors altered in PV-Cre;ErbB4f/f mice. Importantly, microinjection of a Cre-inducible ErbB4 virus (AAV-ErbB4.DIO) into the mesencephalon of TH-Cre;ErbB4f/f mice, which selectively restores ErbB4 expression in DAergic neurons, rescues DA dysfunction and ameliorates behavioral deficits. Our results indicate that direct NRG/ErbB4 signaling in DAergic axonal projections modulates DA homeostasis, and that NRG/ErbB4 signaling in both GABAergic interneurons and DA neurons contribute to the modulation of behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- M Skirzewski
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - I Karavanova
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - A Shamir
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - L Erben
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA ,0000 0001 2240 3300grid.10388.32Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - J Garcia-Olivares
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - J H Shin
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - D Vullhorst
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - V A Alvarez
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - S G Amara
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - A Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
41
|
Neuregulin-Dependent Regulation of Fast-Spiking Interneuron Excitability Controls the Timing of the Critical Period. J Neurosci 2017; 36:10285-10295. [PMID: 27707966 DOI: 10.1523/jneurosci.4242-15.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Maturation of excitatory drive onto fast-spiking interneurons (FS INs) in the visual cortex has been implicated in the control of the timing of the critical period for ocular dominance plasticity. However, the mechanisms that regulate the strength of these synapses over cortical development are not understood. Here we use a mouse model to show that neuregulin (NRG) and the receptor tyrosine kinase erbB4 regulate the timing of the critical period. NRG1 enhanced the strength of excitatory synapses onto FS INs, which inhibited ocular dominance plasticity during the critical period but rescued plasticity in transgenics with hypoexcitable FS INs. Blocking the effects of endogenous neuregulin via inhibition of erbBs rescued ocular dominance plasticity in postcritical period adults, allowing recovery from amblyopia induced by chronic monocular deprivation. Thus, the strength of excitation onto FS INs is a key determinant of critical period plasticity and is maintained at high levels by NRG-erbB4 signaling to constrain plasticity in adulthood. SIGNIFICANCE STATEMENT Despite decades of experimentation, the mechanisms by which critical periods of enhanced synaptic plasticity are initiated and terminated are not completely understood. Here we show that neuregulin (NRG) and the receptor tyrosine kinase erbB4 determine critical period timing by controlling the strength of excitatory synapses onto FS INs. NRG1 enhanced excitatory drive onto fast spiking interneurons, which inhibited ocular dominance plasticity in juveniles but rescued plasticity in transgenics with hypoexcitable FS INs. Blocking the effects of endogenous neuregulin via inhibition of erbBs rescued ocular dominance plasticity in adults, allowing recovery from amblyopia induced by chronic monocular deprivation. Thus, in contrast to prevailing views of the termination of the critical period, active maintenance of strong excitation onto FS INs constrains plasticity in adults.
Collapse
|
42
|
Structural Similarities between Neuregulin 1-3 Isoforms Determine Their Subcellular Distribution and Signaling Mode in Central Neurons. J Neurosci 2017; 37:5232-5249. [PMID: 28432142 DOI: 10.1523/jneurosci.2630-16.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 03/14/2017] [Accepted: 04/09/2017] [Indexed: 11/21/2022] Open
Abstract
The Neuregulin (NRG) family of ErbB ligands is comprised of numerous variants originating from the use of different genes, alternative promoters, and splice variants. NRGs have generally been thought to be transported to axons and presynaptic terminals where they signal via ErbB3/4 receptors in paracrine or juxtacrine mode. However, we recently demonstrated that unprocessed pro-NRG2 accumulates on cell bodies and proximal dendrites, and that NMDAR activity is required for shedding of its ectodomain by metalloproteinases. Here we systematically investigated the subcellular distribution and processing of major NRG isoforms in rat hippocampal neurons. We show that NRG1 isotypes I and II, which like NRG2 are single-pass transmembrane proteins with an Ig-like domain, share the same subcellular distribution and ectodomain shedding properties. We furthermore show that NRG3, like CRD-NRG1, is a dual-pass transmembrane protein that harbors a second transmembrane domain near its amino terminus. Both NRG3 and CRD-NRG1 cluster on axons through juxtacrine interactions with ErbB4 present on GABAergic interneurons. Interestingly, although single-pass NRGs accumulate as unprocessed proforms, axonal puncta of CRD-NRG1 and NRG3 are comprised of processed protein. Mutations of CRD-NRG1 and NRG3 that render them resistant to BACE cleavage, as well as BACE inhibition, result in the loss of axonal puncta and in the accumulation of unprocessed proforms in neuronal soma. Together, these results define two groups of NRGs with distinct membrane topologies and fundamentally different targeting and processing properties in central neurons. The implications of this functional diversity for the regulation of neuronal processes by the NRG/ErbB pathway are discussed.SIGNIFICANCE STATEMENT Numerous Neuregulins (NRGs) are generated through the use of different genes, promoters, and alternative splicing, but the functional significance of this evolutionary conserved diversity remains poorly understood. Here we show that NRGs can be categorized by their membrane topologies. Single-pass NRGs, such as NRG1 Types I/II and NRG2, accumulate as unprocessed proforms on cell bodies, and their ectodomains are shed by metalloproteinases in response to NMDA receptor activation. By contrast, dual-pass CRD-NRG1 and NRG3 are constitutively processed by BACE and accumulate on axons where they interact with ErbB4 in juxtacrine mode. These findings reveal a previously unknown functional relationship between membrane topology, protein processing, and subcellular distribution, and suggest that single- and dual-pass NRGs regulate neuronal functions in fundamentally different ways.
Collapse
|
43
|
Iwakura Y, Wang R, Inamura N, Araki K, Higashiyama S, Takei N, Nawa H. Glutamate-dependent ectodomain shedding of neuregulin-1 type II precursors in rat forebrain neurons. PLoS One 2017; 12:e0174780. [PMID: 28350885 PMCID: PMC5370147 DOI: 10.1371/journal.pone.0174780] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 03/15/2017] [Indexed: 01/30/2023] Open
Abstract
The neurotrophic factor neuregulin 1 (NRG1) regulates neuronal development, glial differentiation, and excitatory synapse maturation. NRG1 is synthesized as a membrane-anchored precursor and is then liberated by proteolytic processing or exocytosis. Mature NRG1 then binds to its receptors expressed by neighboring neurons or glial cells. However, the molecular mechanisms that govern this process in the nervous system are not defined in detail. Here we prepared neuron-enriched and glia-enriched cultures from embryonic rat neocortex to investigate the role of neurotransmitters that regulate the liberation/release of NRG1 from the membrane of neurons or glial cells. Using a two-site enzyme immunoassay to detect soluble NRG1, we show that, of various neurotransmitters, glutamate was the most potent inducer of NRG1 release in neuron-enriched cultures. NRG1 release in glia-enriched cultures was relatively limited. Furthermore, among glutamate receptor agonists, N-Methyl-D-Aspartate (NMDA) and kainate (KA), but not AMPA or tACPD, mimicked the effects of glutamate. Similar findings were acquired from analysis of the hippocampus of rats with KA-induced seizures. To evaluate the contribution of members of a disintegrin and metalloproteinase (ADAM) families to NRG1 release, we transfected primary cultures of neurons with cDNA vectors encoding NRG1 types I, II, or III precursors, each tagged with the alkaline phosphatase reporter. Analysis of alkaline phosphatase activity revealed that the NRG1 type II precursor was subjected to tumor necrosis factor-α-converting enzyme (TACE) / a Disintegrin And Metalloproteinase 17 (ADAM17) -dependent ectodomain shedding in a protein kinase C-dependent manner. These results suggest that glutamatergic neurotransmission positively regulates the ectodomain shedding of NRG1 type II precursors and liberates the active NRG1 domain in an activity-dependent manner.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- * E-mail:
| | - Ran Wang
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Naoko Inamura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazuaki Araki
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular Genetics, Ehime University, Graduate School of Medicine, Ehime, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
44
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
45
|
Nakazawa K, Jeevakumar V, Nakao K. Spatial and temporal boundaries of NMDA receptor hypofunction leading to schizophrenia. NPJ SCHIZOPHRENIA 2017; 3:7. [PMID: 28560253 PMCID: PMC5441533 DOI: 10.1038/s41537-016-0003-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022]
Abstract
The N-methyl-d-aspartate receptor hypofunction is one of the most prevalent models of schizophrenia. For example, healthy subjects treated with uncompetitive N-methyl-d-aspartate receptor antagonists elicit positive, negative, and cognitive-like symptoms of schizophrenia. Patients with anti-N-methyl-d-aspartate receptor encephalitis, which is likely caused by autoantibody-mediated down-regulation of cell surface N-methyl-d-aspartate receptors, often experience psychiatric symptoms similar to schizophrenia initially. However, where and when N-methyl-d-aspartate receptor hypofunction occurs in the brain of schizophrenic patients is poorly understood. Here we review the findings from N-methyl-d-aspartate receptor antagonist and autoantibody models, postmortem studies on N-methyl-d-aspartate receptor subunits, as well as the global and cell-type-specific knockout mouse models of subunit GluN1. We compare various conditional GluN1 knockout mouse strains, focusing on the onset of N-methyl-d-aspartate receptor deletion and on the cortical cell-types. Based on these results, we hypothesize that N-methyl-d-aspartate receptor hypofunction initially occurs in cortical GABAergic neurons during early postnatal development. The resulting GABA neuron maturation deficit may cause reduction of intrinsic excitability and GABA release, leading to disinhibition of pyramidal neurons. The cortical disinhibition in turn could elicit glutamate spillover and subsequent homeostatic down regulation of N-methyl-d-aspartate receptor function in pyramidal neurons in prodromal stage. These two temporally-distinct N-methyl-d-aspartate receptor hypofunctions may be complimentary, as neither alone may not be able to fully explain the entire schizophrenia pathophysiology. Potential underlying mechanisms for N-methyl-d-aspartate receptor hypofunction in cortical GABA neurons are also discussed, based on studies of naturally-occurring N-methyl-d-aspartate receptor antagonists, neuregulin/ErbB4 signaling pathway, and theoretical analysis of excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Kazu Nakazawa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Vivek Jeevakumar
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Kazuhito Nakao
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
46
|
Casanovas A, Salvany S, Lahoz V, Tarabal O, Piedrafita L, Sabater R, Hernández S, Calderó J, Esquerda JE. Neuregulin 1-ErbB module in C-bouton synapses on somatic motor neurons: molecular compartmentation and response to peripheral nerve injury. Sci Rep 2017; 7:40155. [PMID: 28065942 PMCID: PMC5220293 DOI: 10.1038/srep40155] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/02/2016] [Indexed: 12/30/2022] Open
Abstract
The electric activity of lower motor neurons (MNs) appears to play a role in determining cell-vulnerability in MN diseases. MN excitability is modulated by cholinergic inputs through C-type synaptic boutons, which display an endoplasmic reticulum-related subsurface cistern (SSC) adjacent to the postsynaptic membrane. Besides cholinergic molecules, a constellation of proteins involved in different signal-transduction pathways are clustered at C-type synaptic sites (M2 muscarinic receptors, Kv2.1 potassium channels, Ca2+ activated K+ [SK] channels, and sigma-1 receptors [S1R]), but their collective functional significance so far remains unknown. We have previously suggested that neuregulin-1 (NRG1)/ErbBs-based retrograde signalling occurs at this synapse. To better understand signalling through C-boutons, we performed an analysis of the distribution of C-bouton-associated signalling proteins. We show that within SSC, S1R, Kv2.1 and NRG1 are clustered in highly specific, non-overlapping, microdomains, whereas ErbB2 and ErbB4 are present in the adjacent presynaptic compartment. This organization may define highly ordered and spatially restricted sites for different signal-transduction pathways. SSC associated proteins are disrupted in axotomised MNs together with the activation of microglia, which display a positive chemotactism to C-bouton sites. This indicates that C-bouton associated molecules are also involved in neuroinflammatory signalling in diseased MNs, emerging as new potential therapeutic targets.
Collapse
Affiliation(s)
- Anna Casanovas
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Salvany
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Víctor Lahoz
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Olga Tarabal
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Raimundo Sabater
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Hernández
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Jordi Calderó
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Josep E. Esquerda
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| |
Collapse
|
47
|
Proteolytic processing of Neuregulin-1. Brain Res Bull 2016; 126:178-182. [PMID: 27393467 DOI: 10.1016/j.brainresbull.2016.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 06/28/2016] [Accepted: 07/03/2016] [Indexed: 02/07/2023]
Abstract
Neuregulin-1 (NRG1), known also as heregulin, acetylcholine receptor inducing activity (ARIA), glial growth factor (GGF), or sensory and motor neuron derived factor (SMDF), is a key factor for many developmental processes and in adult brain. All known splice variants contain an epidermal growth factor (EGF)-like domain, which is mediating signaling via receptors of the ErbB family. In particular, NRG1 acts as an essential signaling molecule expressed on the axonal surface, where it signals to Schwann cells throughout development and regulates the thickness of the myelin sheath. NRG1 is required also by other cell types in the nervous system, for instance as an axonal signal released by proprioceptive afferents to induce development of the muscle spindle, and it controls aspects of cortical interneuron development as well as the formation of thalamo-cortical projections. The precursor protein of NRG1 can be activated and released from the membrane through limited proteolysis by the β-Secretase (β-site amyloid precursor protein cleaving enzyme 1, BACE1) which was first identified through its function as the rate limiting enzyme of amyloid-β-peptide (Aβ) production. Aβ is the major component of amyloid plaques in Alzheimer's disease (AD). Due to the hairpin nature of NRG1 type III two membrane-bound stubs with a type 1 and a type 2 orientation are generated by an initial proteolytic cleavage and successive release of the EGF-like domain either by dual cleavage by BACE1 or by ADAM17 (a disintegrin and metalloprotease) which is also called TACE (Tumor Necrosis Factor-α-converting enzyme). The cleavages activate NRG1 to allow juxtacrine or paracrine signaling. The type 1 oriented stub is further cleaved by γ-secretase in the transmembrane domain with a putative role in intracellular domain (ICD) signaling, while the type II oriented stub is cleaved by signal peptidase like proteases (SPPLs). Neuregulin-1 was identified as a major physiological substrate of BACE1 during early postnatal development when similarities in BACE1 KO mice and NRG1 heterozygous mice were discovered. Both display severe hypomyelination of peripheral nerves. Later it was shown with genetic and pharmacological evidence that the developmental effect of type I NRG1 on the formation and the maintenance of muscle spindles is BACE1 dependent. Thus, NRG1 functions in PNS and CNS are likely to set limits to an Alzheimer disease therapy with relatively strong BACE1 inhibition.
Collapse
|
48
|
Loos M, Schetters D, Hoogeland M, Spijker S, de Vries TJ, Pattij T. Prefrontal cortical neuregulin-ErbB modulation of inhibitory control in rats. Eur J Pharmacol 2016; 781:157-63. [DOI: 10.1016/j.ejphar.2016.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/13/2022]
|
49
|
Hu X, Fan Q, Hou H, Yan R. Neurological dysfunctions associated with altered BACE1-dependent Neuregulin-1 signaling. J Neurochem 2016; 136:234-49. [PMID: 26465092 PMCID: PMC4833723 DOI: 10.1111/jnc.13395] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Inhibition of BACE1 is being pursued as a therapeutic target to treat patients suffering from Alzheimer's disease because BACE1 is the sole β-secretase that generates β-amyloid peptide. Knowledge regarding other cellular functions of BACE1 is therefore critical for the safe use of BACE1 inhibitors in human patients. Neuregulin-1 (Nrg1) is a BACE1 substrate and BACE1 cleavage of Nrg1 is critical for signaling functions in myelination, remyelination, synaptic plasticity, normal psychiatric behaviors, and maintenance of muscle spindles. This review summarizes the most recent discoveries associated with BACE1-dependent Nrg1 signaling in these areas. This body of knowledge will help to provide guidance for preventing unwanted Nrg1-based side effects following BACE1 inhibition in humans. To initiate its signaling cascade, membrane anchored Neuregulin (Nrg), mainly type I and III β1 Nrg1 isoforms and Nrg3, requires ectodomain shedding. BACE1 is one of such indispensable sheddases to release the functional Nrg signaling fragment. The dependence of Nrg on the cleavage by BACE1 is best manifested by disrupting the critical role of Nrg in the control of axonal myelination, schizophrenic behaviors as well as the formation and maintenance of muscle spindles.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|