1
|
Berrocal-Rubio MA, Pawer YDJ, Dinevska M, De Paoli-Iseppi R, Widodo SS, Gleeson J, Rajab N, De Nardo W, Hallab J, Li A, Mantamadiotis T, Clark MB, Wells CA. Discovery of NRG1-VII: the myeloid-derived class of NRG1. BMC Genomics 2024; 25:814. [PMID: 39210279 PMCID: PMC11360300 DOI: 10.1186/s12864-024-10723-2] [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: 01/30/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The growth factor Neuregulin-1 (NRG1) has pleiotropic roles in proliferation and differentiation of the stem cell niche in different tissues. It has been implicated in gut, brain and muscle development and repair. Six isoform classes of NRG1 and over 28 protein isoforms have been previously described. Here we report a new class of NRG1, designated NRG1-VII to denote that these NRG1 isoforms arise from a myeloid-specific transcriptional start site (TSS) previously uncharacterized. Long-read sequencing was used to identify eight high-confidence NRG1-VII transcripts. These transcripts presented major structural differences from one another, through the use of cassette exons and alternative stop codons. Expression of NRG1-VII was confirmed in primary human monocytes and tissue resident macrophages and induced pluripotent stem cell-derived macrophages (iPSC-derived macrophages). Isoform switching via cassette exon usage and alternate polyadenylation was apparent during monocyte maturation and macrophage differentiation. NRG1-VII is the major class expressed by the myeloid lineage, including tissue-resident macrophages. Analysis of public gene expression data indicates that monocytes and macrophages are a primary source of NRG1. The size and structure of class VII isoforms suggests that they may be more diffusible through tissues than other NRG1 classes. However, the specific roles of class VII variants in tissue homeostasis and repair have not yet been determined.
Collapse
Affiliation(s)
- Miguel A Berrocal-Rubio
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Yair David Joseph Pawer
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Marija Dinevska
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | - Ricardo De Paoli-Iseppi
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Samuel S Widodo
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | - Josie Gleeson
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Nadia Rajab
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Will De Nardo
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Jeannette Hallab
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Anran Li
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
- Department of Microbiology and Immunology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Michael B Clark
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Christine A Wells
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
2
|
Liu J, Geraghty JR, Schram S, Cropper HC, Lei J, Loeb JA, Song F. Prevention of Alzheimer Pathology by Blocking Neuregulin Signaling on Microglia. eNeuro 2023; 10:ENEURO.0422-23.2023. [PMID: 37903620 PMCID: PMC10644371 DOI: 10.1523/eneuro.0422-23.2023] [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: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Plaque formation, microglial activation, and synaptic loss are pathologic hallmarks of Alzheimer's disease; however, removing plaques has had little clinical benefit. Here, we show that neuregulin-1, a glial growth factor, induces inflammatory cytokines and promotes phagocytic activity in vitro and augments microglial activation and plaque formation in 5XFAD Alzheimer's mice. Brain-specific targeting of neuregulin-1 by intraventricular delivery of a novel neuregulin-1 fusion protein antagonist, GlyB4, significantly alters microglial morphology and function to a nonpathogenic morphology in early-stage 5XFAD mice and prevents plaques from forming. Once plaques have already formed, GlyB4 reduces new plaque formation and prevents synaptic loss. Selective, targeted disruption of neuregulin-1 signaling on brain microglia with GlyB4 could be a novel "upstream" approach to slow or stop disease progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Jianguo Liu
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Joseph R Geraghty
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Sarah Schram
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Haley C Cropper
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Justin Lei
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jeffrey A Loeb
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| | - Fei Song
- Department of Neurology and Rehabilitation, The University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
3
|
Vincent B, Maitra S. BACE1-dependent metabolism of neuregulin 1: Bridging the gap in explaining the occurrence of schizophrenia-like symptoms in Alzheimer's disease with psychosis? Ageing Res Rev 2023; 89:101988. [PMID: 37331479 DOI: 10.1016/j.arr.2023.101988] [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: 03/08/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
Alzheimer's disease is a neurodegenerative disease mainly characterized by cortico-neuronal atrophy, impaired memory and other cognitive declines. On the other hand, schizophrenia is a neuro-developmental disorder with an overtly active central nervous system pruning system resulting into abrupt connections with common symptoms including disorganised thoughts, hallucination and delusion. Nevertheless, the fronto-temporal anomaly presents itself as a common denominator for the two pathologies. There is even a strong presumption of increased risk of developing co-morbid dementia for schizophrenic individuals and psychosis for Alzheimer's disease patients, overall leading to a further deteriorated quality of life. However, convincing proofs of how these two disorders, although very distant from each other when considering their aetiology, develop coexisting symptoms is yet to be resolved. At the molecular level, the two primarily neuronal proteins β-amyloid precursor protein and neuregulin 1 have been considered in this relevant context, although the conclusions are for the moment only hypotheses. In order to propose a model for explaining the psychotic schizophrenia-like symptoms that sometimes accompany AD-associated dementia, this review projects out on the similar sensitivity shared by these two proteins regarding their metabolism by the β-site APP cleaving enzyme 1.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560 Valbonne, France.
| | - Subhamita Maitra
- Department of Molecular Biology, Umeå University, Umeå 90736, Sweden
| |
Collapse
|
4
|
Nowrangi MA, Outen JD, Kim J, Avramopoulos D, Lyketsos CG, Rosenberg PB. Neuropsychiatric Symptoms of Alzheimer's Disease: An Anatomic-Genetic Framework for Treatment Development. J Alzheimers Dis 2023; 95:53-68. [PMID: 37522204 DOI: 10.3233/jad-221247] [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] [Indexed: 08/01/2023]
Abstract
BACKGROUND Despite the burden on patients and caregivers, there are no approved therapies for the neuropsychiatric symptoms of Alzheimer's disease (NPS-AD). This is likely due to an incomplete understanding of the underlying mechanisms. OBJECTIVE To review the neurobiological mechanisms of NPS-AD, including depression, psychosis, and agitation. METHODS Understanding that genetic encoding gives rise to the function of neural circuits specific to behavior, we review the genetics and neuroimaging literature to better understand the biological underpinnings of depression, psychosis, and agitation. RESULTS We found that mechanisms involving monoaminergic biosynthesis and function are likely key elements of NPS-AD and while current treatment approaches are in line with this, the lack of effectiveness may be due to contributions from additional mechanisms including neurodegenerative, vascular, inflammatory, and immunologic pathways. CONCLUSION Within an anatomic-genetic framework, development of novel effective biological targets may engage targets within these pathways but will require a better understanding of the heterogeneity in NPS-AD.
Collapse
Affiliation(s)
- Milap A Nowrangi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - John D Outen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios Avramopoulos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Constantine G Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| |
Collapse
|
5
|
Cardona K, Medina J, Orrego-Cardozo M, Restrepo de Mejía F, Elcoroaristizabal X, Naranjo Galvis CA. Inflammatory gene expression profiling in peripheral blood from patients with Alzheimer's disease reveals key pathways and hub genes with potential diagnostic utility: a preliminary study. PeerJ 2021; 9:e12016. [PMID: 34484988 PMCID: PMC8381883 DOI: 10.7717/peerj.12016] [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: 03/15/2021] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related neurodegenerative disease caused by central nervous system disorders. Late-onset Alzheimer disease (LOAD) is the most common neurodegenerative disorder worldwide. Differences at the expression level of certain genes, resulting from either genetic variations or environmental interactions, might be one of the mechanisms underlying differential risks for developing AD. Peripheral blood genome transcriptional profiling may provide a powerful and minimally invasive tool for the identification of novel targets beyond Aβ and tau for AD research. METHODS This preliminary study explores molecular pathogenesis of LOAD-related inflammation through next generation sequencing, to assess RNA expression profiles in peripheral blood from five patients with LOAD and 10 healthy controls. RESULTS The analysis of RNA expression profiles revealed 94 genes up-regulated and 147 down-regulated. Gene function analysis, including Gene Ontology (GO) and KOBAS-Kyoto Encyclopedia of DEGs and Genomes (KEGG) pathways indicated upregulation of interferon family (INF) signaling, while the down-regulated genes were mainly associated with the cell cycle process. KEGG metabolic pathways mapping showed gene expression alterations in the signaling pathways of JAK/STAT, chemokines, MAP kinases and Alzheimer disease. The results of this preliminary study provided not only a comprehensive picture of gene expression, but also the key processes associated with pathology for the regulation of neuroinflammation, to improve the current mechanisms to treat LOAD.
Collapse
Affiliation(s)
- Kelly Cardona
- Facultad de Salud, Universidad Autónoma de Manizales, Manizales, Caldas, Colombia
| | - Javier Medina
- Facultad de Salud, Universidad Autónoma de Manizales, Manizales, Caldas, Colombia
| | - Mary Orrego-Cardozo
- Facultad de Salud, Universidad Autónoma de Manizales, Manizales, Caldas, Colombia
| | | | | | | |
Collapse
|
6
|
Shahsavani N, Alizadeh A, Kataria H, Karimi-Abdolrezaee S. Availability of neuregulin-1beta1 protects neurons in spinal cord injury and against glutamate toxicity through caspase dependent and independent mechanisms. Exp Neurol 2021; 345:113817. [PMID: 34314724 DOI: 10.1016/j.expneurol.2021.113817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/06/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) causes sensorimotor and autonomic impairment that partly reflects extensive, permanent loss of neurons at the epicenter and penumbra of the injury. Strategies aimed at enhancing neuronal protection are critical to attenuate neurodegeneration and improve neurological recovery after SCI. In rat SCI, we previously uncovered that the tissue levels of neuregulin-1beta 1 (Nrg-1β1) are acutely and persistently downregulated in the injured spinal cord. Nrg-1β1 is well-known for its critical roles in the development, maintenance and physiology of neurons and glia in the developing and adult spinal cord. However, despite this pivotal role, Nrg-1β1 specific effects and mechanisms of action on neuronal injury remain largely unknown in SCI. In the present study, using a clinically-relevant model of compressive/contusive SCI in rats and an in vitro model of glutamate toxicity in primary neurons, we demonstrate Nrg-1β1 provides early neuroprotection through attenuation of reactive oxygen species, lipid peroxidation, necrosis and apoptosis in acute and subacute stages of SCI. Mechanistically, availability of Nrg-1β1 following glutamate challenge protects neurons from caspase-dependent and independent cell death that is mediated by modulation of mitochondria associated apoptotic cascades and MAP kinase and AKT signaling pathways. Altogether, our work provides novel insights into the role and mechanisms of Nrg-1β1 in neuronal injury after SCI and introduces its potential as a new neuroprotective target for this debilitating neurological condition.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
7
|
Artificial intelligence and leukocyte epigenomics: Evaluation and prediction of late-onset Alzheimer's disease. PLoS One 2021; 16:e0248375. [PMID: 33788842 PMCID: PMC8011726 DOI: 10.1371/journal.pone.0248375] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
We evaluated the utility of leucocyte epigenomic-biomarkers for Alzheimer’s Disease (AD) detection and elucidates its molecular pathogeneses. Genome-wide DNA methylation analysis was performed using the Infinium MethylationEPIC BeadChip array in 24 late-onset AD (LOAD) and 24 cognitively healthy subjects. Data were analyzed using six Artificial Intelligence (AI) methodologies including Deep Learning (DL) followed by Ingenuity Pathway Analysis (IPA) was used for AD prediction. We identified 152 significantly (FDR p<0.05) differentially methylated intragenic CpGs in 171 distinct genes in AD patients compared to controls. All AI platforms accurately predicted AD with AUCs ≥0.93 using 283,143 intragenic and 244,246 intergenic/extragenic CpGs. DL had an AUC = 0.99 using intragenic CpGs, with both sensitivity and specificity being 97%. High AD prediction was also achieved using intergenic/extragenic CpG sites (DL significance value being AUC = 0.99 with 97% sensitivity and specificity). Epigenetically altered genes included CR1L & CTSV (abnormal morphology of cerebral cortex), S1PR1 (CNS inflammation), and LTB4R (inflammatory response). These genes have been previously linked with AD and dementia. The differentially methylated genes CTSV & PRMT5 (ventricular hypertrophy and dilation) are linked to cardiovascular disease and of interest given the known association between impaired cerebral blood flow, cardiovascular disease, and AD. We report a novel, minimally invasive approach using peripheral blood leucocyte epigenomics, and AI analysis to detect AD and elucidate its pathogenesis.
Collapse
|
8
|
Klein PC, Ettinger U, Schirner M, Ritter P, Rujescu D, Falkai P, Koutsouleris N, Kambeitz-Ilankovic L, Kambeitz J. Brain Network Simulations Indicate Effects of Neuregulin-1 Genotype on Excitation-Inhibition Balance in Cortical Dynamics. Cereb Cortex 2021; 31:2013-2025. [PMID: 33279967 DOI: 10.1093/cercor/bhaa339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/01/2020] [Accepted: 10/11/2020] [Indexed: 11/14/2022] Open
Abstract
Neuregulin-1 (NRG1) represents an important factor for multiple processes including neurodevelopment, brain functioning or cognitive functions. Evidence from animal research suggests an effect of NRG1 on the excitation-inhibition (E/I) balance in cortical circuits. However, direct evidence for the importance of NRG1 in E/I balance in humans is still lacking. In this work, we demonstrate the application of computational, biophysical network models to advance our understanding of the interaction between cortical activity observed in neuroimaging and the underlying neurobiology. We employed a biophysical neuronal model to simulate large-scale brain dynamics and to investigate the role of polymorphisms in the NRG1 gene (rs35753505, rs3924999) in n = 96 healthy adults. Our results show that G/G-carriers (rs3924999) exhibit a significant difference in global coupling (P = 0.048) and multiple parameters determining E/I-balance such as excitatory synaptic coupling (P = 0.047), local excitatory recurrence (P = 0.032) and inhibitory synaptic coupling (P = 0.028). This indicates that NRG1 may be related to excitatory recurrence or excitatory synaptic coupling potentially resulting in altered E/I-balance. Moreover, we suggest that computational modeling is a suitable tool to investigate specific biological mechanisms in health and disease.
Collapse
Affiliation(s)
- Pedro Costa Klein
- Department of Psychiatry, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Germany
| | - Ulrich Ettinger
- Department of Psychology, University of Bonn, Bonn, 53111, Germany
| | - Michael Schirner
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Dept. of Neurology, 10117, Germany.,Bernstein Focus State Dependencies of Learning & Bernstein Center for Computational Neuroscience, Berlin 10115, Germany
| | - Petra Ritter
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Dept. of Neurology, 10117, Germany.,Bernstein Focus State Dependencies of Learning & Bernstein Center for Computational Neuroscience, Berlin 10115, Germany
| | - Dan Rujescu
- University Clinic for Psychiatry, Psychotherapy and Psychosomatic, Martin-Luther-University, Halle-Wittenberg, 06112, Germany
| | - Peter Falkai
- Department of Psychiatry, Ludwig Maximilians Universität München, 80336, Germany
| | | | - Lana Kambeitz-Ilankovic
- Department of Psychiatry, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Germany.,Department of Psychiatry, Ludwig Maximilians Universität München, 80336, Germany
| | - Joseph Kambeitz
- Department of Psychiatry, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Germany
| |
Collapse
|
9
|
Ledonne A, Mercuri NB. Insights on the Functional Interaction between Group 1 Metabotropic Glutamate Receptors (mGluRI) and ErbB Receptors. Int J Mol Sci 2020; 21:ijms21217913. [PMID: 33114459 PMCID: PMC7662933 DOI: 10.3390/ijms21217913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 11/16/2022] Open
Abstract
It is well-appreciated that phosphorylation is an essential post-translational mechanism of regulation for several proteins, including group 1 metabotropic glutamate receptors (mGluRI), mGluR1, and mGluR5 subtypes. While contributions of various serine/threonine protein kinases on mGluRI modulation have been recognized, the functional role of tyrosine kinases (TKs) is less acknowledged. Here, while describing current evidence supporting that mGluRI are targets of TKs, we mainly focus on the modulatory roles of the ErbB tyrosine kinases receptors—activated by the neurotrophic factors neuregulins (NRGs)—on mGluRI function. Available evidence suggests that mGluRI activity is tightly dependent on ErbB signaling, and that ErbB’s modulation profoundly influences mGluRI-dependent effects on neurotransmission, neuronal excitability, synaptic plasticity, and learning and memory processes.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Correspondence: ; Tel.: +39-06-50170-3160
| | - Nicola B. Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, Università di Roma “Tor Vergata”, 00133 Rome, Italy;
| |
Collapse
|
10
|
Clinical Aspects of Neurobehavioral Symptoms of Dementia. Dement Neurocogn Disord 2020; 19:54-64. [PMID: 32602280 PMCID: PMC7326614 DOI: 10.12779/dnd.2020.19.2.54] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/11/2020] [Accepted: 05/17/2020] [Indexed: 12/03/2022] Open
Abstract
Neurobehavioral symptoms of dementia (NBSD) are very common and are significant symptoms of the illness, contributing most to caregiver burdens and often resulting in premature institutionalization of the person with dementia. The main symptoms of NBSD are anxiety, depression, delusions, and hallucinations. NBSD produce significant problems for both patients and caregivers. The pathophysiology of NBSD is determined by genetic, structural, or environmental factors. Therefore, treatment of NBSD requires continuous and organic cooperation between patients, caregivers, social environments, and doctors. Therefore, it is important for neurologists, who mainly view NBSD for dementia patients, to increase their understanding of these more comprehensive areas as well as the latest insights and treatments to help patients and caregivers.
Collapse
|
11
|
Ledonne A, Mercuri NB. On the Modulatory Roles of Neuregulins/ErbB Signaling on Synaptic Plasticity. Int J Mol Sci 2019; 21:ijms21010275. [PMID: 31906113 PMCID: PMC6981567 DOI: 10.3390/ijms21010275] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Neuregulins (NRGs) are a family of epidermal growth factor-related proteins, acting on tyrosine kinase receptors of the ErbB family. NRGs play an essential role in the development of the nervous system, since they orchestrate vital functions such as cell differentiation, axonal growth, myelination, and synapse formation. They are also crucially involved in the functioning of adult brain, by directly modulating neuronal excitability, neurotransmission, and synaptic plasticity. Here, we provide a review of the literature documenting the roles of NRGs/ErbB signaling in the modulation of synaptic plasticity, focusing on evidence reported in the hippocampus and midbrain dopamine (DA) nuclei. The emerging picture shows multifaceted roles of NRGs/ErbB receptors, which critically modulate different forms of synaptic plasticity (LTP, LTD, and depotentiation) affecting glutamatergic, GABAergic, and DAergic synapses, by various mechanisms. Further, we discuss the relevance of NRGs/ErbB-dependent synaptic plasticity in the control of brain processes, like learning and memory and the known involvement of NRGs/ErbB signaling in the modulation of synaptic plasticity in brain’s pathological conditions. Current evidence points to a central role of NRGs/ErbB receptors in controlling glutamatergic LTP/LTD and GABAergic LTD at hippocampal CA3–CA1 synapses, as well as glutamatergic LTD in midbrain DA neurons, thus supporting that NRGs/ErbB signaling is essential for proper brain functions, cognitive processes, and complex behaviors. This suggests that dysregulated NRGs/ErbB-dependent synaptic plasticity might contribute to mechanisms underlying different neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Correspondence: ; Tel.: +3906-501703160; Fax: +3906-501703307
| | - Nicola B. Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier no 1, 00133 Rome, Italy
| |
Collapse
|
12
|
Tsuang DW, Greenwood TA, Jayadev S, Davis M, Shutes-David A, Bird TD. A Genetic Study of Psychosis in Huntington's Disease: Evidence for the Involvement of Glutamate Signaling Pathways. J Huntingtons Dis 2019; 7:51-59. [PMID: 29480208 DOI: 10.3233/jhd-170277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Psychotic symptoms of delusions and hallucinations occur in about 5% of persons with Huntington's disease (HD). The mechanisms underlying these occurrences are unknown, but the same symptoms also occur in schizophrenia, and thus genetic risk factors for schizophrenia may be relevant to the development of psychosis in HD. OBJECTIVE To investigate the possible role of genes associated with schizophrenia in the occurrence of psychotic symptoms in HD. METHODS DNA from subjects with HD and psychosis (HD+P; n = 47), subjects with HD and no psychosis (HD-P; n = 126), and controls (CTLs; n = 207) was genotyped using the Infinium PsychArray-24 v1.1 BeadChip. The allele frequencies of single-nucleotide polymorphisms (SNPs) that were previously associated with schizophrenia and related psychiatric disorders were compared between these groups. RESULTS Of the 30 candidate genes tested, 10 showed an association with psychosis in HD. The majority of these genes, including CTNNA2, DRD2, ERBB4, GRID2, GRIK4, GRM1, NRG1, PCNT, RELN, and SLC1A2, demonstrate network interactions related to glutamate signaling. CONCLUSIONS This study suggests genetic associations between several previously identified candidate genes for schizophrenia and the occurrence of psychotic symptoms in HD. These data support the potential role of genes related to glutamate signaling in HD psychosis.
Collapse
Affiliation(s)
- Debby W Tsuang
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA.,Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Marie Davis
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Neurology, University of Washington, Seattle, WA, USA
| | - Andrew Shutes-David
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.,Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Thomas D Bird
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA.,Department of Neurology, University of Washington, Seattle, WA, USA
| |
Collapse
|
13
|
Cespedes JC, Liu M, Harbuzariu A, Nti A, Onyekaba J, Cespedes HW, Bharti PK, Solomon W, Anyaoha P, Krishna S, Adjei A, Botchway F, Ford B, Stiles JK. Neuregulin in Health and Disease. INTERNATIONAL JOURNAL OF BRAIN DISORDERS AND TREATMENT 2018; 4:024. [PMID: 31032468 PMCID: PMC6483402 DOI: 10.23937/2469-5866/1410024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Juan Carlos Cespedes
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Annette Nti
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - John Onyekaba
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Hanna Watson Cespedes
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | | | - Wesley Solomon
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Precious Anyaoha
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Sri Krishna
- ICMR-National Institute for Research in Tribal Health, India
| | - Andrew Adjei
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Ghana
| | - Felix Botchway
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Ghana
| | - Byron Ford
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, USA
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| |
Collapse
|
14
|
Ledonne A, Mango D, Latagliata EC, Chiacchierini G, Nobili A, Nisticò R, D'Amelio M, Puglisi-Allegra S, Mercuri NB. Neuregulin 1/ErbB signalling modulates hippocampal mGluRI-dependent LTD and object recognition memory. Pharmacol Res 2018; 130:12-24. [PMID: 29427771 DOI: 10.1016/j.phrs.2018.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/19/2018] [Accepted: 02/02/2018] [Indexed: 01/05/2023]
Abstract
The neurotrophic factors neuregulins (NRGs) and their receptors, ErbB tyrosine kinases, regulate neurotransmission, synaptic plasticity and cognitive functions and their alterations have been associated to different neuropsychiatric disorders. Group 1 metabotropic glutamate receptors (mGluRI)-dependent mechanisms are also altered in animal models of neuropsychiatric diseases, especially mGluRI-induced glutamatergic long-term depression (mGluRI-LTD), a form of synaptic plasticity critically involved in learning and memory. Despite this evidence, a potential link between NRGs/ErbB signalling and mGluRI-LTD has never been considered. Here, we aimed to test the hypothesis that NRGs/ErbB signalling regulates mGluRI functions in the hippocampus, thus controlling CA1 pyramidal neurons excitability and synaptic plasticity as well as mGluRI-dependent behaviors. We investigated the functional interaction between NRG1/ErbB signalling and mGluRI in hippocampal CA1 pyramidal neurons, by analyzing the effect of a pharmacological modulation of NRG1/ErbB signalling on the excitation of pyramidal neurons and on the LTD at CA3-CA1 synapses induced by an mGluRI agonist. Furthermore, we verified the involvement of ErbB signalling in mGluRI-dependent learning processes, by evaluating the consequence of an intrahippocampal in vivo injection of a pan-ErbB inhibitor in the object recognition test in mice, a learning task dependent on hippocampal mGluRI. We found that NRG1 potentiates mGluRI-dependent functions on pyramidal neurons excitability and synaptic plasticity at CA3-CA1 synapses. Further, endogenous ErbB signalling per se regulates, through mGluRI, neuronal excitability and LTD in CA1 pyramidal neurons, since ErbB inhibition reduces mGluRI-induced neuronal excitation and mGluRI-LTD. In vivo intrahippocampal injection of the ErbB inhibitor, PD158780, impairs mGluRI-LTD at CA3-CA1 synapses and affects the exploratory behavior in the object recognition test. Thus, our results identify a key role for NRG1/ErbB signalling in the regulation of hippocampal mGluRI-dependent synaptic and cognitive functions, whose alteration might contribute to the pathogenesis of different brain diseases.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation, Rome, Italy.
| | - Dalila Mango
- Pharmacology of Synaptic Disease Lab, European Brain Research Institute, Rome, Italy
| | | | - Giulia Chiacchierini
- Department of Psychology and "Daniel Bovet" Center, University "La Sapienza", Rome, Italy
| | - Annalisa Nobili
- Department of Experimental Neuroscience, Santa Lucia Foundation, Rome, Italy; Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Robert Nisticò
- Pharmacology of Synaptic Disease Lab, European Brain Research Institute, Rome, Italy; Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neuroscience, Santa Lucia Foundation, Rome, Italy; Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Stefano Puglisi-Allegra
- Department of Experimental Neuroscience, Santa Lucia Foundation, Rome, Italy; Department of Psychology and "Daniel Bovet" Center, University "La Sapienza", Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
15
|
Shah C, DeMichele-Sweet MAA, Sweet RA. Genetics of psychosis of Alzheimer disease. Am J Med Genet B Neuropsychiatr Genet 2017; 174:27-35. [PMID: 26756273 PMCID: PMC5154859 DOI: 10.1002/ajmg.b.32413] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023]
Abstract
Psychotic symptoms, comprised of delusions and hallucinations, occur in about half of individuals with Alzheimer disease (AD with psychosis, AD+P). These individuals have greater agitation, aggression, depression, functional impairment, and mortality than individuals without psychosis (AD-P). Although the exact etiopathogenesis of AD+P is unclear, the rapidly developing field of genomics continues to expand our understanding of this disease. Several independent studies have demonstrated familial aggregation and heritability of AD+P. Linkage studies have been suggestive of loci on several chromosomes associated with AD+P. Association studies examining apolipoprotein E gene, the best established genetic risk factor for late-onset AD, did not find any significant association of this gene with AD+P. Other candidate gene studies focusing on monoamine neurotransmitter systems have yielded equivocal results. A genome-wide association study and studies examining copy number variations recently have detected suggestive associations, but have been underpowered. Approaches to increase sizes of AD+P samples for genome wide association studies are discussed. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chintan Shah
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Robert A. Sweet
- Department of Psychiatry and Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
- VISN 4 Mental Health Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Identification of candidate genes for congenital heart defects on proximal chromosome 8p. Sci Rep 2016; 6:36133. [PMID: 27808268 PMCID: PMC5093561 DOI: 10.1038/srep36133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/10/2016] [Indexed: 12/21/2022] Open
Abstract
With the application of advanced molecular cytogenetic techniques, the number of patients identified as having abnormal chromosome 8p has increased progressively. Individuals with terminal 8p deletion have been extensively described in previous studies. The manifestations usually include cardiac anomalies, developmental delay/mental retardation, craniofacial abnormalities, and multiple other minor anomalies. However, some patients with proximal deletion also presented with similar phenotypic features. Here we describe a female child with an 18.5-Mb deletion at 8p11.23–p22 that include the cardiac-associated loci NKX2-6 and NRG1. Further mutation screening of these two candidate genes in 143 atrial septal defect patients, two heterozygous mutations NKX2-6 (c.1A > T) and NRG1 (c.1652G > A) were identified. The mutations were described for the first time in patients with congenital heart disease (CHD). The c.1A > T NKX2-6 generated a protein truncated by 45 amino acids with a decreased level of mRNA expression, whereas the NRG1 mutation had no significant effect on protein functions. Our findings suggest that 8p21-8p12 may be another critical region for 8p-associated CHD, and some cardiac malformations might be due to NKX2-6 haploinsufficiency. This study also links the NKX2-6 mutation to ASD for the first time, providing novel insight into the molecular underpinning of this common form of CHD.
Collapse
|
17
|
|
18
|
Xu J, de Winter F, Farrokhi C, Rockenstein E, Mante M, Adame A, Cook J, Jin X, Masliah E, Lee KF. Neuregulin 1 improves cognitive deficits and neuropathology in an Alzheimer's disease model. Sci Rep 2016; 6:31692. [PMID: 27558862 PMCID: PMC4997345 DOI: 10.1038/srep31692] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/25/2016] [Indexed: 01/06/2023] Open
Abstract
Several lines of evidence suggest that neuregulin 1 (NRG1) signaling may influence cognitive function and neuropathology in Alzheimer’s disease (AD). To test this possibility, full-length type I or type III NRG1 was overexpressed via lentiviral vectors in the hippocampus of line 41 AD mouse. Both type I and type III NRG1 improves deficits in the Morris water-maze behavioral task. Neuropathology was also significantly ameliorated. Decreased expression of the neuronal marker MAP2 and synaptic markers PSD95 and synaptophysin in AD mice was significantly reversed. Levels of Aβ peptides and plaques were markedly reduced. Furthermore, we showed that soluble ectodomains of both type I and type III NRG1 significantly increased expression of Aβ-degrading enzyme neprilysin (NEP) in primary neuronal cultures. Consistent with this finding, immunoreactivity of NEP was increased in the hippocampus of AD mice. These results suggest that NRG1 provides beneficial effects in candidate neuropathologic substrates of AD and, therefore, is a potential target for the treatment of AD.
Collapse
Affiliation(s)
- Jiqing Xu
- Clayton Foundation for Peptide Biology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| | - Fred de Winter
- Clayton Foundation for Peptide Biology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| | - Catherine Farrokhi
- Clayton Foundation for Peptide Biology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Michael Mante
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Anthony Adame
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jonathan Cook
- Molecular Neurobiology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| | - Xin Jin
- Molecular Neurobiology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Kuo-Fen Lee
- Clayton Foundation for Peptide Biology Laboratories, The Salk Institute, La Jolla, CA 92037, USA
| |
Collapse
|
19
|
Huh S, Baek SJ, Lee KH, Whitcomb DJ, Jo J, Choi SM, Kim DH, Park MS, Lee KH, Kim BC. The reemergence of long-term potentiation in aged Alzheimer's disease mouse model. Sci Rep 2016; 6:29152. [PMID: 27377368 PMCID: PMC4932605 DOI: 10.1038/srep29152] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/15/2016] [Indexed: 12/20/2022] Open
Abstract
Mouse models of Alzheimer’s disease (AD) have been developed to study the pathophysiology of amyloid β protein (Aβ) toxicity, which is thought to cause severe clinical symptoms such as memory impairment in AD patients. However, inconsistencies exist between studies using these animal models, specifically in terms of the effects on synaptic plasticity, a major cellular model of learning and memory. Whereas some studies find impairments in plasticity in these models, others do not. We show that long-term potentiation (LTP), in the CA1 region of hippocampal slices from this mouse, is impared at Tg2576 adult 6–7 months old. However, LTP is inducible again in slices taken from Tg2576 aged 14–19 months old. In the aged Tg2576, we found that the percentage of parvalbumin (PV)-expressing interneurons in hippocampal CA1-3 region is significantly decreased, and LTP inhibition or reversal mediated by NRG1/ErbB signaling, which requires ErbB4 receptors in PV interneurons, is impaired. Inhibition of ErbB receptor kinase in adult Tg2576 restores LTP but impairs depotentiation as shown in aged Tg2576. Our study suggests that hippocampal LTP reemerges in aged Tg2576. However, this reemerged LTP is an insuppressible form due to impaired NRG1/ErbB signaling, possibly through the loss of PV interneurons.
Collapse
Affiliation(s)
- Seonghoo Huh
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Soo-Ji Baek
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Daniel J Whitcomb
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Health Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Jihoon Jo
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.,Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Seong-Min Choi
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences and Dong-A Anti-aging Research Center, Dong-A University, Busan 49315, Republic of Korea
| | - Man-Seok Park
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Kun Ho Lee
- National Research Center for Dementia, Gwangju 61452, Republic of Korea
| | - Byeong C Kim
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.,Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.,Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.,National Research Center for Dementia, Gwangju 61452, Republic of Korea
| |
Collapse
|
20
|
Baik TK, Kim YJ, Kang SM, Song DY, Min SS, Woo RS. Blocking the phosphatidylinositol 3-kinase pathway inhibits neuregulin-1-mediated rescue of neurotoxicity induced by Aβ1-42. ACTA ACUST UNITED AC 2016; 68:1021-9. [PMID: 27230708 DOI: 10.1111/jphp.12563] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/29/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Neuregulin-1 (NRG1) has an important role in both the development and the plasticity of the brain as well as neuroprotective properties. In this study, we investigated the downstream pathways of NRG1 signalling and their role in the prevention of Aβ1-42 -induced neurotoxicity. METHODS Lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) generation, superoxide dismutase (SOD) activity and TUNEL staining were assayed to examine the neuroprotective properties in primary rat cortical neurons. KEY FINDINGS The inhibition of PI3K/Akt activation abolished the ability of NRG1 to prevent Aβ1-42 -induced LDH release and increased TUNEL-positive cell count and reactive oxygen species accumulation in primary cortical neurons. CONCLUSIONS Our results demonstrate that NRG1 signalling exerts a neuroprotective effect against Aβ1-42 -induced neurotoxicity via activation of the PI3K/Akt pathway. Furthermore, this suggests that NRG1 has neuroprotective potential for the treatment of AD.
Collapse
Affiliation(s)
- Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| | - Young-Jung Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| | - Se-Mi Kang
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| | - Dae-Yong Song
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, College of Medicine, Eulji University, Daejeon, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| |
Collapse
|
21
|
Ryu J, Hong BH, Kim YJ, Yang EJ, Choi M, Kim H, Ahn S, Baik TK, Woo RS, Kim HS. Neuregulin-1 attenuates cognitive function impairments in a transgenic mouse model of Alzheimer's disease. Cell Death Dis 2016; 7:e2117. [PMID: 26913607 PMCID: PMC4849157 DOI: 10.1038/cddis.2016.30] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/20/2022]
Abstract
The neuregulin (NRG) family of epidermal growth factor-related proteins is composed of a wide variety of soluble and membrane-bound proteins that exert their effects via the tyrosine kinase receptors ErbB2-ErbB4. In the nervous system, the functions of NRG1 are essential for peripheral myelination, the establishment and maintenance of neuromuscular and sensorimotor systems and the plasticity of cortical neuronal circuits. In the present study, we report that an intracerebroventricular infusion of NRG1 attenuated cognitive impairments in 13-month-old Tg2576 mice, an animal model of Alzheimer's disease (AD). In addition, according to Golgi-Cox staining, NRG1 rescued the reduction in the number of dendritic spines detected in the brains of Tg2576 mice compared with vehicle (PBS)-infused mice. This result was also corroborated in vitro as NRG1 attenuated the oligomeric amyloid beta peptide1-42 (Aβ1-42)-induced decrease in dendritic spine density in rat primary hippocampal neuron cultures. NRG1 also alleviated the decrease in neural differentiation induced by oligomeric Aβ1-42 in mouse fetal neural stem cells. Collectively, these results suggest that NRG1 has a therapeutic potential for AD by alleviating the reductions in dendritic spine density and neurogenesis found in AD brains.
Collapse
Affiliation(s)
- J Ryu
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - B-H Hong
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - Y-J Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Republic of Korea
| | - E-J Yang
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - M Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - H Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - S Ahn
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| | - T-K Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Republic of Korea
| | - R-S Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Republic of Korea
| | - H-S Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea.,Seoul National University College of Medicine, Bundang Hospital, Bundang-Gu, Sungnam, Republic of Korea.,Neuroscience Research Institute, College of Medicine, Seoul National University, 103 Daehakro, Jongno-gu, Seoul, Republic of Korea
| |
Collapse
|
22
|
Barrenschee M, Lange C, Cossais F, Egberts JH, Becker T, Wedel T, Böttner M. Expression and function of Neuregulin 1 and its signaling system ERBB2/3 in the enteric nervous system. Front Cell Neurosci 2015; 9:360. [PMID: 26441531 PMCID: PMC4585281 DOI: 10.3389/fncel.2015.00360] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/28/2015] [Indexed: 12/12/2022] Open
Abstract
Neuregulin 1 (NRG1) is suggested to promote the survival and maintenance of the enteric nervous system (ENS). As deficiency in its corresponding receptor signaling complex ERBB2/ERBB3 leads to postnatal colonic hypo/aganglionosis we assessed the distributional and expressional pattern of the NRG1-ERBB2/ERBB3 system in the human colon and explored the neurotrophic capacity of NRG1 on cultured enteric neurons. Site-specific mRNA expression of the NRG1-ERBB2/3 system was determined in microdissected samples harvested from enteric musculature and ganglia. Localization of NRG1, ERBB2 and ERBB3 was determined by dual-label-immunohistochemistry using pan-neuronal and pan-glial markers. Morphometric analysis was performed on NRG1-stimulated rat enteric nerve cultures to evaluate neurotrophic effects. mRNA expression of the NRG1-ERBB2/3 system was determined by qPCR. Co-localization of NRG1 with neuronal or synaptic markers was analyzed in enteric nerve cultures stimulated with glial cell line-derived neurotrophic factor (GDNF). The NRG1 system was expressed in both neurons and glial cells of enteric ganglia and in nerve fibers. NRG1 significantly enhanced growth parameters in enteric nerve cell cultures and ErB3 mRNA expression was down-regulated upon NRG1 stimulation. GDNF negatively regulates ErbB2 and ErbB3 mRNA expression. The NRG1-ERBB2/3 system is physiologically present in the human ENS and NRG1 acts as a neurotrophic factor for the ENS. The down-regulation of ErbB3/ErbB2 in GDNF stimulated nerve cell cultures points to an interaction of both neurotrophic factors. Thus, the data may provide a basis to assess disturbed signaling components of the NRG1 system in enteric neuropathies.
Collapse
Affiliation(s)
- Martina Barrenschee
- Neurogastroenterology, Institute of Anatomy, Christian-Albrechts-University of Kiel Kiel, Germany
| | - Christina Lange
- Neurogastroenterology, Institute of Anatomy, Christian-Albrechts-University of Kiel Kiel, Germany
| | - François Cossais
- Neurogastroenterology, Institute of Anatomy, Christian-Albrechts-University of Kiel Kiel, Germany
| | - Jan-Hendrik Egberts
- Department of General, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel Kiel, Germany
| | - Thomas Becker
- Department of General, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel Kiel, Germany
| | - Thilo Wedel
- Neurogastroenterology, Institute of Anatomy, Christian-Albrechts-University of Kiel Kiel, Germany
| | - Martina Böttner
- Neurogastroenterology, Institute of Anatomy, Christian-Albrechts-University of Kiel Kiel, Germany
| |
Collapse
|
23
|
Barral S, Vardarajan BN, Reyes-Dumeyer D, Faber KM, Bird TD, Tsuang D, Bennett DA, Rosenberg R, Boeve BF, Graff-Radford NR, Goate AM, Farlow M, Lantigua R, Medrano MZ, Wang X, Kamboh MI, Barmada MM, Schaid DJ, Foroud TM, Weamer EA, Ottman R, Sweet RA, Mayeux R. Genetic variants associated with susceptibility to psychosis in late-onset Alzheimer's disease families. Neurobiol Aging 2015; 36:3116.e9-3116.e16. [PMID: 26359528 DOI: 10.1016/j.neurobiolaging.2015.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/01/2015] [Accepted: 08/08/2015] [Indexed: 11/16/2022]
Abstract
Psychotic symptoms are frequent in late-onset Alzheimer's disease (LOAD) patients. Although the risk for psychosis in LOAD is genetically mediated, no genes have been identified. To identify loci potentially containing genetic variants associated with risk of psychosis in LOAD, a total of 263 families from the National Institute of Aging-LOAD cohort were classified into psychotic (LOAD+P, n = 215) and nonpsychotic (LOAD-P, n = 48) families based on the presence/absence of psychosis during the course of LOAD. The LOAD+P families yielded strong evidence of linkage on chromosome 19q13 (two-point [2-pt] logarithm of odds [LOD] = 3.8, rs2285513 and multipoint LOD = 2.7, rs541169). Joint linkage and association in 19q13 region detected strong association with rs2945988 (p = 8.7 × 10(-7)). Linkage results for the LOAD-P families yielded nonsignificant 19q13 LOD scores. Several 19q13 single-nucleotide polymorphisms generalized the association of LOAD+P in a Caribbean Hispanic (CH) cohort, and the strongest signal was rs10410711 (pmeta = 5.1 × 10(-5)). A variant located 24 kb upstream of rs10410711 and rs10421862 was strongly associated with LOAD+P (pmeta = 1.0 × 10(-5)) in a meta-analysis of the CH cohort and an additional non-Hispanic Caucasian dataset. Identified variants rs2945988 and rs10421862 affect brain gene expression levels. Our results suggest that genetic variants in genes on 19q13, some of which are involved in brain development and neurodegeneration, may influence the susceptibility to psychosis in LOAD patients.
Collapse
Affiliation(s)
- Sandra Barral
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Badri N Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Kelley M Faber
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Debby Tsuang
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Roger Rosenberg
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Alison M Goate
- Icanhn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Farlow
- Department of Neurology, Indiana University Center for Alzheimer's Disease and Related Disorders, Indianapolis, IN, USA
| | - Rafael Lantigua
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Medicine, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Martin Z Medrano
- Department of Geriatrics, Pontificia Universidad Católica Madre y Maestra, Santiago, Dominican Republic
| | - Xinbing Wang
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Tatiana M Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Elise A Weamer
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruth Ottman
- Gertrude H. Sergievsky Center, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Epidemiology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA.
| | | |
Collapse
|
24
|
Zahodne LB, Ornstein K, Cosentino S, Devanand DP, Stern Y. Longitudinal relationships between Alzheimer disease progression and psychosis, depressed mood, and agitation/aggression. Am J Geriatr Psychiatry 2015; 23:130-40. [PMID: 23871118 PMCID: PMC3858495 DOI: 10.1016/j.jagp.2013.03.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/19/2013] [Accepted: 03/27/2013] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Behavioral and psychological symptoms of dementia (BPSD) are prevalent in Alzheimer disease (AD) and are related to poor outcomes such as nursing home placement. No study has examined the impact of individual BPSD on dependence, a clinically important feature that reflects changing patient needs and their effect on caregivers. The current study characterized independent cross-sectional and longitudinal relationships between three BPSD (psychosis, depressed mood, and agitation/aggression), cognition, and dependence to better understand the interplay between these symptoms over time. DESIGN The Predictors Study measured changes in BPSD, cognition, and dependence every 6 months in patients with AD. Cross-sectional and longitudinal relationships between individual BPSD, cognition, and dependence over 6 years were characterized by using multivariate latent growth curve modeling. This approach characterizes independent changes in multiple outcome measures over time. SETTING Four memory clinics in the United States and Europe. PARTICIPANTS A total of 517 patients with probable AD. MEASUREMENTS Columbia University Scale for Psychopathology, modified Mini-Mental State Examination, and Dependence Scale. RESULTS Both psychosis and depressed mood at study entry were associated with worse subsequent cognitive decline. Independent of cognitive decline, initial psychosis was associated with worse subsequent increases in dependence. Rates of increase in agitation/aggression separately correlated with rates of declines in both cognition and independence. CONCLUSIONS Although purely observational, our findings support the poor prognosis associated with psychosis and depression in AD. Results also show that agitation/aggression tracks declines in cognition and independence independently over time. Targeted intervention for individual BPSD, particularly psychosis, could have broad effects not only on patient well-being but also on care costs and family burden.
Collapse
Affiliation(s)
- Laura B Zahodne
- Cognitive Neuroscience Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University College of Physicians and Surgeons, New York, NY
| | - Katherine Ornstein
- Department of Geriatrics and Palliative Medicine, Mount Sinai School of Medicine, New York, NY
| | - Stephanie Cosentino
- Cognitive Neuroscience Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University College of Physicians and Surgeons, New York, NY
| | - D P Devanand
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY
| | - Yaakov Stern
- Cognitive Neuroscience Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University College of Physicians and Surgeons, New York, NY.
| |
Collapse
|
25
|
Zheng X, Demirci FY, Barmada MM, Richardson GA, Lopez OL, Sweet RA, Kamboh MI, Feingold E. A rare duplication on chromosome 16p11.2 is identified in patients with psychosis in Alzheimer's disease. PLoS One 2014; 9:e111462. [PMID: 25379732 PMCID: PMC4224411 DOI: 10.1371/journal.pone.0111462] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/29/2014] [Indexed: 01/10/2023] Open
Abstract
Epidemiological and genetic studies suggest that schizophrenia and autism may share genetic links. Besides common single nucleotide polymorphisms, recent data suggest that some rare copy number variants (CNVs) are risk factors for both disorders. Because we have previously found that schizophrenia and psychosis in Alzheimer's disease (AD+P) share some genetic risk, we investigated whether CNVs reported in schizophrenia and autism are also linked to AD+P. We searched for CNVs associated with AD+P in 7 recurrent CNV regions that have been previously identified across autism and schizophrenia, using the Illumina HumanOmni1-Quad BeadChip. A chromosome 16p11.2 duplication CNV (chr16: 29,554,843-30,105,652) was identified in 2 of 440 AD+P subjects, but not in 136 AD subjects without psychosis, or in 593 AD subjects with intermediate psychosis status, or in 855 non-AD individuals. The frequency of this duplication CNV in AD+P (0.46%) was similar to that reported previously in schizophrenia (0.46%). This duplication CNV was further validated using the NanoString nCounter CNV Custom CodeSets. The 16p11.2 duplication has been associated with developmental delay, intellectual disability, behavioral problems, autism, schizophrenia (SCZ), and bipolar disorder. These two AD+P patients had no personal of, nor any identified family history of, SCZ, bipolar disorder and autism. To the best of our knowledge, our case report is the first suggestion that 16p11.2 duplication is also linked to AD+P. Although rare, this CNV may have an important role in the development of psychosis.
Collapse
Affiliation(s)
- Xiaojing Zheng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - F. Yesim Demirci
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - M. Michael Barmada
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Gale A. Richardson
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Oscar L. Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VISN 4 Mental Illness Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Robert A. Sweet
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VISN 4 Mental Illness Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - M. Ilyas Kamboh
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VISN 4 Mental Illness Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Eleanor Feingold
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
26
|
Mei L, Nave KA. Neuregulin-ERBB signaling in the nervous system and neuropsychiatric diseases. Neuron 2014; 83:27-49. [PMID: 24991953 DOI: 10.1016/j.neuron.2014.06.007] [Citation(s) in RCA: 413] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neuregulins (NRGs) comprise a large family of growth factors that stimulate ERBB receptor tyrosine kinases. NRGs and their receptors, ERBBs, have been identified as susceptibility genes for diseases such as schizophrenia (SZ) and bipolar disorder. Recent studies have revealed complex Nrg/Erbb signaling networks that regulate the assembly of neural circuitry, myelination, neurotransmission, and synaptic plasticity. Evidence indicates there is an optimal level of NRG/ERBB signaling in the brain and deviation from it impairs brain functions. NRGs/ERBBs and downstream signaling pathways may provide therapeutic targets for specific neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Goettingen, Germany.
| |
Collapse
|
27
|
Cao L, Deng W, Guan L, Yang Z, Lin Y, Ma X, Li X, Liu Y, Ye B, Lao G, Chen Y, Liang H, Wu Y, Ou Y, Huang W, Liu W, Wang Q, Wang Y, Zhao L, Li T, Hu X. Association of the 3' region of the neuregulin 1 gene with bipolar I disorder in the Chinese Han population. J Affect Disord 2014; 162:81-8. [PMID: 24767010 DOI: 10.1016/j.jad.2014.03.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Based on the function of neuregulin 1 (NRG1) in neurodevelopment, susceptibility to bipolar disorder presumably involves this gene. The 3' region of NRG1 contains the majority of the coding exons, and transcripts from this region encode 8 of the 9 known NRG1 isoforms; therefore, this region is likely to be predominant versus the 5' region in terms of their relative contributions to NRG1 function. We investigated the association between the 3' region of the NRG1 gene and bipolar I disorder (BPI) in the Chinese Han population and performed further analyses depending on the presence or absence of psychotic features. METHODS A total of 385 BPI patients and 475 healthy controls were recruited for this study. Thirty tag single nucleotide polymorphisms (SNPs) across the 3' region of the NRG1 gene were genotyped for allelic and haplotypic associations with BPI and subgroups with psychotic features (BPI-P) or without psychotic features (BPI-NP). RESULTS Individual marker analysis showed that 2 SNPs (rs12547858 and rs6468121) in this region were significantly associated with BPI. Moreover, subgroup analyses showed significant but marginal associations of rs6468121 with BPI-P and rs3757933 with BPI-NP. Haplotype analyses showed that 6 haplotypes were associated with BPI only. LIMITATIONS The sample size was relatively small. The investigated tag SNPs only represented 83% of the information on the targeted region. There might be a retrospective bias in the subgroup analyses. CONCLUSION The results suggest that the 3' region of the NRG1 gene plays a role in BPI susceptibility in the Chinese Han population. In addition, the preliminary results show that BPI with psychotic features and BPI without psychotic features may constitute different sub-phenotypes; however, this finding should be confirmed in a larger population sample.
Collapse
Affiliation(s)
- Liping Cao
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China.
| | - Wenhao Deng
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Lijie Guan
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Zhenxing Yang
- Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yin Lin
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China; Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xiaohong Ma
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China; Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xuan Li
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Yuping Liu
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Biyu Ye
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Guohui Lao
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Yuwei Chen
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Huiwei Liang
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Yuanfei Wu
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Yufen Ou
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Weijie Huang
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Wentao Liu
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China
| | - Qiang Wang
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China; Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yingcheng Wang
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Brain Hospital, Guangzhou, Guangdong, PR China; Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Liansheng Zhao
- Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Tao Li
- Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xun Hu
- Psychiatric Laboratory and Mental Health Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
28
|
Ford AH. Neuropsychiatric aspects of dementia. Maturitas 2014; 79:209-15. [PMID: 24794580 DOI: 10.1016/j.maturitas.2014.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 03/28/2014] [Accepted: 04/01/2014] [Indexed: 11/15/2022]
Abstract
Dementia affects approximately 6.5% of people over the age of 65. Whilst cognitive impairment is central to the dementia concept, neuropsychiatric symptoms are invariably present at some stage of the illness. Neuropsychiatric symptoms result in a number of negative outcomes for the individual and their caregivers and are associated with higher rates of institutionalization and mortality. A number of factors have been associated with neuropsychiatric symptoms including neurobiological changes, dementia type, and illness severity and duration. Specific patient, caregiver and environmental factors are also important. Neuropsychiatric symptoms can be broadly divided into four clusters: psychotic symptoms, mood/affective symptoms, apathy, and agitation/aggression. Neuropsychiatric symptoms tend to persist over time although differing symptom profiles exist at various stages of the illness. Assessment should take into account the presenting symptoms together with an appreciation of the myriad of likely underlying causes for the symptoms. A structured assessment/rating tool can be helpful. Management should focus on non-pharmacological measures initially with pharmacological approaches reserved for more troubling symptoms. Pharmacological approaches should target specific symptoms although the evidence-base for pharmacological management is quite modest. Any medication trial should include an adequate appreciation of the risk-benefit profile in individual patients and discussion of these with both the individual and their caregiver.
Collapse
Affiliation(s)
- Andrew H Ford
- Western Australian Centre for Health & Ageing (M573), Centre for Medical Research, University of Western Australia, 35 Stirling Highway, Crawley, Perth, WA 6009, Australia.
| |
Collapse
|
29
|
Murray PS, Kumar S, Demichele-Sweet MAA, Sweet RA. Psychosis in Alzheimer's disease. Biol Psychiatry 2014; 75:542-52. [PMID: 24103379 PMCID: PMC4036443 DOI: 10.1016/j.biopsych.2013.08.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/22/2013] [Accepted: 08/19/2013] [Indexed: 12/13/2022]
Abstract
Psychotic symptoms, delusions and hallucinations, occur in approximately 50% of individuals with Alzheimer's disease (AD) (AD with psychosis [AD + P]). Pharmacotherapies for AD + P have limited efficacy and can increase short-term mortality. These observations have motivated efforts to identify the underlying biology of AD + P. Psychosis in AD indicates a more severe phenotype, with more rapid cognitive decline beginning even before psychosis onset. Neuroimaging studies suggest that AD + P subjects demonstrate greater cortical synaptic impairments than AD subjects without psychosis, reflected in reduced gray matter volume, reduced regional blood flow, and reduced regional glucose metabolism. Neuroimaging and available postmortem evidence further indicate that the impairments in AD + P, relative to AD subjects without psychosis, are localized to neocortex rather than medial temporal lobe. Neuropathologic studies provide consistent evidence of accelerated accumulation of hyperphosphorylated microtubule associated protein tau in AD + P. Finally, studies of familial aggregation of AD + P have established that the risk for psychosis in AD is, in part, genetically mediated. Although no genes are established as associated with AD + P, the first genome-wide association study of AD + P has generated some promising leads. The study of the neurobiology of AD + P is rapidly accelerating and may be poised for translational discovery. This process can be enhanced by identifying points of convergence and divergence with the neurobiology of AD proper and of schizophrenia, by innovative extension of current approaches, and by development of relevant animal models.
Collapse
Affiliation(s)
- Patrick S Murray
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Veterans Integrated Service Network 4 Mental Illness Research, Education and Clinical Center, US Department of Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Sanjeev Kumar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania; Veterans Integrated Service Network 4 Mental Illness Research, Education and Clinical Center, US Department of Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.
| |
Collapse
|
30
|
Ubhi K, Rockenstein E, Kragh C, Inglis C, Spencer B, Michael S, Mante M, Adame A, Galasko D, Masliah E. Widespread microRNA dysregulation in multiple system atrophy - disease-related alteration in miR-96. Eur J Neurosci 2014; 39:1026-1041. [PMID: 24304186 PMCID: PMC4052839 DOI: 10.1111/ejn.12444] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 10/26/2013] [Accepted: 11/05/2013] [Indexed: 12/15/2022]
Abstract
MicroRNA (miRNA) are short sequences of RNA that function as post-transcriptional regulators by binding to target mRNA transcripts resulting in translational repression. A number of recent studies have identified miRNA as being involved in neurodegenerative disorders including Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the role of miRNA in multiple system atrophy (MSA), a progressive neurodegenerative disorder characterized by oligodendroglial accumulation of alpha-synuclein remains unexamined. In this context, this study examined miRNA profiles in MSA cases compared with controls and in transgenic (tg) models of MSA compared with non-tg mice. The results demonstrate a widespread dysregulation of miRNA in MSA cases, which is recapitulated in the murine models. The study employed a cross-disease, cross-species approach to identify miRNA that were either specifically dysregulated in MSA or were commonly dysregulated in neurodegenerative conditions such as Alzheimer's disease, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal degeneration or the tg mouse model equivalents of these disorders. Using this approach we identified a number of miRNA that were commonly dysregulated between disorders and those that were disease-specific. Moreover, we identified miR-96 as being up-regulated in MSA. Consistent with the up-regulation of miR-96, mRNA and protein levels of members of the solute carrier protein family SLC1A1 and SLC6A6, miR-96 target genes, were down-regulated in MSA cases and a tg model of MSA. These results suggest that miR-96 dysregulation may play a role in MSA and its target genes may be involved in the pathogenesis of MSA.
Collapse
Affiliation(s)
- Kiren Ubhi
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Christine Kragh
- Department of Biomedicine, University of Aarhus, DK-8000 Aarhus, Denmark
| | - Chandra Inglis
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Brian Spencer
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Sarah Michael
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, California 92093-0624, USA
- Department of Pathology, University of California, San Diego, California 92093-0624, USA
| |
Collapse
|
31
|
Abstract
Psychosis occurs in approximately half of patients with Alzheimer disease (AD with psychosis, AD+P). AD+P patients have more rapid cognitive decline, greater behavioral symptoms, and higher mortality than do AD patients without psychosis. Studies in three independent cohorts have shown that psychosis in AD aggregates in families, with estimated heritability of 29.5 - 60.8%. These findings have motivated studies to investigate and uncover the genes responsible for the development of psychosis, with the ultimate goal of identifying potential biologic mechanisms that may serve as leads to specific therapies. Linkage analyses have implicated loci on chromosomes 2, 6, 7, 8, 15, and 21 with AD+P. Association studies of APOE do not support it as a risk gene for psychosis in AD. No other candidate genes, such as neurodegenerative and monoamine genes, show conclusive evidence of association with AD+P. However, a recent genome-side association study has produced some promising leads, including among them genes that have been associated with schizophrenia. This review summarizes the current knowledge of the genetic basis of AD+P.
Collapse
Affiliation(s)
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
- VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA
| |
Collapse
|
32
|
Kukshal P, Bhatia T, Bhagwat AM, Gur RE, Gur RC, Deshpande SN, Nimgaonkar VL, Thelma BK. Association study of neuregulin-1 gene polymorphisms in a North Indian schizophrenia sample. Schizophr Res 2013; 144:24-30. [PMID: 23360725 PMCID: PMC4040109 DOI: 10.1016/j.schres.2012.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/20/2012] [Accepted: 12/17/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neuregulin-1 (NRG1) gene polymorphisms have been proposed as risk factors for several common disorders. Associations with cognitive variation have also been tested. With regard to schizophrenia (SZ) risk, studies of Caucasian ancestry samples indicate associations more consistently than East Asian samples, suggesting heterogeneity. To exploit the differences in linkage disequilibrium (LD) structure across ethnic groups, we conducted a SZ case-control study (that included cognitive evaluations) in a sample from the north Indian population. METHODS NRG1 variants (n=35 SNPs, three microsatellite markers) were initially analyzed among cases (DSM IV criteria, n=1007) and controls (n=1019, drawn from two groups) who were drawn from the same geographical region in North India. Nominally significant associations with SZ were next analyzed in relation to neurocognitive measures estimated with a computerized neurocognitive battery in a subset of the sample (n=116 cases, n=170 controls). RESULTS Three variants and one microsatellite showed allelic association with SZ (rs35753505, rs4733263, rs6994992, and microsatellite 420M9-1395, p≤0.05 uncorrected for multiple comparisons). A six marker haplotype 221121 (rs35753505-rs6994992-rs1354336-rs10093107-rs3924999-rs11780123) showed (p=0.0004) association after Bonferroni corrections. Regression analyses with the neurocognitive measures showed nominal (uncorrected) associations with emotion processing and attention at rs35753505 and rs6994992, respectively. CONCLUSIONS Suggestive associations with SZ and SZ-related neurocognitive measures were detected with two SNPs from the NRG1 promoter region in a north Indian cohort. The functional role of the alleles merits further investigation.
Collapse
Affiliation(s)
- Prachi Kukshal
- Department of Genetics, University of Delhi South campus, Benito
Juarez Road, New Delhi – 110 021, India
- C.B. Patel Research Centre, Vile Parle (West), Mumbai, India
| | - Triptish Bhatia
- Department of Psychiatry, Dr. RML Hospital, New Delhi – 110
001, India
| | - A. M. Bhagwat
- C.B. Patel Research Centre, Vile Parle (West), Mumbai, India
| | - Raquel E. Gur
- Department of Psychiatry, Neuropsychiatry Section, University of
Pennsylvania, Philadelphia, PA, USA
| | - Ruben C. Gur
- Department of Psychiatry, Neuropsychiatry Section, University of
Pennsylvania, Philadelphia, PA, USA
| | | | - Vishwajit L. Nimgaonkar
- Department of Psychiatry and Human Genetics, Western Psychiatric
Institute and Clinic, University of Pittsburgh School of Medicine and Graduate
School of Public Health, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - B. K. Thelma
- Department of Genetics, University of Delhi South campus, Benito
Juarez Road, New Delhi – 110 021, India
| |
Collapse
|
33
|
Unveiling clusters of RNA transcript pairs associated with markers of Alzheimer's disease progression. PLoS One 2012; 7:e45535. [PMID: 23029078 PMCID: PMC3448659 DOI: 10.1371/journal.pone.0045535] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 08/23/2012] [Indexed: 12/17/2022] Open
Abstract
Background One primary goal of transcriptomic studies is identifying gene expression patterns correlating with disease progression. This is usually achieved by considering transcripts that independently pass an arbitrary threshold (e.g. p<0.05). In diseases involving severe perturbations of multiple molecular systems, such as Alzheimer’s disease (AD), this univariate approach often results in a large list of seemingly unrelated transcripts. We utilised a powerful multivariate clustering approach to identify clusters of RNA biomarkers strongly associated with markers of AD progression. We discuss the value of considering pairs of transcripts which, in contrast to individual transcripts, helps avoid natural human transcriptome variation that can overshadow disease-related changes. Methodology/Principal Findings We re-analysed a dataset of hippocampal transcript levels in nine controls and 22 patients with varying degrees of AD. A large-scale clustering approach determined groups of transcript probe sets that correlate strongly with measures of AD progression, including both clinical and neuropathological measures and quantifiers of the characteristic transcriptome shift from control to severe AD. This enabled identification of restricted groups of highly correlated probe sets from an initial list of 1,372 previously published by our group. We repeated this analysis on an expanded dataset that included all pair-wise combinations of the 1,372 probe sets. As clustering of this massive dataset is unfeasible using standard computational tools, we adapted and re-implemented a clustering algorithm that uses external memory algorithmic approach. This identified various pairs that strongly correlated with markers of AD progression and highlighted important biological pathways potentially involved in AD pathogenesis. Conclusions/Significance Our analyses demonstrate that, although there exists a relatively large molecular signature of AD progression, only a small number of transcripts recurrently cluster with different markers of AD progression. Furthermore, considering the relationship between two transcripts can highlight important biological relationships that are missed when considering either transcript in isolation.
Collapse
|
34
|
Reeves SJ, Gould RL, Powell JF, Howard RJ. Origins of delusions in Alzheimer's disease. Neurosci Biobehav Rev 2012; 36:2274-87. [PMID: 22910677 DOI: 10.1016/j.neubiorev.2012.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/19/2012] [Accepted: 08/03/2012] [Indexed: 11/26/2022]
Abstract
Research over the past two decades supports a shared aetiology for delusions in Alzheimer's disease (AD) and schizophrenia. Functional networks involved in salience attribution and belief evaluation have been implicated in the two conditions, and striatal D2/3 receptors are increased to a comparable extent. Executive/frontal deficits are common to both disorders and predict emergent symptoms. Putative risk genes for schizophrenia, which may modify the AD process, have been more strongly implicated in delusions than those directly linked with late-onset AD. Phenotypic correlates of delusions in AD may be dependent upon delusional subtype. Persecutory delusions occur early in the disease and are associated with neurochemical and neuropathological changes in frontostriatal circuits. In contrast, misidentification delusions are associated with greater global cognitive deficits and advanced limbic pathology. It is unclear whether the two subtypes are phenomenologically and biologically distinct or are part of a continuum, in which misidentification delusions manifest increasingly as the pathological process extends. This has treatment implications, particularly if they are found to have discrete chemical and/or pathological markers.
Collapse
Affiliation(s)
- Suzanne J Reeves
- Department of Old Age Psychiatry, Institute of Psychiatry, Kings College London, De Crespigny Park, Camberwell, London SE58AF, UK.
| | | | | | | |
Collapse
|
35
|
Jellinger KA. Cerebral correlates of psychotic syndromes in neurodegenerative diseases. J Cell Mol Med 2012; 16:995-1012. [PMID: 21418522 PMCID: PMC4365880 DOI: 10.1111/j.1582-4934.2011.01311.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 03/01/2011] [Indexed: 12/20/2022] Open
Abstract
Psychosis has been recognized as a common feature in neurodegenerative diseases and a core feature of dementia that worsens most clinical courses. It includes hallucinations, delusions including paranoia, aggressive behaviour, apathy and other psychotic phenomena that occur in a wide range of degenerative disorders including Alzheimer's disease, synucleinopathies (Parkinson's disease, dementia with Lewy bodies), Huntington's disease, frontotemporal degenerations, motoneuron and prion diseases. Many of these psychiatric manifestations may be early expressions of cognitive impairment, but often there is a dissociation between psychotic/behavioural symptoms and the rather linear decline in cognitive function, suggesting independent pathophysiological mechanisms. Strictly neuropathological explanations are likely to be insufficient to explain them, and a large group of heterogeneous factors (environmental, neurochemical changes, genetic factors, etc.) may influence their pathogenesis. Clinico-pathological evaluation of behavioural and psychotic symptoms (PS) in the setting of neurodegenerative and dementing disorders presents a significant challenge for modern neurosciences. Recognition and understanding of these manifestations may lead to the development of more effective preventive and therapeutic options that can serve to delay long-term progression of these devastating disorders and improve the patients' quality of life. A better understanding of the pathophysiology and distinctive pathological features underlying the development of PS in neurodegenerative diseases may provide important insights into psychotic processes in general.
Collapse
|
36
|
Proitsi P, Powell JF. Missense substitutions associated with behavioural disturbances in Alzheimer's disease (AD). Brain Res Bull 2012; 88:394-405. [PMID: 22414959 DOI: 10.1016/j.brainresbull.2012.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/03/2012] [Accepted: 02/24/2012] [Indexed: 11/18/2022]
Abstract
Behavioural and psychological symptoms in dementia, or BPSD, occur in the majority of Alzheimer's disease (AD) patients. They are associated with considerable patient morbidity and greater care-giver stress. There is some evidence suggesting that BPSD have a genetic component and a large number of studies have examined the association of candidate genes with these symptoms. This review provides a comprehensive summary of all the published studies investigating the association of candidate gene missense substitutions with BPSD. Missense substitutions could potentially alter protein function or render the protein non-functional, resulting in phenotypic consequences. More than 80 studies investigating the association of 8 missense substitutions in 7 genes with BPSD were identified. However, results of these studies are contradictory and do not provide firm support for these associations. Larger studies and more systematic approaches will delineate the association of missense substitutions with behavioural symptoms in AD.
Collapse
Affiliation(s)
- Petroula Proitsi
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, London SE5 8AF, UK.
| | | |
Collapse
|
37
|
Abstract
Neuregulin 1 (NRG1) is a trophic factor that has been implicated in neural development, neurotransmission, and synaptic plasticity. NRG1 has multiple isoforms that are generated by usage of different promoters and alternative splicing of a single gene. However, little is known about NRG1 isoform composition profile, whether it changes during development, or the underlying mechanisms. We found that each of the six types of NRG1 has a distinct expression pattern in the brain at different ages, resulting in a change in NRG1 isoform composition. In both human and rat, the most dominant are types III and II, followed by either type I or type V, while types IV and VI are the least abundant. The expression of NRG1 isoforms is higher in rat brains at ages of E13 and P5 (in particular type V), suggesting roles in early neural development and in the neonatal critical period. At the cellular level, the majority of NRG1 isoforms (types I, II, and III) are expressed in excitatory neurons, although they are also present in GABAergic neurons and astrocytes. Finally, the expression of each NRG1 isoform is distinctly regulated by neuronal activity, which causes significant increase in type I and IV NRG1 levels. Neuronal activity regulation of type IV expression requires a CRE cis-element in the 5' untranslated region (UTR) that binds to CREB. These results indicate that expression of NRG1 isoforms is regulated by distinct mechanisms, which may contribute to versatile functions of NRG1 and pathologic mechanisms of brain disorders such as schizophrenia.
Collapse
|
38
|
Lack of Associations of Neuregulin 1 Variations with Schizophrenia and Smooth Pursuit Eye Movement Abnormality in a Korean Population. J Mol Neurosci 2011; 46:476-82. [DOI: 10.1007/s12031-011-9619-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 08/04/2011] [Indexed: 10/17/2022]
|
39
|
Hollingworth P, Harold D, Jones L, Owen MJ, Williams J. Alzheimer's disease genetics: current knowledge and future challenges. Int J Geriatr Psychiatry 2011; 26:793-802. [PMID: 20957767 DOI: 10.1002/gps.2628] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 07/29/2010] [Indexed: 11/08/2022]
Abstract
Alzheimer's disease (AD) is highly heritable, but genetically complex. Recently, three large-scale genome-wide association studies have made substantial breakthroughs in disentangling the genetic architecture of the disease. These studies combined include data from over 43 000 independent individuals and provide compelling evidence that variants in four novel susceptibility genes (CLU, PICALM, CR1, BIN1) are associated with disease risk. These findings are tremendously exciting, not only in providing new avenues for exploration, but also highlighting the potential for further gene discovery when larger samples are analysed. Here we discuss progress to date in identifying risk genes for dementia, ways forward and how current findings are refining previous ideas and defining new putative primary disease mechanisms.
Collapse
Affiliation(s)
- Paul Hollingworth
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK.
| | | | | | | | | |
Collapse
|
40
|
Woo RS, Lee JH, Yu HN, Song DY, Baik TK. Expression of ErbB4 in the neurons of Alzheimer's disease brain and APP/PS1 mice, a model of Alzheimer's disease. Anat Cell Biol 2011; 44:116-27. [PMID: 21829755 PMCID: PMC3145840 DOI: 10.5115/acb.2011.44.2.116] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 04/21/2011] [Accepted: 05/02/2011] [Indexed: 01/10/2023] Open
Abstract
Neuregulin-1 (NRG1) plays important roles in the development and plasticity of the brain, and has also been reported to exhibit potent neuroprotective properties. Although ErbB4, a key NRG1 receptor, is expressed in multiple regions in the adult animal brain, little is known about its role in Alzheimer's disease (AD). AD is characterized by progressive impairment of cognition and behavioral disturbance that strongly correlate with degeneration and death of neurons in the cerebral cortex and limbic brain areas, such as the hippocampus and the amygdala. Here, we show that the ErbB4 and phospho-ErbB4 immunoreactivities were higher intensity in the neurons of the CA1-2 transitional field of AD brains as compared to age-matched controls. Also, ErbB4 expression was increased in the neurons of the cortico medial nucleus amygdala, human basal forebrain and superior frontal gyrus of AD brains. In cerebral cortex and hippocampus of amyloid precursor protein/presenilin 1 double transgenic mice, ErbB4 immunoreactivity significantly increased in comparison to age-matched wild type control. These results suggest that up-regulating of ErbB4 immunoreactivity may involve in the progression of pathology of AD.
Collapse
Affiliation(s)
- Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, Korea
| | | | | | | | | |
Collapse
|
41
|
Flirski M, Sobow T, Kloszewska I. Behavioural genetics of Alzheimer's disease: a comprehensive review. Arch Med Sci 2011; 7:195-210. [PMID: 22291757 PMCID: PMC3258720 DOI: 10.5114/aoms.2011.22068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 07/12/2010] [Accepted: 08/24/2010] [Indexed: 12/16/2022] Open
Abstract
Behavioural and psychological symptoms of dementia (BPSD) are present in the course of the illness in up to 90% of patients with Alzheimer's disease (AD). They are the main source of caregiver burden and one of the major factors contributing to early institutionalization. The involvement of a genetic component in BPSD aetiology seems beyond controversy, though the exact significance of particular polymorphisms is uncertain in the majority of cases. Multiple genes have been assessed for their putative influence on BPSD risk. In this paper we review the behavioural genetics of AD, particularly the importance, with respect to BPSD risk, of genes coding for apolipoprotein E and proteins involved in the process of neurotransmission: serotonin receptors, serotonin transporter, COMT, MAO-A, tryptophan hydroxylase and dopamine receptors. A general conclusion is the striking inconsistency of the findings, unsurprising in the field of psychiatric genetics. The potential reasons for such discrepancy are exhaustively discussed.
Collapse
Affiliation(s)
- Marcin Flirski
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Poland
| | | | | |
Collapse
|
42
|
DeMichele-Sweet MA, Sweet RA. Genetics of psychosis in Alzheimer's disease: a review. J Alzheimers Dis 2010; 19:761-80. [PMID: 20157235 DOI: 10.3233/jad-2010-1274] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In and of itself, late-onset Alzheimer's disease (AD) can be a devastating illness. However, a sub-group of AD patients develop psychosis as the disease progresses. These patients have an added burden of greater cognitive impairment, higher rates of institutionalization, and higher mortality than AD patients without psychosis. While the etiopathogenesis such as psychosis in AD (AD+P) is not known, mounting evidence accrued over the past ten years indicates that AD+P represents a distinct phenotype with a genetic basis. Elucidating the genetic mechanism of AD+P is crucial if better pharmaceutical treatments are to be developed for these patients. The goal of this review is to summarize what is currently known regarding the genetic basis of psychosis in AD. Specific attention is given to familial aggregation and heritability, linkage to chromosomal loci, and associations of candidate genes of APOE and the monoamine neurotransmitter system.
Collapse
|
43
|
Abstract
Presenilins form the catalytic part of the gamma-secretases, protein complexes that are responsible for the intramembranous cleavage of transmembrane proteins. The presenilins are involved in several biological functions, but are best known for their role in the generation of the beta-amyloid (Abeta) peptide in Alzheimer's disease and are therefore thought to be important drug targets for this disorder. Mutations in the presenilin genes cause early-onset familial Alzheimer's disease, but mutation carriers have substantial phenotypic heterogeneity. Recent evidence implicating presenilin mutations in non-Alzheimer's dementias, including frontotemporal dementia and Lewy body dementia, warrants further investigation. An increased understanding of the diversity of the molecular cell biology of the gamma-secretase complex and the effects of clinical mutations in the presenilin genes might help pave the way for improved development of drugs that are designed to target gamma-secretase enzymatic activity in Alzheimer's disease and potentially in other neurological diseases.
Collapse
Affiliation(s)
- Bruno A Bergmans
- Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium; Center for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | | |
Collapse
|
44
|
Haraldsson HM, Ettinger U, Magnusdottir BB, Ingason A, Hutton SB, Sigmundsson T, Sigurdsson E, Petursson H. Neuregulin-1 genotypes and eye movements in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2010; 260:77-85. [PMID: 19575259 DOI: 10.1007/s00406-009-0032-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 06/15/2009] [Indexed: 01/26/2023]
Abstract
Neuregulin-1 (NRG-1) is a putative susceptibility gene for schizophrenia but the neurocognitive processes that may involve NRG-1 in schizophrenia are unknown. Deficits in antisaccade (AS) and smooth pursuit eye movements (SPEM) are promising endophenotypes, which may be associated with brain dysfunctions underlying the pathophysiology of schizophrenia. The aim of this study was to investigate the associations of NRG-1 genotypes with AS and SPEM in schizophrenia patients and healthy controls. Patients (N = 113) and controls (N = 106) were genotyped for two NRG-1 single nucleotide polymorphisms (SNPs); SNP8NRG222662, a surrogate marker for the originally described Icelandic NRG-1 risk haplotype, and SNP8NRG243177, which has recently been associated with individual differences in brain function. Subjects underwent infrared oculographic assessment of AS and SPEM. The study replicates previous findings of impaired AS and SPEM performance in schizophrenia patients (all P < 0.005; all d = 0.5-1.5). SNP8NRG243177 risk allele carriers had marginally increased variability of AS spatial error (P = 0.050, d = 0.3), but there were no significant genotype effects on other eye movement variables and no significant diagnosis-by-genotype interactions. Generally, risk allele carriers (G allele for SNP8NRG222662 and T allele for SNP8NRG243177) had numerically worse performance than non-carriers on most AS and SPEM variables. The results do not suggest that NRG-1 genotype significantly affects AS and SPEM task performance. However, the power of the sample to identify small effects is limited and the possibility of a type II error must be kept in mind. Larger samples may be needed to reliably investigate such gene effects on oculomotor endophenotypes.
Collapse
Affiliation(s)
- H Magnus Haraldsson
- Division of Psychiatry, Landspitali University Hospital, Hringbraut, 101, Reykjavik, Iceland.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Tabarés-Seisdedos R, Rubenstein JLR. Chromosome 8p as a potential hub for developmental neuropsychiatric disorders: implications for schizophrenia, autism and cancer. Mol Psychiatry 2009; 14:563-89. [PMID: 19204725 DOI: 10.1038/mp.2009.2] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Defects in genetic and developmental processes are thought to contribute susceptibility to autism and schizophrenia. Presumably, owing to etiological complexity identifying susceptibility genes and abnormalities in the development has been difficult. However, the importance of genes within chromosomal 8p region for neuropsychiatric disorders and cancer is well established. There are 484 annotated genes located on 8p; many are most likely oncogenes and tumor-suppressor genes. Molecular genetics and developmental studies have identified 21 genes in this region (ADRA1A, ARHGEF10, CHRNA2, CHRNA6, CHRNB3, DKK4, DPYSL2, EGR3, FGF17, FGF20, FGFR1, FZD3, LDL, NAT2, NEF3, NRG1, PCM1, PLAT, PPP3CC, SFRP1 and VMAT1/SLC18A1) that are most likely to contribute to neuropsychiatric disorders (schizophrenia, autism, bipolar disorder and depression), neurodegenerative disorders (Parkinson's and Alzheimer's disease) and cancer. Furthermore, at least seven nonprotein-coding RNAs (microRNAs) are located at 8p. Structural variants on 8p, such as copy number variants, microdeletions or microduplications, might also contribute to autism, schizophrenia and other human diseases including cancer. In this review, we consider the current state of evidence from cytogenetic, linkage, association, gene expression and endophenotyping studies for the role of these 8p genes in neuropsychiatric disease. We also describe how a mutation in an 8p gene (Fgf17) results in a mouse with deficits in specific components of social behavior and a reduction in its dorsomedial prefrontal cortex. We finish by discussing the biological connections of 8p with respect to neuropsychiatric disorders and cancer, despite the shortcomings of this evidence.
Collapse
Affiliation(s)
- R Tabarés-Seisdedos
- Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, CIBER-SAM, University of Valencia, Valencia, Spain.
| | | |
Collapse
|
46
|
Sims R, Hollingworth P, Moskvina V, Dowzell K, O'Donovan MC, Powell J, Lovestone S, Brayne C, Rubinsztein D, Owen MJ, Williams J, Abraham R. Evidence that variation in the oligodendrocyte lineage transcription factor 2 (OLIG2) gene is associated with psychosis in Alzheimer's disease. Neurosci Lett 2009; 461:54-9. [PMID: 19477230 DOI: 10.1016/j.neulet.2009.05.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 05/14/2009] [Accepted: 05/18/2009] [Indexed: 01/29/2023]
Abstract
Psychotic symptoms are common in individuals with Alzheimer's disease (AD), and define a phenotype associated with more rapid cognitive and functional decline. Evidence suggests that psychotic symptoms may be influenced by genetic factors, and recent studies in schizophrenia, bipolar affective disorder (BPAD) and Alzheimer's disease with psychosis (AD+P) suggest that psychosis susceptibility or modifier genes may act across diseases. We hypothesised that oligodendrocyte lineage transcription factor 2 (OLIG2), a regulator of white matter development and a candidate gene for schizophrenia, may also be associated with psychotic symptoms in AD. We genotyped 11 SNPs in OLIG2 previously tested for association with schizophrenia [L. Georgieva, V. Moskvina, T. Peirce, N. Norton, N.J. Bray, L. Jones, P. Holmans, S. Macgregor, S. Zammit, J. Wilkinson, H. Williams, I. Nikolov, N. Williams, D. Ivanov, K.L. Davis, V. Haroutunian, J.D. Buxbaum, N. Craddock, G. Kirov, M.J. Owen, M.C. O'Donovan, Convergent evidence that oligodendrocyte lineage transcription factor 2 (OLIG2) and interacting genes influence susceptibility to schizophrenia, Proc. Natl. Acad. Sci. U.S.A. 103 (33) (2006) 12469-12474] and tested these for association with AD and AD+P. Significant evidence for association of psychotic symptoms within cases was identified for two SNPs, rs762237 (allelic P=0.002, OR=1.42, corrected P=0.019) and rs2834072 (allelic P=0.004, OR=1.41, corrected P=0.05).
Collapse
Affiliation(s)
- R Sims
- Department of Psychological Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF144XN, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bora E, Yucel M, Fornito A, Berk M, Pantelis C. Major psychoses with mixed psychotic and mood symptoms: are mixed psychoses associated with different neurobiological markers? Acta Psychiatr Scand 2008; 118:172-87. [PMID: 18699952 DOI: 10.1111/j.1600-0447.2008.01230.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Evidence related to overlapping clinical and genetic risk factors in schizophrenia and bipolar disorder (BD) have raised concerns about the validity of 'Kraepelinian dichotomy'. As controversies mainly arise in mixed psychoses that occupy the intermediate zone between schizophrenia and BD, investigating neurobiological markers of mixed psychoses may be relevant to understanding the nature of psychotic disorders. METHOD In this article, we review studies comparing magnetic resonance imaging, neuropsychological and electrophysiological findings in mixed psychoses with each other, as well as with more prototypical cases of schizophrenia and BD. RESULTS The evidence reviewed suggests that mixed psychoses may be associated with different genetic and neurobiological markers compared with prototypical forms of schizophrenia and BD. CONCLUSION These findings may be compatible with more sophisticated versions of dimensional and continuum models or, alternatively, they may suggest that there is an intermediate third category between prototypical schizophrenia and BD.
Collapse
Affiliation(s)
- E Bora
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, and Melbourne Health, ORYGEN research Centre, The University of Melbourne, Melbourne, Vic, Australia.
| | | | | | | | | |
Collapse
|
48
|
Wieduwilt MJ, Moasser MM. The epidermal growth factor receptor family: biology driving targeted therapeutics. Cell Mol Life Sci 2008; 65:1566-84. [PMID: 18259690 PMCID: PMC3060045 DOI: 10.1007/s00018-008-7440-8] [Citation(s) in RCA: 497] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The epidermal growth factor family of receptor tyrosine kinases (ErbBs) plays essential roles in regulating cell proliferation, survival, differentiation and migration. The ErbB receptors carry out both redundant and restricted functions in mammalian development and in the maintenance of tissues in the adult mammal. Loss of regulation of the ErbB receptors underlies many human diseases, most notably cancer. Our understanding of the function and complex regulation of these receptors has fueled the development of targeted therapeutic agents for human malignancies in the last 15 years. Here we review the biology of ErbB receptors, including their structure, signaling, regulation, and roles in development and disease, then briefly touch on their increasing roles as targets for cancer therapy.
Collapse
Affiliation(s)
- M. J. Wieduwilt
- Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, UCSF, Box 0875, San Francisco, CA 94143-0875 USA
| | - M. M. Moasser
- Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, UCSF, Box 0875, San Francisco, CA 94143-0875 USA
| |
Collapse
|
49
|
Chen Z, Simmons MS, Perry RT, Wiener HW, Harrell LE, Go RCP. Genetic association of neurotrophic tyrosine kinase receptor type 2 (NTRK2) With Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 2008; 147:363-9. [PMID: 17918233 DOI: 10.1002/ajmg.b.30607] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase (TRK) signaling pathway activates a wide range of downstream intracellular cascades, regulating neuronal development and plasticity, long-term potentiation, and apoptosis. The NTRK family encodes the receptors TRKA, TRKB, and TRKC, to which the neurotrophins, nerve growth factor (NGF), BDNF and neurotrophin-3 (NT-3) bind, respectively, with high affinity. Signaling through these receptors appears to be compromised in Alzheimer's disease (AD). This study is the most comprehensive investigation of genetic variants of NTRK2, and the first to show significant association between NTRK2 with AD. Fourteen single nucleotide polymorphisms (SNPs), located in 8 of 18 linkage disequilibrium (LD) blocks, were genotyped in 203 families with at least two AD affected siblings with mean age of onset (MAO) of 70.9 +/- 7.4 years and one unaffected sibling from the NIMH-ADGJ dataset. Family based association testing found no single SNP association, however, significant associations were found for two and three locus haplotypes (P = 0.012, P = 0.009, respectively) containing SNPs rsl624327, rsl443445, and rs378645. These SNPs are located in areas of the gene containing sequences that could be involved in alternative splicing and/or regulation of NTRK2. Our results suggest that NTRK2 may be a genetic susceptibility gene contributing to AD pathology.
Collapse
Affiliation(s)
- Zuomin Chen
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
50
|
Hong LE, Wonodi I, Stine OC, Mitchell BD, Thaker GK. Evidence of missense mutations on the neuregulin 1 gene affecting function of prepulse inhibition. Biol Psychiatry 2008; 63:17-23. [PMID: 17631867 PMCID: PMC3569848 DOI: 10.1016/j.biopsych.2007.05.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 05/10/2007] [Accepted: 05/11/2007] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neuregulin 1 (NRG1) is one of the leading candidate genes in schizophrenia. Rodents with NRG1 knock-out showed significantly impaired prepulse inhibition (PPI) in the original report linking NRG1 to schizophrenia. A widely used surrogate measure of psychosis in animal models, PPI is considered a schizophrenia endophenotype. We hypothesized that if NRG1 influences PPI in rodents, then it should have a similar effect on PPI in humans. METHODS We examined the potential neurophysiological effects of two nonsynonymous single nucleotide polymorphisms located on NRG1 (rs3924999 and rs10503929) on PPI. Genotyping was completed in 430 unrelated individuals, including 244 schizophrenia cases and 186 controls. PPI was available in a subgroup of 113 cases and 63 controls. RESULTS Rs3924999 genotype was significantly associated with PPI (p = .003): PPI was lowest in the subjects who were homozygous for the minor allele A/A carriers, intermediate in A/G carriers, and highest in homozygous major alleles G/G carriers. The associations persisted within cases (p = .02) and controls (p = .02) analyzed separately. An additive model suggested that rs3924999 alone contributes to 7.9% of the PPI variance. In contrast, rs10503929 genotype was not associated with PPI (p = .85). Schizophrenia patients had reduced PPI compared to control subjects (p = .04). Neither single nucleotide polymorphism was associated with schizophrenia (all p > .37). However, schizophrenia patients with abnormal PPI may be associated with rs3924999 (p = .05). CONCLUSIONS A missense mutation on rs3924999 of the neuregulin 1 gene may have a functional effect on prepulse inhibition in both schizophrenia and healthy control populations.
Collapse
Affiliation(s)
- L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, Baltimore, Maryland 21228, USA.
| | | | | | | | | |
Collapse
|