1
|
Jover E, Garaikoetxea M, Martín-Núñez E, Goñi-Olóriz M, San-Ildefonso-García S, Navarro A, Fernández-Celis A, Álvarez V, Sádaba R, Calvier L, López-Andrés N. Expression of the lymphangiogenic reelin is associated with sex-dependent calcific aortic stenosis in men. Atherosclerosis 2025; 403:119162. [PMID: 40188710 DOI: 10.1016/j.atherosclerosis.2025.119162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 01/29/2025] [Accepted: 03/10/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND AND AIMS Aortic stenosis is a major form of adult valvulopathy with strong sex-related phenotypes. Circulating reelin, a large extracellular glycoprotein, regulates lymphangiogenesis and inflammation and promotes atherosclerosis, a risk factor in aortic stenosis. We sought to investigate the sex-dependent expression of reelin in stenotic aortic valves to comprehend its role in aortic stenosis progression. METHODS Reelin was studied in aortic valves and serum samples from severe aortic stenosis and aortic regurgitation patients. In vitro calcification modelling of human valve interstitial cells (VICs) (n = 18 donors, 50 % men) was conducted for 2, 4 and 8 days. RESULTS Reelin (RELN) expression was enhanced within the fibrocalcific areas of stenotic aortic valves, especially in men. Expression of RELN was associated with angiogenic and lymphangiogenic, inflammation and osteogenic markers only in aortic stenosis but not in aortic regurgitation. The VIC, along with inflammatory cells and valve endothelial cells, expressed reelin. In vitro, we confirmed the VIC to display sex-dependent responses as those reported within the valve. Male VICs expressed higher RELN than women's, and that was significantly associated with enhanced Dab2/Akt/NFkB signaling as well as with lymphangiogenesis, inflammation, and osteogenesis markers. CONCLUSIONS This study suggests a sex-dependent expression of reelin in stenotic aortic valves. This observation is partly due to different responses in VIC between men and women. In men, reelin was associated with inflammation, angiogenesis, lymphangiogenesis, and osteogenesis, which contributes to more calcific phenotypes, clinically relevant in male patients. However, further mechanistic studies are necessary to fully understand these processes. It's important to note that these findings were not reflected in circulating levels of reelin.
Collapse
Affiliation(s)
- Eva Jover
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Miriam Goñi-Olóriz
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Susana San-Ildefonso-García
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Adela Navarro
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Virginia Álvarez
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Rafael Sádaba
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Laurent Calvier
- Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas TX, USA.
| | - Natalia López-Andrés
- Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain; F-CRIN, INI-CRCT, Cardiovascular Translational Research. Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.
| |
Collapse
|
2
|
Lopera F, Marino C, Chandrahas AS, O'Hare M, Villalba-Moreno ND, Aguillon D, Baena A, Sanchez JS, Vila-Castelar C, Ramirez Gomez L, Chmielewska N, Oliveira GM, Littau JL, Hartmann K, Park K, Krasemann S, Glatzel M, Schoemaker D, Gonzalez-Buendia L, Delgado-Tirado S, Arevalo-Alquichire S, Saez-Torres KL, Amarnani D, Kim LA, Mazzarino RC, Gordon H, Bocanegra Y, Villegas A, Gai X, Bootwalla M, Ji J, Shen L, Kosik KS, Su Y, Chen Y, Schultz A, Sperling RA, Johnson K, Reiman EM, Sepulveda-Falla D, Arboleda-Velasquez JF, Quiroz YT. Resilience to autosomal dominant Alzheimer's disease in a Reelin-COLBOS heterozygous man. Nat Med 2023; 29:1243-1252. [PMID: 37188781 PMCID: PMC10202812 DOI: 10.1038/s41591-023-02318-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/22/2023] [Indexed: 05/17/2023]
Abstract
We characterized the world's second case with ascertained extreme resilience to autosomal dominant Alzheimer's disease (ADAD). Side-by-side comparisons of this male case and the previously reported female case with ADAD homozygote for the APOE3 Christchurch (APOECh) variant allowed us to discern common features. The male remained cognitively intact until 67 years of age despite carrying a PSEN1-E280A mutation. Like the APOECh carrier, he had extremely elevated amyloid plaque burden and limited entorhinal Tau tangle burden. He did not carry the APOECh variant but was heterozygous for a rare variant in RELN (H3447R, termed COLBOS after the Colombia-Boston biomarker research study), a ligand that like apolipoprotein E binds to the VLDLr and APOEr2 receptors. RELN-COLBOS is a gain-of-function variant showing stronger ability to activate its canonical protein target Dab1 and reduce human Tau phosphorylation in a knockin mouse. A genetic variant in a case protected from ADAD suggests a role for RELN signaling in resilience to dementia.
Collapse
Affiliation(s)
- Francisco Lopera
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia
- Medicine School, University of Antioquia, Medellín, Colombia
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Anita S Chandrahas
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Michael O'Hare
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | | | - David Aguillon
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia
- Medicine School, University of Antioquia, Medellín, Colombia
| | - Ana Baena
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia
| | - Justin S Sanchez
- Department of Neurology at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Clara Vila-Castelar
- Department of Psychiatry at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Liliana Ramirez Gomez
- Department of Neurology at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Natalia Chmielewska
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Gabriel M Oliveira
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
- Department of Psychiatry at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Lisa Littau
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kyungeun Park
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorothee Schoemaker
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
- Department of Psychiatry at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Said Arevalo-Alquichire
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Kahira L Saez-Torres
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Leo A Kim
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Randall C Mazzarino
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Harper Gordon
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA
| | - Yamile Bocanegra
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia
| | - Andres Villegas
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia
- Medicine School, University of Antioquia, Medellín, Colombia
| | - Xiaowu Gai
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Moiz Bootwalla
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jianling Ji
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Lishuang Shen
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Kenneth S Kosik
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Yi Su
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Yinghua Chen
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Aaron Schultz
- Department of Neurology at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology at Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology at Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Eric M Reiman
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
- University of Arizona, Tucson, AZ, USA
- Arizona State University, Tucson, AZ, USA
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical School, Boston, MA, USA.
| | - Yakeel T Quiroz
- Neuroscience Group of Antioquia, Medicine School, University of Antioquia, Medellín, Colombia.
- Department of Neurology at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry at Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Di Donato N, Guerrini R, Billington CJ, Barkovich AJ, Dinkel P, Freri E, Heide M, Gershon ES, Gertler TS, Hopkin RJ, Jacob S, Keedy SK, Kooshavar D, Lockhart PJ, Lohmann DR, Mahmoud IG, Parrini E, Schrock E, Severi G, Timms AE, Webster RI, Willis MJH, Zaki MS, Gleeson JG, Leventer RJ, Dobyns WB. Monoallelic and biallelic mutations in RELN underlie a graded series of neurodevelopmental disorders. Brain 2022; 145:3274-3287. [PMID: 35769015 PMCID: PMC9989350 DOI: 10.1093/brain/awac164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/02/2022] [Accepted: 04/19/2022] [Indexed: 11/14/2022] Open
Abstract
Reelin, a large extracellular protein, plays several critical roles in brain development and function. It is encoded by RELN, first identified as the gene disrupted in the reeler mouse, a classic neurological mutant exhibiting ataxia, tremors and a 'reeling' gait. In humans, biallelic variants in RELN have been associated with a recessive lissencephaly variant with cerebellar hypoplasia, which matches well with the homozygous mouse mutant that has abnormal cortical structure, small hippocampi and severe cerebellar hypoplasia. Despite the large size of the gene, only 11 individuals with RELN-related lissencephaly with cerebellar hypoplasia from six families have previously been reported. Heterozygous carriers in these families were briefly reported as unaffected, although putative loss-of-function variants are practically absent in the population (probability of loss of function intolerance = 1). Here we present data on seven individuals from four families with biallelic and 13 individuals from seven families with monoallelic (heterozygous) variants of RELN and frontotemporal or temporal-predominant lissencephaly variant. Some individuals with monoallelic variants have moderate frontotemporal lissencephaly, but with normal cerebellar structure and intellectual disability with severe behavioural dysfunction. However, one adult had abnormal MRI with normal intelligence and neurological profile. Thorough literature analysis supports a causal role for monoallelic RELN variants in four seemingly distinct phenotypes including frontotemporal lissencephaly, epilepsy, autism and probably schizophrenia. Notably, we observed a significantly higher proportion of loss-of-function variants in the biallelic compared to the monoallelic cohort, where the variant spectrum included missense and splice-site variants. We assessed the impact of two canonical splice-site variants observed as biallelic or monoallelic variants in individuals with moderately affected or normal cerebellum and demonstrated exon skipping causing in-frame loss of 46 or 52 amino acids in the central RELN domain. Previously reported functional studies demonstrated severe reduction in overall RELN secretion caused by heterozygous missense variants p.Cys539Arg and p.Arg3207Cys associated with lissencephaly suggesting a dominant-negative effect. We conclude that biallelic variants resulting in complete absence of RELN expression are associated with a consistent and severe phenotype that includes cerebellar hypoplasia. However, reduced expression of RELN remains sufficient to maintain nearly normal cerebellar structure. Monoallelic variants are associated with incomplete penetrance and variable expressivity even within the same family and may have dominant-negative effects. Reduced RELN secretion in heterozygous individuals affects only cortical structure whereas the cerebellum remains intact. Our data expand the spectrum of RELN-related neurodevelopmental disorders ranging from lethal brain malformations to adult phenotypes with normal brain imaging.
Collapse
Affiliation(s)
- Nataliya Di Donato
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, University of Florence, 50139 Florence, Italy
| | - Charles J Billington
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN 55454, USA
| | - A James Barkovich
- Departments of Radiology and Biomedical Imaging, Neurology, Pediatrics, and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Philine Dinkel
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
- German Primate Center, Leibniz Institute for Primate Research, 37077 Goettingen, Germany
| | - Elliot S Gershon
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Tracy S Gertler
- Division of Neurology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Robert J Hopkin
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Department of Pediatrics, Division of Human Genetics, Cincinnati, OH 45229, USA
| | - Suma Jacob
- Department of Psychiatry, University of Minnesota, Minneapolis, MN 55454, USA
| | - Sarah K Keedy
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Daniz Kooshavar
- Bruce Lefory Centre, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - Paul J Lockhart
- Bruce Lefory Centre, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - Dietmar R Lohmann
- Institut fur Humangenetik, Universitatsklinikum Essen, 45147 Essen, Germany
| | - Iman G Mahmoud
- Pediatric Neurology Department, Cairo University Children's Hospital, Cairo, Egypt
| | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, University of Florence, 50139 Florence, Italy
| | - Evelin Schrock
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Giulia Severi
- Medical Genetics Unit, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Andrew E Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Richard I Webster
- T. Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Sydney 2145, Australia
| | - Mary J H Willis
- Uniformed Services University School of Medicine and Naval Medical Center, Department of Pediatrics, San Diego, CA 92134, USA
| | - Maha S Zaki
- Pediatric Neurology Department, Cairo University Children's Hospital, Cairo, Egypt
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo Governorate 12622, Egypt
| | - Joseph G Gleeson
- Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Richard J Leventer
- Department of Neurology, Royal Children's Hospital, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - William B Dobyns
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN 55454, USA
| |
Collapse
|
4
|
Chaves Filho AJM, Mottin M, Lós DB, Andrade CH, Macedo DS. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022; 201:79-99. [PMID: 35931337 DOI: 10.1016/j.biochi.2022.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/26/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Inflammation and an exacerbated immune response are widely accepted contributing mechanisms to the genesis and progression of major neuropsychiatric disorders. However, despite the impressive advances in understanding the neurobiology of these disorders, there is still no approved drug directly linked to the regulation of inflammation or brain immune responses. Importantly, matrix metalloproteinases (MMPs) comprise a group of structurally related endopeptidases primarily involved in remodeling extracellular matrix (ECM). In the central nervous system (CNS), these proteases control synaptic plasticity and strength, patency of the blood-brain barrier, and glia-neuron interactions through cleaved and non-cleaved mediators. Several pieces of evidence have pointed to a complex scenario of MMPs dysregulation triggered by neuroinflammation. Furthermore, major psychiatric disorders' affective symptoms and neurocognitive abnormalities are related to MMPs-mediated ECM changes and neuroglia activation. In the past decade, research efforts have been directed to broad-spectrum MMPs inhibitors with frustrating clinical results. However, in the light of recent advances in combinatorial chemistry and drug design technologies, specific and CNS-oriented MMPs modulators have been proposed as a new frontier of therapy for regulating ECM properties in the CNS. Therefore, here we aim to discuss the state of the art of MMPs and ECM abnormalities in major neuropsychiatric disorders, namely depression, bipolar disorder, and schizophrenia, the possible neuro-immune interactions involved in this complex scenario of MMPs dysregulation and propose these endopeptidases as promising targets for rational drug design.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Deniele Bezerra Lós
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
5
|
Priyadarshi S, Hansdah K, Singh N, Bouzid A, Ray CS, Panda KC, Biswal NC, Desai A, Choudhury JC, Tekari A, Masmoudi S, Ramchander PV. The risks of RELN polymorphisms and its expression in the development of otosclerosis. PLoS One 2022; 17:e0269558. [PMID: 35658052 PMCID: PMC9165908 DOI: 10.1371/journal.pone.0269558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
Otosclerosis (OTSC) is the primary form of conductive hearing loss characterized by abnormal bone remodelling within the otic capsule of the human middle ear. A genetic association of the RELN SNP rs3914132 with OTSC has been identified in European population. Previously, we showed a trend towards association of this polymorphism with OTSC and identified a rare variant rs74503667 in a familial case. Here, we genotyped these variants in an Indian cohort composed of 254 OTSC cases and 262 controls. We detected a significant association of rs3914132 with OTSC (OR = 0.569, 95%CI = 0.386–0.838, p = 0.0041). To confirm this finding, we completed a meta-analysis which revealed a significant association of the rs3914132 polymorphism with OTSC (Z = 6.707, p<0.0001) across different ethnic populations. Linkage analysis found the evidence of linkage at RELN locus (LOD score 2.1059) in the OTSC family which has shown the transmission of rare variant rs74503667 in the affected individuals. To understand the role of RELN and its receptors in the development of OTSC, we went further to perform a functional analysis of RELN/reelin. Here we detected a reduced RELN (p = 0.0068) and VLDLR (p = 0.0348) mRNA levels in the otosclerotic stapes tissues. Furthermore, a reduced reelin protein expression by immunohistochemistry was confirmed in the otosclerotic tissues. Electrophoretic mobility shift assays for rs3914132 and rs74503667 variants revealed an altered binding of transcription factors in the mutated sequences which indicates the regulatory role of these variations in the RELN gene regulation. Subsequently, we showed by scanning electron microscopy a change in stapes bone morphology of otosclerotic patients. In conclusion, this study evidenced that the rare variation rs74503667 and the common polymorphism rs3914132 in the RELN gene and its reduced expressions that were associated with OTSC.
Collapse
Affiliation(s)
- Saurabh Priyadarshi
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, India
| | - Kirtal Hansdah
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, India
| | - Neha Singh
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, India
| | - Amal Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Chinmay Sundar Ray
- Department of Ear, Nose, and Throat (ENT), Shrirama Chandra Bhanj (SCB) Medical College & Hospital, Cuttack, India
| | | | - Narayan Chandra Biswal
- Department of Ear, Nose, and Throat (ENT), Shrirama Chandra Bhanj (SCB) Medical College & Hospital, Cuttack, India
| | - Ashim Desai
- Dr. ABR Desai Ear, Nose and Throat (ENT) Clinic and Research Centre, Mumbai, India
| | - Jyotish Chandra Choudhury
- Department of Forensic Medicine & Toxicology (FMT), Shrirama Chandra Bhanj (SCB) Medical College & Hospital, Cuttack, India
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | | |
Collapse
|
6
|
Castello MA, Gleeson JG. Insight into developmental mechanisms of global and focal migration disorders of cortical development. Curr Opin Neurobiol 2020; 66:77-84. [PMID: 33099181 DOI: 10.1016/j.conb.2020.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
Abstract
Cortical development involves neurogenesis followed by migration, maturation, and myelination of immature neurons. Disruptions in these processes can cause malformations of cortical development (MCD). Radial glia (RG) are the stem cells of the brain, both generating neurons and providing the scaffold upon which immature neurons radially migrate. Germline mutations in genes required for cell migration, or cell-cell contact, often lead to global MCDs. Somatic mutations in RG in genes involved in homeostatic function, like mTOR signaling, often lead to focal MCDs. Two different mutations occurring in the same patient can combine in ways we are just beginning to understand. Our growing knowledge about MCD suggests mTOR inhibitors may have expanded utility in treatment-resistant epilepsy, while imaging techniques can better delineate the type and extent of these lesions.
Collapse
Affiliation(s)
- Michael A Castello
- Department of Neurosciences, Division of Child Neurology, University of California San Diego, San Diego, CA, USA
| | - Joseph G Gleeson
- Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
7
|
Zhang Z, Chen G. A logical relationship for schizophrenia, bipolar, and major depressive disorder. Part 1: Evidence from chromosome 1 high density association screen. J Comp Neurol 2020; 528:2620-2635. [PMID: 32266715 DOI: 10.1002/cne.24921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
Familial clustering of schizophrenia (SCZ), bipolar disorder (BPD), and major depressive disorder (MDD) was investigated systematically (Aukes et al., Genetics in Medicine, 2012, 14, 338-341) and any two or even three of these disorders could coexist in some families. Furthermore, evidence from symptomatology and psychopharmacology also imply the existence of intrinsic connections between these three major psychiatric disorders. A total of 71,445 SNPs on chromosome 1 were genotyped on 119 SCZ, 253 BPD (type-I), 177 MDD cases and 1000 controls and further validated in 986 SCZ patients in the population of Shandong province of China. Outstanding psychosis genes are systematically revealed( ATP1A4, ELTD1, FAM5C, HHAT, KIF26B, LMX1A, NEGR1, NFIA, NR5A2, NTNG1, PAPPA2, PDE4B, PEX14, RYR2, SYT6, TGFBR3, TTLL7, and USH2A). Unexpectedly, flanking genes for up to 97.09% of the associated SNPs were also replicated in an enlarged cohort of 986 SCZ patients. From the perspective of etiological rather than clinical psychiatry, bipolar, and major depressive disorder could be subtypes of schizophrenia. Meanwhile, the varied clinical feature and prognosis might be the result of interaction of genetics and epigenetics, for example, irreversible or reversible shut down, and over or insufficient expression of certain genes, which may gives other aspects of these severe mental disorders.
Collapse
Affiliation(s)
- Zhihua Zhang
- Shandong Mental Health Center, Jinan, Shandong, China
| | - Gang Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
8
|
Notaras MJ, Vivian B, Wilson C, van den Buuse M. Interaction of reelin and stress on immobility in the forced swim test but not dopamine-mediated locomotor hyperactivity or prepulse inhibition disruption: Relevance to psychotic and mood disorders. Schizophr Res 2020; 215:485-492. [PMID: 28711473 DOI: 10.1016/j.schres.2017.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/28/2022]
Abstract
RATIONALE Psychotic disorders, such as schizophrenia, as well as some mood disorders, such as bipolar disorder, have been suggested to share common biological risk factors. One such factor is reelin, a large extracellular matrix glycoprotein that regulates neuronal migration during development as well as numerous activity-dependent processes in the adult brain. The current study sought to evaluate whether a history of stress exposure interacts with endogenous reelin levels to modify behavioural endophenotypes of relevance to psychotic and mood disorders. METHODS Heterozygous Reeler Mice (HRM) and wildtype (WT) controls were treated with 50mg/L of corticosterone (CORT) in their drinking water from 6 to 9weeks of age, before undergoing behavioural testing in adulthood. We assessed methamphetamine-induced locomotor hyperactivity, prepulse inhibition (PPI) of acoustic startle, short-term spatial memory in the Y-maze, and depression-like behaviour in the Forced-Swim Test (FST). RESULTS HRM genotype or CORT treatment did not affect methamphetamine-induced locomotor hyperactivity, a model of psychosis-like behaviour. At baseline, HRM showed decreased PPI at the commonly used 100msec interstimulus interval (ISI), but not at the 30msec ISI or following challenge with apomorphine. A history of CORT exposure potentiated immobility in the FST amongst HRM, but not WT mice. In the Y-maze, chronic CORT treatment decreased novel arm preference amongst HRM, reflecting reduced short-term spatial memory. CONCLUSION These data confirm a significant role of endogenous reelin levels on stress-related behaviour, supporting a possible role in both bipolar disorder and schizophrenia. However, an interaction of reelin deficiency with dopaminergic regulation of psychosis-like behaviour remains unclear.
Collapse
Affiliation(s)
- Michael J Notaras
- Florey Institute of Neuroscience & Mental Health, Melbourne, Victoria, Australia
| | - Billie Vivian
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Carey Wilson
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Pharmacology, University of Melbourne, Victoria, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia.
| |
Collapse
|
9
|
Subarachnoid cerebrospinal fluid is essential for normal development of the cerebral cortex. Semin Cell Dev Biol 2019; 102:28-39. [PMID: 31786096 DOI: 10.1016/j.semcdb.2019.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
The central nervous system develops around a fluid filled space which persists in the adult within the ventricles, spinal canal and around the outside of the brain and spinal cord. Ventricular fluid is known to act as a growth medium and stimulator of proliferation and differentiation to neural stem cells but the role of CSF in the subarachnoid space has not been fully investigated except for its role in the recently described "glymphatic" system. Fundamental changes occur in the control and coordination of CNS development upon completion of brain stem and spinal cord development and initiation of cortical development. These include changes in gene expression, changes in fluid and fluid source from neural tube fluid to cerebrospinal fluid (CSF), changes in fluid volume, composition and fluid flow pathway, with exit of high volume CSF into the subarachnoid space and the critical need for fluid drainage. We used a number of experimental approaches to test a predicted critical role for CSF in development of the cerebral cortex in rodents and humans. Data from fetuses affected by spina bifida and/or hydrocephalus are correlated with experimental evidence on proliferation and migration of cortical cells from the germinal epithelium in rodent neural tube defects, as well as embryonic brain slice experiments demonstrating a requirement for CSF to contact both ventricular and pial surfaces of the developing cortex for normal proliferation and migration. We discuss the possibility that complications with the fluid system are likely to underlie developmental disorders affecting the cerebral cortex as well as function and integrity of the cortex throughout life.
Collapse
|
10
|
Abstract
Mental disorders are important diseases with a high prevalence rate in the general population. Common mental disorders are complex diseases with high heritability, and their pathogenesis is the result of interactions between genetic and environmental factors. However, the relationship between mental disorders and genes is complex and difficult to evaluate. Additionally, some mental disorders involve numerous genes, and a single gene can also be associated with different types of mental disorders.This study used text mining (including word frequency analysis, cluster analysis, and association analysis) of the PubMed database to identify genes related to mental disorders.Word frequency analysis revealed 52 high-frequency genes important in studies of mental disorders. Cluster analysis showed that 5-HTT, SLC6A4, and MAOA are common genetic factors in most mental disorders; the intra-group genes in each cluster were highly correlated. Some mental disorders may have common genetic factors; for example, there may be common genetic factors between 'Affective Disorders' and 'Schizophrenia.' Association analysis revealed 35 frequent itemsets and 25 association rules, indicating close associations among genes. The results of association rules showed that CCK, MAOA, and 5-HTT are the most closely related.We used text mining technology to analyze genes related to mental disorders to further summarize and clarify the relationships between mental disorders and genes as well as identify potential relationships, providing a foundation for future experiments. The results of the associative analysis also provide a reference for multi-gene studies of mental disorders.
Collapse
Affiliation(s)
- Ying Wu
- School of Humanities and Social Sciences, Shanxi Medical University,
| | - Meilin Dang
- School of Humanities and Social Sciences, Shanxi Medical University,
| | - Hongxia Li
- School of Humanities and Social Sciences, Shanxi Medical University,
| | - Xing Jin
- Affiliated Tumor Hospital, Shanxi Medical University, Taiyuan, China
| | - Wenxiao Yang
- School of Humanities and Social Sciences, Shanxi Medical University,
| |
Collapse
|
11
|
Prata DP, Costa-Neves B, Cosme G, Vassos E. Unravelling the genetic basis of schizophrenia and bipolar disorder with GWAS: A systematic review. J Psychiatr Res 2019; 114:178-207. [PMID: 31096178 DOI: 10.1016/j.jpsychires.2019.04.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES To systematically review findings of GWAS in schizophrenia (SZ) and in bipolar disorder (BD); and to interpret findings, with a focus on identifying independent replications. METHOD PubMed search, selection and review of all independent GWAS in SZ or BD, published since March 2011, i.e. studies using non-overlapping samples within each article, between articles, and with those of the previous review (Li et al., 2012). RESULTS From the 22 GWAS included in this review, the genetic associations surviving standard GWAS-significance were for genetic markers in the regions of ACSL3/KCNE4, ADCY2, AMBRA1, ANK3, BRP44, DTL, FBLN1, HHAT, INTS7, LOC392301, LOC645434/NMBR, LOC729457, LRRFIP1, LSM1, MDM1, MHC, MIR2113/POU3F2, NDST3, NKAPL, ODZ4, PGBD1, RENBP, TRANK1, TSPAN18, TWIST2, UGT1A1/HJURP, WHSC1L1/FGFR1 and ZKSCAN4. All genes implicated across both reviews are discussed in terms of their function and implication in neuropsychiatry. CONCLUSION Taking all GWAS to date into account, AMBRA1, ANK3, ARNTL, CDH13, EFHD1 (albeit with different alleles), MHC, PLXNA2 and UGT1A1 have been implicated in either disorder in at least two reportedly non-overlapping samples. Additionally, evidence for a SZ/BD common genetic basis is most strongly supported by the implication of ANK3, NDST3, and PLXNA2.
Collapse
Affiliation(s)
- Diana P Prata
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Portugal; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, UK; Instituto Universitário de Lisboa (ISCTE-IUL), Centro de Investigação e Intervenção Social, Lisboa, Portugal.
| | - Bernardo Costa-Neves
- Lisbon Medical School, University of Lisbon, Av. Professor Egas Moniz, 1649-028, Lisbon, Portugal; Centro Hospitalar Psiquiátrico de Lisboa, Av. do Brasil, 53 1749-002, Lisbon, Portugal
| | - Gonçalo Cosme
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Portugal
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, 16 De Crespigny Park, SE5 8AF, UK
| |
Collapse
|
12
|
Zhang-James Y, Fernàndez-Castillo N, Hess JL, Malki K, Glatt SJ, Cormand B, Faraone SV. An integrated analysis of genes and functional pathways for aggression in human and rodent models. Mol Psychiatry 2019; 24:1655-1667. [PMID: 29858598 PMCID: PMC6274606 DOI: 10.1038/s41380-018-0068-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/04/2018] [Accepted: 04/03/2018] [Indexed: 11/12/2022]
Abstract
Human genome-wide association studies (GWAS), transcriptome analyses of animal models, and candidate gene studies have advanced our understanding of the genetic architecture of aggressive behaviors. However, each of these methods presents unique limitations. To generate a more confident and comprehensive view of the complex genetics underlying aggression, we undertook an integrated, cross-species approach. We focused on human and rodent models to derive eight gene lists from three main categories of genetic evidence: two sets of genes identified in GWAS studies, four sets implicated by transcriptome-wide studies of rodent models, and two sets of genes with causal evidence from online Mendelian inheritance in man (OMIM) and knockout (KO) mice reports. These gene sets were evaluated for overlap and pathway enrichment to extract their similarities and differences. We identified enriched common pathways such as the G-protein coupled receptor (GPCR) signaling pathway, axon guidance, reelin signaling in neurons, and ERK/MAPK signaling. Also, individual genes were ranked based on their cumulative weights to quantify their importance as risk factors for aggressive behavior, which resulted in 40 top-ranked and highly interconnected genes. The results of our cross-species and integrated approach provide insights into the genetic etiology of aggression.
Collapse
Affiliation(s)
- Yanli Zhang-James
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY, USA.
| | - Noèlia Fernàndez-Castillo
- 0000 0004 1937 0247grid.5841.8Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain ,0000 0004 1791 1185grid.452372.5Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain ,0000 0004 1937 0247grid.5841.8Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain ,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Spain
| | - Jonathan L Hess
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA
| | - Karim Malki
- 0000 0001 2322 6764grid.13097.3cKing’s College London, MRC Social, Genetic and Developmental Psychiatry Centre at the Institute of Psychiatry, Psychology and Neuroscience (IOPPN), London, UK
| | - Stephen J Glatt
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0000 9159 4457grid.411023.5Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, NY USA
| | - Bru Cormand
- 0000 0004 1937 0247grid.5841.8Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain ,0000 0004 1791 1185grid.452372.5Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain ,0000 0004 1937 0247grid.5841.8Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain ,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Spain
| | - Stephen V Faraone
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0000 9159 4457grid.411023.5Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0004 1936 7443grid.7914.bK.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
Alfimova MV, Kondratyev NV, Golov AK, Golubev SA, Galaktionova DY, Nasedkina TV, Golimbet VE. [Relationships of rs7341475 polymorphism and DNA methylation in the reelin gene with schizophrenia symptoms]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:71-76. [PMID: 30335076 DOI: 10.17116/jnevro201811809171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To study the role of polymorphism rs7341475 and methylation of the reelin gene in symptoms of schizophrenia and semantic verbal fluency. MATERIAL AND METHODS Genotypes at the locus rs7341475 were identified in 556 patients with schizophrenic disorders. PANSS scores were obtained for 549 patients and 221 patients performed a test for semantic verbal fluency. The association of the reelin promoter methylation with the PANSS and verbal fluency measures was evaluated in 35 patients. A five-factor model of the PANSS was used. RESULTS The interaction effect of sex with genotype on the PANSS scores was found (F=2.70, p=0.020). Schizophrenic men homozygous for a common allele G had the lowest scores of the positive syndrome. Verbal fluency was related to the reelin promoter methylation. CONCLUSION The results suggest that polymorphism rs7341475 may be associated with the variability of positive symptomatology in schizophrenic men. At the same time, the reelin gene methylation pattern, which consists of a higher methylation level in the region of the transcription start site and a lower one in the distal region of the promoter, may be beneficial for verbal fluency.
Collapse
Affiliation(s)
| | | | - A K Golov
- Mental Health Research Center, Moscow, Russia
| | - S A Golubev
- Mental Health Research Center, Moscow, Russia
| | - D Yu Galaktionova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - T V Nasedkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
14
|
Kara B, Sahin N, Kara M, Sakalli Cetin E, Topal H. The relationship between attention deficit hyperactivity disorder and reelin gene polymorphisms in Turkish population. PSYCHIAT CLIN PSYCH 2018. [DOI: 10.1080/24750573.2018.1478192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Affiliation(s)
- Bilge Kara
- Faculty of Medicine, Department of Psychiatry, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nilfer Sahin
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Mugla Sitki Kocman University, Mugla, Turkey
| | - Murat Kara
- Faculty of Medicine, Department of Medical Genetics, Mugla Sitki Kocman University, Mugla, Turkey
| | - Esin Sakalli Cetin
- Faculty of Medicine, Department of Medical Biology, Mugla Sitki Kocman University, Mugla, Turkey
| | - Hatice Topal
- Faculty of Medicine, Department of Pediatrics, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
15
|
Imai H, Shoji H, Ogata M, Kagawa Y, Owada Y, Miyakawa T, Sakimura K, Terashima T, Katsuyama Y. Dorsal Forebrain-Specific Deficiency of Reelin-Dab1 Signal Causes Behavioral Abnormalities Related to Psychiatric Disorders. Cereb Cortex 2018; 27:3485-3501. [PMID: 26762856 DOI: 10.1093/cercor/bhv334] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Reelin-Dab1 signaling is involved in brain development and neuronal functions. The abnormalities in the signaling through either reduction of Reelin and Dab1 gene expressions or the genomic mutations in the brain have been reported to be associated with psychiatric disorders. However, it has not been clear if the deficiency in Reelin-Dab1 signaling is responsible for symptoms of the disorders. Here, to examine the function of Reelin-Dab1 signaling in the forebrain, we generated dorsal forebrain-specific Dab1 conditional knockout mouse (Dab1 cKO) and performed a behavioral test battery on the Dab1 cKO mice. Although conventional Dab1 null mutant mice exhibit cerebellar atrophy and cerebellar ataxia, the Dab1 cKO mice had normal cerebellum and showed no motor dysfunction. Dab1 cKO mice exhibited behavioral abnormalities, including hyperactivity, decreased anxiety-like behavior, and impairment of working memory, which are reminiscent of symptoms observed in patients with psychiatric disorders such as schizophrenia and bipolar disorder. These results suggest that deficiency of Reelin-Dab1 signal in the dorsal forebrain is involved in the pathogenesis of some symptoms of human psychiatric disorders.
Collapse
Affiliation(s)
- Hideaki Imai
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Kawaguchi 332-0012, Japan
| | - Masaki Ogata
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Kawaguchi 332-0012, Japan.,Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Toshio Terashima
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Yu Katsuyama
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan.,Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
16
|
Han L, Jia Z, Cao C, Liu Z, Liu F, Wang L, Ren W, Sun M, Wang B, Li C, Chen L. Potential contribution of the neurodegenerative disorders risk loci to cognitive performance in an elderly male gout population. Medicine (Baltimore) 2017; 96:e8195. [PMID: 28953682 PMCID: PMC5626325 DOI: 10.1097/md.0000000000008195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cognitive impairment has been described in elderly subjects with high normal concentrations of serum uric acid. However, it remains unclear if gout confers an increased poorer cognition than those in individuals with asymptomatic hyperuricemia. The present study aimed at evaluating cognitive function in patients suffering from gout in an elderly male population, and further investigating the genetic contributions to the risk of cognitive function.This study examined the cognitive function as assessed by Mini-Mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA) in 205 male gout patients and 204 controls. The genetic basis of these cognitive measures was evaluated by genome-wide association study (GWAS) data in 102 male gout patients. Furthermore, 7 loci associated with cognition in GWAS were studied for correlation with gout in 1179 male gout patients and 1848 healthy male controls.Compared with controls, gout patients had significantly lower MoCA scores [22.78 ± 3.01 vs 23.42 ± 2.95, P = .023, adjusted by age, body mass index (BMI), education, and emotional disorder]. GWAS revealed 7 single-nucleotide polymorphisms (SNPs) associations with MoCA test at a level of conventional genome-wide significance (P < 9.6 × 10). The most significant association was observed between rs12895072 and rs12434554 within the KTN1 gene (Padjusted = 4.2 × 10, Padjusted = 4.7 × 10) at 14q22. The next best signal was in RELN gene (rs155333, Padjusted = 1.3 × 10) at 7q22, while the other variants at rs17458357 (Padjusted = 3.98 × 10), rs2572683 (Padjusted = 8.9 × 10), rs12555895 (Padjusted = 2.6 × 10), and rs3764030 (Padjusted = 9.4 × 10) were also statistically significant. The 7 SNPs were not associated with gout in further analysis (all P > .05).Elderly male subjects with gout exhibit accelerated decline in cognition performance. Several neurodegenerative disorders risk loci were identified for genetic contributors to cognitive performance in our Chinese elderly male gout population. Larger prospective studies of the cognitive performance and genetic analysis in gout subjects are recommended.
Collapse
Affiliation(s)
- Lin Han
- Department of Endocrinology Qilu Hospital of Shandong University, Jinan
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Zhaotong Jia
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhen Liu
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Fuqiang Liu
- Department of Endocrinology Qilu Hospital of Shandong University, Jinan
| | - Lin Wang
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Wei Ren
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Mingxia Sun
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Baoping Wang
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Changgui Li
- Gout Laboratory, The Affiliated Hospital of Qingdao University, Qingdao
| | - Li Chen
- Department of Endocrinology Qilu Hospital of Shandong University, Jinan
| |
Collapse
|
17
|
Chen N, Bao Y, Xue Y, Sun Y, Hu D, Meng S, Lu L, Shi J. Meta-analyses of RELN variants in neuropsychiatric disorders. Behav Brain Res 2017; 332:110-119. [PMID: 28506622 DOI: 10.1016/j.bbr.2017.05.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 10/19/2022]
Abstract
Reelin is a critical extracellular matrix glycoprotein and implicated in neurodevelopment and psychiatric disorders in animal model studies. The genetic polymorphism of RELN has also been reported to be associated with several psychiatric disorders, but the results remain controversial. Here, we conducted meta-analyses of RELN gene SNPs and related neuropsychiatric disorders (schizophrenia, autistic spectrum disorders, attention-deficit hyperactivity disorder, Alzheimer's disease and bipolar disorders). A total of 12 SNPs (rs736707, rs362691, rs607755, rs2229864, rs7341475, rs262355, rs362719, rs11496125, g.-888G>C, rs2299356, rs528528, and rs4298437) in RELN gene were included into meta-analyses. Subgroup analyses based on ethnicity were performed. We found that RELN rs736707 was significantly related with psychiatric disorders (schizophrenia, autism spectrum disorders and attention-deficit hyperactivity disorder) in Asian group (C vs T, OR=1.26, 95% CI=1.13-1.41, P<0.01, FDR<0.01), and rs7341475 was only significantly associated with reduced risk of schizophrenia in Caucasian (A vs G, OR=0.88, 95% CI=0.82-0.95, P<0.01, FDR<0.01). No association of other SNPs and psychiatric disorders is found. These findings suggest a role of RELN SNPs in psychiatric diseases, and indicate that further researches in populations with different genetic background and studies with larger sample size are of great value.
Collapse
Affiliation(s)
- Na Chen
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China
| | - Yanping Bao
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China; Beijing Key Laboratory on Drug Dependence Research, Beijing, China
| | - Yan Sun
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China; Beijing Key Laboratory on Drug Dependence Research, Beijing, China
| | - Die Hu
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China; Beijing Key Laboratory on Drug Dependence Research, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China; Beijing Key Laboratory on Drug Dependence Research, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China; Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China; School of Basic Medical Science, Peking University, Beijing, China; Beijing Key Laboratory on Drug Dependence Research, Beijing, China; The State Key Laboratory of Natural and Biomimetic Drugs, Beijing, China; Key Laboratory for Neuroscience of the Ministry of Education and Ministry of Public Healthy, Beijing, China.
| |
Collapse
|
18
|
Whittaker DE, Riegman KL, Kasah S, Mohan C, Yu T, Sala BP, Hebaishi H, Caruso A, Marques AC, Michetti C, Smachetti MES, Shah A, Sabbioni M, Kulhanci O, Tee WW, Reinberg D, Scattoni ML, Volk H, McGonnell I, Wardle FC, Fernandes C, Basson MA. The chromatin remodeling factor CHD7 controls cerebellar development by regulating reelin expression. J Clin Invest 2017; 127:874-887. [PMID: 28165338 PMCID: PMC5330721 DOI: 10.1172/jci83408] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
The mechanisms underlying the neurodevelopmental deficits associated with CHARGE syndrome, which include cerebellar hypoplasia, developmental delay, coordination problems, and autistic features, have not been identified. CHARGE syndrome has been associated with mutations in the gene encoding the ATP-dependent chromatin remodeler CHD7. CHD7 is expressed in neural stem and progenitor cells, but its role in neurogenesis during brain development remains unknown. Here we have shown that deletion of Chd7 from cerebellar granule cell progenitors (GCps) results in reduced GCp proliferation, cerebellar hypoplasia, developmental delay, and motor deficits in mice. Genome-wide expression profiling revealed downregulated expression of the gene encoding the glycoprotein reelin (Reln) in Chd7-deficient GCps. Recessive RELN mutations have been associated with severe cerebellar hypoplasia in humans. We found molecular and genetic evidence that reductions in Reln expression contribute to GCp proliferative defects and cerebellar hypoplasia in GCp-specific Chd7 mouse mutants. Finally, we showed that CHD7 is necessary for maintaining an open, accessible chromatin state at the Reln locus. Taken together, this study shows that Reln gene expression is regulated by chromatin remodeling, identifies CHD7 as a previously unrecognized upstream regulator of Reln, and provides direct in vivo evidence that a mammalian CHD protein can control brain development by modulating chromatin accessibility in neuronal progenitors.
Collapse
Affiliation(s)
- Danielle E. Whittaker
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Kimberley L.H. Riegman
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Sahrunizam Kasah
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Conor Mohan
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Tian Yu
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Blanca Pijuan Sala
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Husam Hebaishi
- King’s College London, Randall Division, New Hunt’s House, London, United Kingdom
| | - Angela Caruso
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
- School of Behavioural Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Ana Claudia Marques
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Caterina Michetti
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy
| | | | - Apar Shah
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Mara Sabbioni
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
| | - Omer Kulhanci
- MRC Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Wee-Wei Tee
- Howard Hughes Medical Institute, Department of Molecular Pharmacology and Biochemistry, New York University School of Medicine, New York, New York, USA
| | - Danny Reinberg
- Howard Hughes Medical Institute, Department of Molecular Pharmacology and Biochemistry, New York University School of Medicine, New York, New York, USA
| | - Maria Luisa Scattoni
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
| | - Holger Volk
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Imelda McGonnell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Fiona C. Wardle
- King’s College London, Randall Division, New Hunt’s House, London, United Kingdom
| | - Cathy Fernandes
- MRC Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- King’s College London, MRC Centre for Neurodevelopmental Disorders, New Hunt’s House, London, United Kingdom
| | - M. Albert Basson
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
- King’s College London, MRC Centre for Neurodevelopmental Disorders, New Hunt’s House, London, United Kingdom
| |
Collapse
|
19
|
Reis VNDS, Kitajima JP, Tahira AC, Feio-dos-Santos AC, Fock RA, Lisboa BCG, Simões SN, Krepischi ACV, Rosenberg C, Lourenço NC, Passos-Bueno MR, Brentani H. Integrative Variation Analysis Reveals that a Complex Genotype May Specify Phenotype in Siblings with Syndromic Autism Spectrum Disorder. PLoS One 2017; 12:e0170386. [PMID: 28118382 PMCID: PMC5261619 DOI: 10.1371/journal.pone.0170386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/31/2016] [Indexed: 12/30/2022] Open
Abstract
It has been proposed that copy number variations (CNVs) are associated with increased risk of autism spectrum disorder (ASD) and, in conjunction with other genetic changes, contribute to the heterogeneity of ASD phenotypes. Array comparative genomic hybridization (aCGH) and exome sequencing, together with systems genetics and network analyses, are being used as tools for the study of complex disorders of unknown etiology, especially those characterized by significant genetic and phenotypic heterogeneity. Therefore, to characterize the complex genotype-phenotype relationship, we performed aCGH and sequenced the exomes of two affected siblings with ASD symptoms, dysmorphic features, and intellectual disability, searching for de novo CNVs, as well as for de novo and rare inherited point variations—single nucleotide variants (SNVs) or small insertions and deletions (indels)—with probable functional impacts. With aCGH, we identified, in both siblings, a duplication in the 4p16.3 region and a deletion at 8p23.3, inherited by a paternal balanced translocation, t(4, 8) (p16; p23). Exome variant analysis found a total of 316 variants, of which 102 were shared by both siblings, 128 were in the male sibling exome data, and 86 were in the female exome data. Our integrative network analysis showed that the siblings’ shared translocation could explain their similar syndromic phenotype, including overgrowth, macrocephaly, and intellectual disability. However, exome data aggregate genes to those already connected from their translocation, which are important to the robustness of the network and contribute to the understanding of the broader spectrum of psychiatric symptoms. This study shows the importance of using an integrative approach to explore genotype-phenotype variability.
Collapse
MESH Headings
- Autism Spectrum Disorder/genetics
- Child
- Chromosomes, Human, Pair 4/genetics
- Chromosomes, Human, Pair 4/ultrastructure
- Chromosomes, Human, Pair 8/genetics
- Chromosomes, Human, Pair 8/ultrastructure
- Comparative Genomic Hybridization
- DNA Copy Number Variations
- Exome/genetics
- Female
- Gene Duplication
- Gene Regulatory Networks
- Genetic Association Studies
- Humans
- In Situ Hybridization, Fluorescence
- Intellectual Disability/genetics
- Learning Disabilities/genetics
- Male
- Megalencephaly/genetics
- Nerve Tissue Proteins/genetics
- Nucleic Acid Amplification Techniques
- Sequence Deletion
- Siblings
- Syndrome
- Translocation, Genetic
Collapse
Affiliation(s)
| | | | - Ana Carolina Tahira
- LIM23-Institute of Psychiatry, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Rodrigo Ambrósio Fock
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
| | | | - Sérgio Nery Simões
- Department of Informatics, Federal Institute of Espírito Santo, Serra, Brazil
| | - Ana C. V. Krepischi
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, São Paulo, Brazil
| | - Carla Rosenberg
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, São Paulo, Brazil
| | - Naila Cristina Lourenço
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, São Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, São Paulo, Brazil
| | - Helena Brentani
- LIM23-Institute of Psychiatry, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
20
|
Monson ET, Pirooznia M, Parla J, Kramer M, Goes FS, Gaine ME, Gaynor SC, de Klerk K, Jancic D, Karchin R, McCombie WR, Zandi PP, Potash JB, Willour VL. Assessment of Whole-Exome Sequence Data in Attempted Suicide within a Bipolar Disorder Cohort. MOLECULAR NEUROPSYCHIATRY 2017; 3:1-11. [PMID: 28879196 DOI: 10.1159/000454773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/24/2016] [Indexed: 11/19/2022]
Abstract
Suicidal behavior is a complex and devastating phenotype with a heritable component that has not been fully explained by existing common genetic variant analyses. This study represents the first large-scale DNA sequencing project designed to assess the role of rare functional genetic variation in suicidal behavior risk. To accomplish this, whole-exome sequencing data for ∼19,000 genes were generated for 387 bipolar disorder subjects with a history of suicide attempt and 631 bipolar disorder subjects with no prior suicide attempts. Rare functional variants were assessed in all exome genes as well as pathways hypothesized to contribute to suicidal behavior risk. No result survived conservative Bonferroni correction, though many suggestive findings have arisen that merit additional attention. In addition, nominal support for past associations in genes, such as BDNF, and pathways, such as the hypothalamic-pituitary-adrenal axis, was also observed. Finally, a novel pathway was identified that is driven by aldehyde dehydrogenase genes. Ultimately, this investigation explores variation left largely untouched by existing efforts in suicidal behavior, providing a wealth of novel information to add to future investigations, such as meta-analyses.
Collapse
Affiliation(s)
- Eric T Monson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Mehdi Pirooznia
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Parla
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Melissa Kramer
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Marie E Gaine
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Sophia C Gaynor
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Kelly de Klerk
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Dubravka Jancic
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rachel Karchin
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Maryland, USA
| | - W Richard McCombie
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - James B Potash
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Virginia L Willour
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
21
|
Ishii K, Kubo KI, Nakajima K. Reelin and Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:229. [PMID: 27803648 PMCID: PMC5067484 DOI: 10.3389/fncel.2016.00229] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Proper neuronal migration and laminar formation during corticogenesis is essential for normal brain function. Disruption of these developmental processes is thought to be involved in the pathogenesis of some neuropsychiatric conditions. Especially, Reelin, a glycoprotein mainly secreted by the Cajal-Retzius cells and a subpopulation of GABAergic interneurons, has been shown to play a critical role, both during embryonic and postnatal periods. Indeed, animal studies have clearly revealed that Reelin is an essential molecule for proper migration of cortical neurons and finally regulates the cell positioning in the cortex during embryonic and early postnatal stages; by contrast, Reelin signaling is closely involved in synaptic function in adulthood. In humans, genetic studies have shown that the reelin gene (RELN) is associated with a number of psychiatric diseases, including Schizophrenia (SZ), bipolar disorder (BP) and autistic spectrum disorder. Indeed, Reln haploinsufficiency has been shown to cause cognitive impairment in rodents, suggesting the expression level of the Reelin protein is closely related to the higher brain functions. However, the molecular abnormalities in the Reelin pathway involved in the pathogenesis of psychiatric disorders are not yet fully understood. In this article, we review the current progress in the understanding of the Reelin functions that could be related to the pathogenesis of psychiatric disorders. Furthermore, we discuss the basis for selecting Reelin and molecules in its downstream signaling pathway as potential therapeutic targets for psychiatric illnesses.
Collapse
Affiliation(s)
- Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
22
|
Mice that lack the C-terminal region of Reelin exhibit behavioral abnormalities related to neuropsychiatric disorders. Sci Rep 2016; 6:28636. [PMID: 27346785 PMCID: PMC4921851 DOI: 10.1038/srep28636] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/06/2016] [Indexed: 12/29/2022] Open
Abstract
The secreted glycoprotein Reelin is believed to play critical roles in the pathogenesis of several neuropsychiatric disorders. The highly basic C-terminal region (CTR) of Reelin is necessary for efficient activation of its downstream signaling, and the brain structure of knock-in mice that lack the CTR (ΔC-KI mice) is impaired. Here, we performed a comprehensive behavioral test battery on ΔC-KI mice, in order to evaluate the effects of partial loss-of-function of Reelin on brain functions. The ΔC-KI mice were hyperactive and exhibited reduced anxiety-like and social behaviors. The working memory in ΔC-KI mice was impaired in a T-maze test. There was little difference in spatial reference memory, depression-like behavior, prepulse inhibition, or fear memory between ΔC-KI and wild-type mice. These results suggest that CTR-dependent Reelin functions are required for some specific normal brain functions and that ΔC-KI mice recapitulate some aspects of neuropsychiatric disorders, such as schizophrenia, bipolar disorder, and autism spectrum disorder.
Collapse
|
23
|
Lintas C, Sacco R, Persico AM. Differential methylation at the RELN gene promoter in temporal cortex from autistic and typically developing post-puberal subjects. J Neurodev Disord 2016; 8:18. [PMID: 27134686 PMCID: PMC4850686 DOI: 10.1186/s11689-016-9151-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/12/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Reelin plays a pivotal role in neurodevelopment and in post-natal synaptic plasticity and has been implicated in the pathogenesis of autism spectrum disorder (ASD). The reelin (RELN) gene expression is significantly decreased in ASD, both in the brain and peripherally. Methylation at the RELN gene promoter is largely triggered at puberty, and hypermethylation has been found in post-mortem brains of schizophrenic and bipolar patients. METHODS In this study, we assessed RELN gene methylation status in post-mortem temporocortical tissue samples (BA41/42 or 22) of six pairs of post-puberal individuals with ASD and typically developing subjects, matched for sex (male:female, M:F = 5:1), age, and post-mortem interval. RESULTS ASD patients display a significantly higher number of methylated CpG islands and heavier methylation in the 5' region of the RELN gene promoter, spanning from -458 to -223 bp, whereas controls have more methylated CpG positions and greater extent of methylation at the 3' promoter region, spanning from -222 to +1 bp. The most upstream promoter region (-458 to -364 bp) is methylated only in ASD brains, while the most downstream region (-131 to +1 bp) is methylated exclusively in control brains. Within this general framework, three different methylation patterns are discernible, each correlated with different extents of reduction in reelin gene expression among ASD individuals compared to controls. CONCLUSIONS The methylation pattern is different in ASD and control post-mortem brains. ASD-specific CpG positions, located in the most upstream gene promoter region, may exert a functional role potentially conferring ASD risk by blunting RELN gene expression.
Collapse
Affiliation(s)
- Carla Lintas
- Unit of Child and Adolescent Neuropsychiatry, University Campus Bio-Medico, Rome, Italy ; Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Roberto Sacco
- Unit of Child and Adolescent Neuropsychiatry, University Campus Bio-Medico, Rome, Italy ; Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Antonio M Persico
- Unit of Child and Adolescent Neuropsychiatry, "Gaetano Martino" University Hospital, University of Messina, via Consolare Valeria 1, I-98125 Messina, Italy ; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| |
Collapse
|
24
|
Kwon HJ, Jang WC, Lim MH. Association between RELN Gene Polymorphisms and Attention Deficit Hyperactivity Disorder in Korean Children. Psychiatry Investig 2016; 13:210-6. [PMID: 27081382 PMCID: PMC4823197 DOI: 10.4306/pi.2016.13.2.210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Attention deficit hyperactivity disorder (ADHD) is common disorder of the school-age population. ADHD is familial and genetic studies estimate heritability at 80-90%. The aim of the present study was to investigate the association between the genetic type and alleles for RELNgene (rs736707, rs2229864, rs362746, rs362726, rs362691, rs1062831, rs607755, and rs2072403) in Korean children with ADHD. METHODS The sample consisted of 180 ADHD children and 159 control children. We diagnosed ADHD according to DSM-IV. ADHD symptoms were evaluated with Conners' Parent Rating Scales and Dupaul Parent ADHD Rating Scales. Blood samples were taken from the 339 subjects, DNA was extracted from blood lymphocytes, and PCR was performed for RELN Polymorphism. Alleles and genotype frequencies were compared using the chi-square test. We compared the allele and genotype frequencies of RELN gene polymorphism in the ADHD and control groups. RESULTS This study showed that there was a significant correlation among the frequencies of the rs736707 (OR=1.40, 95% CI=1.03-1.90, p=0.031) of alleles of RELN, but the final conclusions are not definite. CONCLUSION Follow up studies with larger patient or pure subgroups are expected. These results suggested that RELN might be related to ADHD symptoms.
Collapse
Affiliation(s)
- Ho Jang Kwon
- Environmental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Won-Cheol Jang
- Department of Chemistry, College of Natural Science, Dankook University, Cheonan, Republic of Korea
| | - Myung Ho Lim
- Environmental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
- Department of Psychology, College of Public Welfare, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
25
|
Neuronal Heterotopias Affect the Activities of Distant Brain Areas and Lead to Behavioral Deficits. J Neurosci 2015; 35:12432-45. [PMID: 26354912 DOI: 10.1523/jneurosci.3648-14.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuronal heterotopia refers to brain malformations resulting from deficits of neuronal migration. Individuals with heterotopias show a high incidence of neurological deficits, such as epilepsy. More recently, it has come to be recognized that focal heterotopias may also show a range of psychiatric problems, including cognitive and behavioral impairments. However, because focal heterotopias are not always located in the brain areas responsible for the symptoms, the causal relationship between the symptoms and heterotopias remains elusive. In this study, we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited spatial working memory deficit and low competitive dominance behavior, which have been shown to be closely associated with the activity of the medial prefrontal cortex (mPFC) in rodents. Analysis of the mPFC activity revealed that the immediate-early gene expression was decreased and the local field potentials of the mPFC were altered in the mice with heterotopias compared with the control mice. Moreover, activation of these ectopic and overlying sister neurons using the DREADD (designer receptor exclusively activated by designer drug) system improved the working memory deficits. These findings suggest that cortical regions containing focal heterotopias can affect distant brain regions and give rise to behavioral abnormalities. Significance statement: Recent studies reported that patients with heterotopias have a variety of clinical symptoms, such as cognitive disturbance, psychiatric symptoms, and autistic behavior. However, the causal relationship between the symptoms and heterotopias remains elusive. Here we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited behavioral deficits that have been shown to be associated with the mPFC activity in rodents. The existence of heterotopias indeed altered the neural activities of the mPFC, and direct manipulation of the neural activity of the ectopic neurons and their sister neurons in the overlying cortex improved the behavioral deficit. Thus, our results indicate that focal heterotopias could affect the activities of distant brain areas and cause behavioral abnormalities.
Collapse
|
26
|
Convergent Lines of Evidence Support LRP8 as a Susceptibility Gene for Psychosis. Mol Neurobiol 2015; 53:6608-6619. [PMID: 26637325 DOI: 10.1007/s12035-015-9559-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/22/2015] [Indexed: 12/23/2022]
Abstract
Reelin (RELN) is identified as a risk gene for major psychiatric disorders such as schizophrenia (SCZ) and bipolar disorder (BPD). However, the role of its downstream signaling molecule, the low-density lipoprotein receptor-related protein 8 (LRP8) in these illnesses is still unclear. To detect whether LRP8 is a susceptibility gene for SCZ and BPD, we analyzed the associations of single nucleotide polymorphisms (SNPs) in LRP8 in a total of 47,187 subjects (including 9379 SCZ patients; 6990 BPD patients; and 12,556 controls in a screening sample, and 1397 SCZ families, 3947 BPD patients, and 8387 controls in independent replications), and identified a non-synonymous SNP rs5174 in LRP8 significantly associated with SCZ and BPD as well as the combined psychosis phenotype (P meta = 1.99 × 10-5, odds ratio (OR) = 1.066, 95 % confidence interval (CI) = 1.035-1.098). The risk SNP rs5174 was also associated with LRP8 messenger RNA (mRNA) expression in multiple brain tissues across independent samples (lowest P = 0.00005). Further exploratory analysis revealed that LRP8 was preferentially expressed in fetal brain tissues. Protein-protein interaction (PPI) analysis demonstrated that LRP8 significantly participated in a highly interconnected PPI network build by top risk genes for SCZ and BPD (P = 7.0 × 10-4). Collectively, we confirmed that LRP8 is a risk gene for psychosis, and our results provide useful information toward a better understanding of genetic mechanism involving LRP8 underlying risk of complex psychiatric disorders.
Collapse
|
27
|
Heilbronner U, Malzahn D, Strohmaier J, Maier S, Frank J, Treutlein J, Mühleisen TW, Forstner AJ, Witt SH, Cichon S, Falkai P, Nöthen MM, Rietschel M, Schulze TG. A common risk variant in CACNA1C supports a sex-dependent effect on longitudinal functioning and functional recovery from episodes of schizophrenia-spectrum but not bipolar disorder. Eur Neuropsychopharmacol 2015; 25:2262-70. [PMID: 26475575 DOI: 10.1016/j.euroneuro.2015.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/26/2015] [Accepted: 09/24/2015] [Indexed: 12/13/2022]
Abstract
Sex is a powerful modulator of disease susceptibility, course and outcome. The gene CACNA1C is among the best replicated vulnerability genes of bipolar disorder and schizophrenia. The aim of the present study was to investigate whether sex and a variant in CACNA1C (rs10774035 as a proxy for the well-acknowledged risk variant rs1006737) influence psychosocial adaptation in a large German patient sample with schizophrenia-spectrum (n=297) and bipolar (n=516) disorders. We analyzed Global Assessment of Functioning (GAF) scores, retrospectively collected for different time points during disease course. We investigated whether CACNA1C sex-dependently modulates longitudinal GAF scores and recovery from episodes of psychiatric disturbance in the above mentioned disorders. Psychosocial recovery was measured as difference score between the current GAF score (assessing the last remission) and the worst GAF score ever during an illness episode. Covariate- adjusted association analyses revealed a sex × rs10774035 genotype interaction on longitudinal GAF and recovery from illness episodes only in schizophrenia-spectrum but not in bipolar disorders. In schizophrenia-spectrum affected males, rs10774035 minor allele (T) carriers had higher GAF scores at three time points (premorbid, worst ever, current). In contrast, females carrying rs10774035 minor alleles had impaired recovery from schizophrenia-spectrum episodes. These results encourage further investigations of gene × sex interactions and longitudinal quantitative phenotypes to unravel the rich variety of behavioral consequences of genetic individuality.
Collapse
Affiliation(s)
- Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich, Germany.
| | - Dörthe Malzahn
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Jana Strohmaier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Sandra Maier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Jens Treutlein
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich (FZJ), Jülich, Germany; Institute of Human Genetics, University of Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn, Germany; Division of Medical Genetics, University Hospital Basel, University of Basel, Switzerland; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University, Göttingen, Germany
| |
Collapse
|
28
|
Chen X, Long F, Cai B, Chen X, Chen G. A novel relationship for schizophrenia, bipolar and major depressive disorder Part 7: A hint from chromosome 7 high density association screen. Behav Brain Res 2015; 293:241-51. [PMID: 26192912 DOI: 10.1016/j.bbr.2015.06.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/25/2015] [Accepted: 06/25/2015] [Indexed: 11/18/2022]
Abstract
Convergent evidence from genetics, symptology and psychopharmacology imply that there are intrinsic connection between schizophrenia (SCZ), bipolar disorder (BPD) and major depressive disorder (MDD). Also, any two or even three of these disorders could co-existe in some families. A total of 47,144 single nucleotide polymorphism (SNPs) on chromosome 7 were genotyped by Affymetrix Genome-Wide Human SNP array 6.0 on 119 SCZ, 253 BPD (type-I), 177 MDD, and 1000 controls. Associated SNP loci were comprehensively revealed and outstanding susceptibility genes were identified including CNTNAP2. a neurexin family gene. Unexpectedly, flanking genes for up to 94.74 % of of the associated SNPs were replicated (P≤9.9 E-8) in an enlarged cohort of 986 SCZ patients. Considering other convergent evidence, our results further implicate that BPD and MDD are subtypes of SCZ.
Collapse
Affiliation(s)
- Xing Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, Shandong, People's Republic of China
| | - Feng Long
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, Shandong, People's Republic of China
| | - Bin Cai
- CapitalBio corporation, 18 Life Science Parkway, Changping District, Beijing 102206, People's Republic of China
| | - Xiaohong Chen
- CapitalBio corporation, 18 Life Science Parkway, Changping District, Beijing 102206, People's Republic of China
| | - Gang Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, Shandong, People's Republic of China.
| |
Collapse
|
29
|
Ishii K, Nagai T, Hirota Y, Noda M, Nabeshima T, Yamada K, Kubo KI, Nakajima K. Reelin has a preventive effect on phencyclidine-induced cognitive and sensory-motor gating deficits. Neurosci Res 2015; 96:30-6. [PMID: 25573715 DOI: 10.1016/j.neures.2014.12.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 12/26/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022]
Abstract
Reelin has recently attracted attention because of its connection to several neuropsychiatric diseases. We previously reported the finding that prior transplantation of GABAergic neuron precursor cells into the medial prefrontal cortex (mPFC) of mice significantly prevented the induction of cognitive and sensory-motor gating deficits induced by phencyclidine (PCP). The majority of the precursor cells transplanted into the mPFC of the recipient mice differentiated into members of a somatostatin/Reelin-expressing class of GABAergic interneurons. These findings raised the possibility that Reelin secreted by the transplanted cells plays an important role in preventing the deficits induced by PCP. In this study, we investigated whether Reelin itself has a preventive effect on PCP-induced behavioral phenotypes by injecting conditioned medium containing Reelin into the lateral ventricle of the brains of 6- to 7-week-old male mice before administrating PCP. Behavioral analyses showed that the prior Reelin injection had a preventive effect against induction of the cognitive and sensory-motor gating deficits associated with PCP. Moreover, one of the types of Reelin receptor was found to be expressed by neurons in the mPFC. The results of this study point to the Reelin signaling pathway as a candidate target for the pharmacologic treatment of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University School of Medicine, Nagoya, Japan
| | - Yuki Hirota
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Mariko Noda
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Toshitaka Nabeshima
- Department of Regional Pharmaceutical Care & Sciences, Meijo University, Nagoya, Japan; NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University School of Medicine, Nagoya, Japan
| | - Ken-ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
30
|
Bicker S, Lackinger M, Weiß K, Schratt G. MicroRNA-132, -134, and -138: a microRNA troika rules in neuronal dendrites. Cell Mol Life Sci 2014; 71:3987-4005. [PMID: 25008044 PMCID: PMC11113804 DOI: 10.1007/s00018-014-1671-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/11/2014] [Accepted: 06/20/2014] [Indexed: 01/19/2023]
Abstract
Dendritic mRNA transport and local translation in the postsynaptic compartment play an important role in synaptic plasticity, learning and memory. Local protein synthesis at the synapse has to be precisely orchestrated by a plethora of factors including RNA binding proteins as well as microRNAs, an extensive class of small non-coding RNAs. By binding to complementary sequences in target mRNAs, microRNAs fine-tune protein synthesis and thereby represent critical regulators of gene expression at the post-transcriptional level. Research over the last years identified an entire network of dendritic microRNAs that fulfills an essential role in synapse development and physiology. Recent studies provide evidence that these small regulatory molecules are highly regulated themselves, at the level of expression as well as function. The importance of microRNAs for correct function of the nervous system is reflected by an increasing number of studies linking dysregulation of microRNA pathways to neurological disorders. By focusing on three extensively studied examples (miR-132, miR-134, miR-138), this review will attempt to illustrate the complex regulatory roles of dendritic microRNAs at the synapse and their implications for pathological conditions.
Collapse
Affiliation(s)
- Silvia Bicker
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Martin Lackinger
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Kerstin Weiß
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gerhard Schratt
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
31
|
Abstract
The extracellular matrix (ECM) has a prominent role in brain development, maturation of neural circuits, and adult neuroplasticity. This multifactorial role of the ECM suggests that processes that affect composition or turnover of ECM in the brain could lead to altered brain function, possibly underlying conditions of impaired mental health, such as neuropsychiatric or neurodegenerative disease. In support of this, in the last two decades, clinical and preclinical research provided evidence of correlations and to some degree causal links, between aberrant ECM function and neuropsychiatric disorders, the most prominent being addiction and schizophrenia. Based on these initial observations of involvement of different classes of ECM molecules (laminin, reelin, and their integrin receptors, as well as tenascins and chondroitin sulfate proteoglycans), ECM targets have been suggested as a novel entry point in the treatment of neuropsychiatric disorders. Hence, understanding how ECM molecules contribute to proper neuronal functioning and how this is dysregulated in conditions of mental illness is of pivotal importance. In this chapter, we will review available literature that implicates the different classes of brain ECM molecules in psychiatric disorders, with a primary focus on addiction (opiates, psychostimulants, and alcohol), and we will compare these ECM adaptations with those implicated in schizophrenia and mood disorders.
Collapse
|
32
|
The psychiatric vulnerability gene CACNA1C and its sex-specific relationship with personality traits, resilience factors and depressive symptoms in the general population. Mol Psychiatry 2013; 18:607-13. [PMID: 22665259 DOI: 10.1038/mp.2012.53] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genome-wide association studies have reported an association between the A-allele of rs1006737 within CACNA1C and affective disorders and schizophrenia. The aim of the present study was to investigate the relationship between rs1006737 and established and potential endophenotypes for these disorders in a population-based cohort of 3793 subjects, using an analytical method designed to assess a previously reported sex-specific effect of CACNA1C. The investigated endophenotypes included personality traits and resilience factors. At 10-year follow-up, subjects were screened for depressive symptoms. All subjects were genotyped for rs1006737. The direction of the effect and mode of inheritance of rs1006737 differed between the sexes. In men, the A-allele was associated with higher emotional lability and lower resilience, that is, lower sense of coherence (P=0.021), lower perceived social support (P=0.018), lower dispositional optimism (P=0.032) and more depressive symptoms at follow-up (P=0.007). In women, the A-allele was associated with lower emotional lability and stronger resilience, that is, higher sense of coherence (P=0.00028), higher perceived social support (P=0.010), lower neuroticism (P=0.022) and fewer depressive symptoms at follow-up (P=0.035). After conservative Bonferroni correction for 32 tests, results only remained significant for sense of coherence in women (P=0.009). These results suggest that CACNA1C is involved in the genetic architecture of endophenotypes for affective disorders and schizophrenia, and that it shows a distinct sex-specific effect. Comprehensive phenotype characterization in case-control samples and the general population, as well as an adequate modeling of sex-specific genetic effects, may be warranted to elucidate the pathogenetic mechanisms conferred by robustly identified susceptibility genes.
Collapse
|
33
|
Folsom TD, Fatemi SH. The involvement of Reelin in neurodevelopmental disorders. Neuropharmacology 2013; 68:122-35. [PMID: 22981949 PMCID: PMC3632377 DOI: 10.1016/j.neuropharm.2012.08.015] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 08/14/2012] [Accepted: 08/16/2012] [Indexed: 12/21/2022]
Abstract
Reelin is a glycoprotein that serves important roles both during development (regulation of neuronal migration and brain lamination) and in adulthood (maintenance of synaptic function). A number of neuropsychiatric disorders including autism, schizophrenia, bipolar disorder, major depression, Alzheimer's disease and lissencephaly share a common feature of abnormal Reelin expression in the brain. Altered Reelin expression has been hypothesized to impair neuronal connectivity and synaptic plasticity, leading ultimately to the cognitive deficits present in these disorders. The mechanisms for abnormal Reelin expression in some of these disorders are currently unknown although possible explanations include early developmental insults, mutations, hypermethylation of the promoter for the Reelin gene (RELN), miRNA silencing of Reelin mRNA, FMRP underexpression and Reelin processing abnormalities. Increasing Reelin expression through pharmacological therapies may help ameliorate symptoms resulting from Reelin deficits. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Timothy D. Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
| | - S. Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
- Department of Pharmacology, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
| |
Collapse
|
34
|
Hill RA, Wu YWC, Gogos A, van den Buuse M. Sex-dependent alterations in BDNF-TrkB signaling in the hippocampus of reelin heterozygous mice: a role for sex steroid hormones. J Neurochem 2013; 126:389-99. [DOI: 10.1111/jnc.12205] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/13/2013] [Accepted: 02/13/2013] [Indexed: 12/01/2022]
Affiliation(s)
- Rachel A. Hill
- Behavioural Neuroscience Laboratory; Mental Health Research Institute; Melbourne Australia
- Centre for Neuroscience; University of Melbourne; Melbourne Australia
| | - Yee-Wen Candace Wu
- Behavioural Neuroscience Laboratory; Mental Health Research Institute; Melbourne Australia
- Department of Pharmacology; University of Melbourne; Melbourne Australia
| | - Andrea Gogos
- Behavioural Neuroscience Laboratory; Mental Health Research Institute; Melbourne Australia
- Centre for Neuroscience; University of Melbourne; Melbourne Australia
| | - Maarten van den Buuse
- Behavioural Neuroscience Laboratory; Mental Health Research Institute; Melbourne Australia
- Department of Pharmacology; University of Melbourne; Melbourne Australia
| |
Collapse
|
35
|
Reelin and its complex involvement in brain development and function. Int J Biochem Cell Biol 2012; 44:1501-4. [PMID: 22705982 DOI: 10.1016/j.biocel.2012.06.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 05/25/2012] [Accepted: 06/04/2012] [Indexed: 12/29/2022]
Abstract
Reelin is a neuroprotein with crucial role during neurodevelopment and also in postnatal period. It regulates neuronal migration and positioning in developing neocortex and cerebellar cortex. Postnatally it participates in regulation of dendritic and axonal growth, synaptogenesis, neurotransmission and it contribute to synaptic plasticity necessary for learning and memory functions. Role of Reelin seems to be rather complex, profound research gradually uncovers its further functions. Deficits of Reelin were detected in neuropsychiatric disorders such as schizophrenia, bipolar disorder and autism. Pathogenesis of these disorders is far from being clearly understood. Reelin contribution to these diseases seems to be vital, since genetic variants of Reelin were associated with these diseases and often influence symptom severity. Reelin is a promising candidate molecule with potential future use in diagnostics and therapy, however further detailed research is essential.
Collapse
|
36
|
Brummelte S, Galea L, Devlin A, Oberlander T. Antidepressant use during pregnancy and serotonin transporter genotype (SLC6A4) Affect newborn serum reelin levels. Dev Psychobiol 2012; 55:518-29. [DOI: 10.1002/dev.21056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/16/2012] [Indexed: 12/31/2022]
|
37
|
Altered N-methyl-D-aspartate receptor function in reelin heterozygous mice: male-female differences and comparison with dopaminergic activity. Prog Neuropsychopharmacol Biol Psychiatry 2012; 37:237-46. [PMID: 22361156 DOI: 10.1016/j.pnpbp.2012.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/07/2012] [Accepted: 02/09/2012] [Indexed: 01/19/2023]
Abstract
The aim of this study was to investigate the in vivo relationship between reelin and NMDA receptor function in schizophrenia. We assessed the effect of reelin deficiency in behavioral models of aspects of this illness, NMDA receptor subunit levels, and NMDA receptor, dopamine D₂ receptor, and dopamine transporter density. Male, but not female, reelin heterozygous mice showed significantly enhanced MK-801-induced locomotor hyperactivity compared to wildtype controls (7.4-fold vs. 5.2-fold effect of MK-801 over saline, respectively) but there were no genotype differences in the response to amphetamine. Both male and female reelin heterozygous mice showed enhanced effects of MK-801 on startle, but not prepulse inhibition (PPI) of startle. There were no group differences in the effect of apomorphine on startle or PPI. The levels of NMDA receptor subunits were not altered in the striatum. In the frontal cortex, male and female reelin heterozygous mice showed significant up-regulation of NR1 subunits, but down-regulation of NR2C subunits, which was associated with significantly elevated NR1/NR2A and NR1/NR2C ratios. However, there were no differences in [³H]MK-801 binding density in the nucleus accumbens or caudate nucleus, nor in the density of [³H]YM-09151 or [³H]GBR12935 in these brain regions. The enhanced effects of MK-801 in reelin heterozygous mice in this study could be reflective of the role of reelin deficiency in schizophrenia. This genotype effect was male-specific for locomotor hyperactivity, a model of psychosis, but was seen in male and female mice for startle, which could be an indication of changes in anxiety. Changes in NMDA receptor subunit levels and ratios were also seen in both male and female mice. These results suggest that the role of reelin deficiency in schizophrenia may be particularly mediated by altered NMDA receptor responses, with some of these effects being strictly sex-specific.
Collapse
|
38
|
Kim SH, Shin SY, Lee KY, Joo EJ, Song JY, Ahn YM, Lee YH, Kim YS. The genetic association of DUSP6 with bipolar disorder and its effect on ERK activity. Prog Neuropsychopharmacol Biol Psychiatry 2012; 37:41-9. [PMID: 22155192 DOI: 10.1016/j.pnpbp.2011.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/05/2011] [Accepted: 11/24/2011] [Indexed: 12/21/2022]
Abstract
The dual-specificity phosphatase 6 (DUSP6) gene resides at chromosome location 12q22-23, which is one of the candidate loci for susceptibility to bipolar disorder and which encodes a phosphatase selective for extracellular signal-regulated kinase (ERK). Previously, we reported a positive association between the functional Leu114Val polymorphism (rs2279574) in DUSP6 and bipolar disorder. Given that the association between DUSP6 and the reported down-regulation of DUSP6 transcript in bipolar postmortem brains were sex-dimorphic, showing significance in women but not men, we performed two independent analyses in homogenous samples of male and female Korean patients with bipolar disorder or schizophrenia using samples enlarged from our previous report. Among the examined DUSP6 SNPs, five (rs769700, rs704076, rs770087, rs808820, and rs2279574) showed positive allelic associations, with the frequency of minor alleles (C, T, G, G, and G) in each SNP significantly increased in women with BD. Consequently, the "C-T-G-G-G" haplotype was significantly over-represented (P=0.016; OR=3.242), whereas the "T-G-T-A-T" haplotype was significantly under-represented (P=0.014; OR=0.697). We found no significant associations with DUSP6 SNPs in men with bipolar disorder or schizophrenia. We also investigated the functions of the functional SNPs' positive associations and found that Leu114Val (rs2279574; T/G) and Ser144Ala (rs770087; T/G) mutations in DUSP6 proteins reduced lithium-induced ERK1/2 phosphorylation in vitro, implicating the dominant active functions. Thus, DUSP6 may not only play important roles in the pathogenesis of bipolar disorder, particularly in women, but also affect the therapeutic response to lithium through modulating lithium's effects on intracellular signaling.
Collapse
Affiliation(s)
- Se Hyun Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Migración neuronal, apoptosis y trastorno bipolar. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2012; 5:127-33. [DOI: 10.1016/j.rpsm.2011.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 11/12/2011] [Accepted: 11/28/2011] [Indexed: 11/23/2022]
|
41
|
Liu LY, Schaub MA, Sirota M, Butte AJ. Sex differences in disease risk from reported genome-wide association study findings. Hum Genet 2012; 131:353-64. [PMID: 21858542 PMCID: PMC3260375 DOI: 10.1007/s00439-011-1081-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/08/2011] [Indexed: 01/01/2023]
Abstract
Men and women differ in susceptibility to many diseases and in responses to treatment. Recent advances in genome-wide association studies (GWAS) provide a wealth of data for associating genetic profiles with disease risk; however, in general, these data have not been systematically probed for sex differences in gene-disease associations. Incorporating sex into the analysis of GWAS results can elucidate new relationships between single nucleotide polymorphisms (SNPs) and human disease. In this study, we performed a sex-differentiated analysis on significant SNPs from GWAS data of the seven common diseases studied by the Wellcome Trust Case Control Consortium. We employed and compared three methods: logistic regression, Woolf's test of heterogeneity, and a novel statistical metric that we developed called permutation method to assess sex effects (PMASE). After correction for false discovery, PMASE finds SNPs that are significantly associated with disease in only one sex. These sexually dimorphic SNP-disease associations occur in Coronary Artery Disease and Crohn's Disease. GWAS analyses that fail to consider sex-specific effects may miss discovering sexual dimorphism in SNP-disease associations that give new insights into differences in disease mechanism between men and women.
Collapse
Affiliation(s)
- Linda Y. Liu
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| | - Marc A. Schaub
- Computer Science Department, Stanford University, 353 Serra Mall, Stanford, CA 94305, USA,
| | - Marina Sirota
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| | - Atul J. Butte
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA,
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| |
Collapse
|
42
|
Claes S, Tang YL, Gillespie CF, Cubells JF. Human genetics of schizophrenia. HANDBOOK OF CLINICAL NEUROLOGY 2012; 106:37-52. [DOI: 10.1016/b978-0-444-52002-9.00003-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
Overexpression of Reelin prevents the manifestation of behavioral phenotypes related to schizophrenia and bipolar disorder. Neuropsychopharmacology 2011; 36:2395-405. [PMID: 21814183 PMCID: PMC3194085 DOI: 10.1038/npp.2011.153] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Despite the impact of schizophrenia and mood disorders, which in extreme cases can lead to death, recent decades have brought little progress in the development of new treatments. Recent studies have shown that Reelin, an extracellular protein that is critical for neuronal development, is reduced in schizophrenia and bipolar disorder patients. However, data on a causal or protective role of Reelin in psychiatric diseases is scarce. In order to study the direct influence of Reelin's levels on behavior, we subjected two mouse lines, in which Reelin levels are either reduced (Reelin heterozygous mice) or increased (Reelin overexpressing mice), to a battery of behavioral tests: open-field, black-white box, novelty-suppressed-feeding, forced-swim-test, chronic corticosterone treatment followed by forced-swim-test, cocaine sensitization and pre-pulse inhibition (PPI) deficits induced by N-methyl-D-aspartate (NMDA) antagonists. These tests were designed to model some aspects of psychiatric disorders such as schizophrenia, mood, and anxiety disorders. We found no differences between Reeler heterozygous mice and their wild-type littermates. However, Reelin overexpression in the mouse forebrain reduced the time spent floating in the forced-swim-test in mice subjected to chronic corticosterone treatment, reduced behavioral sensitization to cocaine, and reduced PPI deficits induced by a NMDA antagonist. In addition, we demonstrate that while stress increased NMDA NR2B-mediated synaptic transmission, known to be implicated in depression, Reelin overexpression significantly reduced it. Together, these results point to the Reelin signaling pathway as a relevant drug target for the treatment of a range of psychiatric disorders.
Collapse
|
44
|
Abstract
Schizophrenia (SCZ) is among the most disabling of mental disorders. Several neurobiological hypotheses have been postulated as responsible for SCZ pathogenesis: polygenic/multifactorial genomic defects, intrauterine and perinatal environment-genome interactions, neurodevelopmental defects, dopaminergic, cholinergic, serotonergic, gamma-aminobutiric acid (GABAergic), neuropeptidergic and glutamatergic/N-Methyl-D-Aspartate (NMDA) dysfunctions, seasonal infection, neuroimmune dysfunction, and epigenetic dysregulation. SCZ has a heritability estimated at 60-90%. Genetic studies in SCZ have revealed the presence of chromosome anomalies, copy number variants, multiple single-nucleotide polymorphisms of susceptibility distributed across the human genome, aberrant single nucleotide polymorphisms (SNPs) in microRNA genes, mitochondrial DNA mutations, and epigenetic phenomena. Pharmacogenetic studies of psychotropic drug response have focused on determining the relationship between variation in specific candidate genes and the positive and adverse effects of drug treatment. Approximately, 18% of neuroleptics are major substrates of CYP1A2 enzymes, 40% of CYP2D6, and 23% of CYP3A4; 24% of antidepressants are major substrates of CYP1A2 enzymes, 5% of CYP2B6, 38% of CYP2C19, 85% of CYP2D6, and 38% of CYP3A4; 7% of benzodiazepines are major substrates of CYP2C19 enzymes, 20% of CYP2D6, and 95% of CYP3A4. About 10-20% of Western populations are defective in genes of the CYP superfamily. Only 26% of Southern Europeans are pure extensive metabolizers for the trigenic cluster integrated by the CYP2D6+CYP2C19+CYP2C9 genes. The pharmacogenomic response of SCZ patients to conventional psychotropic drugs also depends on genetic variants associated with SCZ-related genes. Consequently, the incorporation of pharmacogenomic procedures both to drugs in development and drugs on the market would help to optimize therapeutics in SCZ and other central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, 15165-Bergondo, Coruña, Spain.
| | | |
Collapse
|
45
|
Li W, Song X, Zhang H, Yang Y, Jiang C, Xiao B, Li W, Yang G, Zhao J, Guo W, Lv L. Association study of RELN polymorphisms with schizophrenia in Han Chinese population. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1505-11. [PMID: 21549172 DOI: 10.1016/j.pnpbp.2011.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 04/13/2011] [Accepted: 04/19/2011] [Indexed: 01/08/2023]
Abstract
Schizophrenia (SZ) is a common and complex psychiatric disorder with a strong genetic component. Previous research suggests that mutations altering genes in neurodevelopmental pathways contribute to SZ. Reelin gene (RELN) maps to chromosome 7q22.1, the encoded protein plays a pivotal role in guiding neuronal migration, lamination and connection during embryonic brain development. Several reports had indicated that reduced RELN expression is associated with human mental illnesses such as SZ, mood disorders and autism. In this study, case-control association analyses were performed in the Han Chinese population to determine if the RELN gene is a susceptibility gene for SZ. Thirty-seven single nucleotide polymorphisms (SNPs) were genotyped in 528 paranoid SZ patients and 528 control subjects. A significant association was found between rs12705169 and SZ (p=0.001). Moreover, the haplotypes constructed from five SNPs showed significant differences between cases and controls (p=0.041). When subjects were divided by gender, rs12705169 remained significant difference only in females (OR=0.24, 95%CI=0.14-0.40 for CC and OR=0.40, 95%CI=0.27-0.58 for AC), both in the allele and genotype (p=0.0001 for both). This study describes a positive association between RELN and SZ in the Han Chinese population, and provides genetic evidence to support the gender difference of SZ.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Samuels J, Wang Y, Riddle MA, Greenberg BD, Fyer AJ, McCracken JT, Rauch SL, Murphy DL, Grados MA, Knowles JA, Piacentini J, Cullen B, Bienvenu OJ, Rasmussen SA, Geller D, Pauls DL, Liang KY, Shugart YY, Nestadt G. Comprehensive family-based association study of the glutamate transporter gene SLC1A1 in obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:472-7. [PMID: 21445956 PMCID: PMC3082623 DOI: 10.1002/ajmg.b.31184] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 03/02/2011] [Indexed: 12/25/2022]
Abstract
SLC1A1 encodes a neuronal glutamate transporter and is a promising candidate gene for obsessive-compulsive disorder (OCD). Several independent research groups have reported significant associations between OCD and single nucleotide polymorphisms (SNPs) in this gene. Previously, we evaluated 13 SNPs in, or near, SLC1A1 and reported a strong association signal with rs301443, a SNP 7.5 kb downstream of the gene [Shugart et al. (2009); Am J Med Genet Part B 150B:886–892]. The aims of the current study were first, to further investigate this finding by saturating the region around rs301443; and second, to explore the entire gene more thoroughly with a dense panel of SNP markers. We genotyped an additional 111 SNPs in or near SLC1A1, covering from 9 kb upstream to 84 kb downstream of the gene at average spacing of 1.7 kb per SNP, and conducted family-based association analyses in 1,576 participants in 377 families.We found that none of the surrounding markers were in linkage disequilibrium with rs301443, nor were any associated with OCD. We also found that SNP rs4740788, located about 8.8 kb upstream of the gene, was associated with OCD in all families (P = 0.003) and in families with male affecteds (P = 0.002). A three-SNP haplotype (rs4740788–rs10491734–rs10491733) was associated with OCD in the total sample (P = 0.00015) and in families with male affecteds (P = 0.0007). Although of nominal statistical significance considering the number of comparisons, these findings provide further support for the involvement of SLC1A1 in the pathogenesis of OCD.
Collapse
Affiliation(s)
- Jack Samuels
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Ying Wang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark A. Riddle
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Benjamin D. Greenberg
- Department of Psychiatry and Human Behavior, Brown Medical School, Butler Hospital, Providence, Rhode Island
| | - Abby J. Fyer
- Department of Psychiatry, College of Physicians and Surgeons at Columbia University and the New York State Psychiatric Institute, New York City, New York
| | - James T. McCracken
- Department of Psychiatry and Biobehavioral Sciences University of California, Los Angeles, School of Medicine, Los Angeles, California
| | - Scott L. Rauch
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | | | - Marco A. Grados
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - James A. Knowles
- Department of Psychiatry, University of Southern California School of Medicine, Los Angeles, California
| | - John Piacentini
- Department of Psychiatry and Biobehavioral Sciences University of California, Los Angeles, School of Medicine, Los Angeles, California
| | - Bernadette Cullen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - O. Joseph Bienvenu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven A. Rasmussen
- Department of Psychiatry and Human Behavior, Brown Medical School, Butler Hospital, Providence, Rhode Island
| | - Daniel Geller
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - David L. Pauls
- Department of Psychiatry and Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kung-Yee Liang
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Yin Y. Shugart
- Genomic Research Branch, Division of Neuroscience and Basic Behavioral Science, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Gerald Nestadt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
Genetic variants in the RELN gene are associated with otosclerosis in multiple European populations. Hum Genet 2009; 127:155-62. [DOI: 10.1007/s00439-009-0754-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 10/12/2009] [Indexed: 10/20/2022]
|