1
|
Morini E, Gao D, Logan EM, Salani M, Krauson AJ, Chekuri A, Chen YT, Ragavendran A, Chakravarty P, Erdin S, Stortchevoi A, Svejstrup JQ, Talkowski ME, Slaugenhaupt SA. Developmental regulation of neuronal gene expression by Elongator complex protein 1 dosage. J Genet Genomics 2022; 49:654-665. [PMID: 34896608 PMCID: PMC9254147 DOI: 10.1016/j.jgg.2021.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/27/2021] [Accepted: 11/04/2021] [Indexed: 01/21/2023]
Abstract
Familial dysautonomia (FD), a hereditary sensory and autonomic neuropathy, is caused by a mutation in the Elongator complex protein 1 (ELP1) gene that leads to a tissue-specific reduction of ELP1 protein. Our work to generate a phenotypic mouse model for FD headed to the discovery that homozygous deletion of the mouse Elp1 gene leads to embryonic lethality prior to mid-gestation. Given that FD is caused by a reduction, not loss, of ELP1, we generated two new mouse models by introducing different copy numbers of the human FD ELP1 transgene into the Elp1 knockout mouse (Elp1-/-) and observed that human ELP1 expression rescues embryonic development in a dose-dependent manner. We then conducted a comprehensive transcriptome analysis in mouse embryos to identify genes and pathways whose expression correlates with the amount of ELP1. We found that ELP1 is essential for the expression of genes responsible for nervous system development. Further, gene length analysis of the differentially expressed genes showed that the loss of Elp1 mainly impacts the expression of long genes and that by gradually restoring Elongator, their expression is progressively rescued. Finally, through evaluation of co-expression modules, we identified gene sets with unique expression patterns that depended on ELP1 expression.
Collapse
Affiliation(s)
- Elisabetta Morini
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Dadi Gao
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Emily M Logan
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Monica Salani
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Aram J Krauson
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Anil Chekuri
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Yei-Tsung Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taiwan
| | - Ashok Ragavendran
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Probir Chakravarty
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Serkan Erdin
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Alexei Stortchevoi
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jesper Q Svejstrup
- Mechanisms of Transcription Laboratory, The Francis Crick Institute, London, UK; Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Susan A Slaugenhaupt
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Havula E, Ghazanfar S, Lamichane N, Francis D, Hasygar K, Liu Y, Alton LA, Johnstone J, Needham EJ, Pulpitel T, Clark T, Niranjan HN, Shang V, Tong V, Jiwnani N, Audia G, Alves AN, Sylow L, Mirth C, Neely GG, Yang J, Hietakangas V, Simpson SJ, Senior AM. Genetic variation of macronutrient tolerance in Drosophila melanogaster. Nat Commun 2022; 13:1637. [PMID: 35347148 PMCID: PMC8960806 DOI: 10.1038/s41467-022-29183-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 02/28/2022] [Indexed: 11/08/2022] Open
Abstract
Carbohydrates, proteins and lipids are essential nutrients to all animals; however, closely related species, populations, and individuals can display dramatic variation in diet. Here we explore the variation in macronutrient tolerance in Drosophila melanogaster using the Drosophila genetic reference panel, a collection of ~200 strains derived from a single natural population. Our study demonstrates that D. melanogaster, often considered a "dietary generalist", displays marked genetic variation in survival on different diets, notably on high-sugar diet. Our genetic analysis and functional validation identify several regulators of macronutrient tolerance, including CG10960/GLUT8, Pkn and Eip75B. We also demonstrate a role for the JNK pathway in sugar tolerance and de novo lipogenesis. Finally, we report a role for tailless, a conserved orphan nuclear hormone receptor, in regulating sugar metabolism via insulin-like peptide secretion and sugar-responsive CCHamide-2 expression. Our study provides support for the use of nutrigenomics in the development of personalized nutrition.
Collapse
Affiliation(s)
- E Havula
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - S Ghazanfar
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - N Lamichane
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - D Francis
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - K Hasygar
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Y Liu
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - L A Alton
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - J Johnstone
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - E J Needham
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Pulpitel
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Clark
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - H N Niranjan
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Shang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Tong
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - N Jiwnani
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - G Audia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A N Alves
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - L Sylow
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - C Mirth
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - G G Neely
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - J Yang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Hietakangas
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - S J Simpson
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A M Senior
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
3
|
Cerebellin-2 regulates a serotonergic dorsal raphe circuit that controls compulsive behaviors. Mol Psychiatry 2021; 26:7509-7521. [PMID: 34158618 PMCID: PMC8692491 DOI: 10.1038/s41380-021-01187-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Cerebellin-1 (Cbln1) and cerebellin-2 (Cbln2) are secreted glycoproteins that are expressed in distinct subsets of neurons throughout the brain. Cbln1 and Cbln2 simultaneously bind to presynaptic neurexins and postsynaptic GluD1 and GluD2, thereby forming trans-synaptic adhesion complexes. Genetic associations link cerebellins, neurexins and GluD's to neuropsychiatric disorders involving compulsive behaviors, such as Tourette syndrome, attention-deficit hyperactivity disorder (ADHD), and obsessive-compulsive disorder (OCD). Extensive evidence implicates dysfunction of serotonergic signaling in these neuropsychiatric disorders. Here, we report that constitutive Cbln2 KO mice, but not Cbln1 KO mice, display robust compulsive behaviors, including stereotypic pattern running, marble burying, explosive jumping, and excessive nest building, and exhibit decreased brain serotonin levels. Strikingly, treatment of Cbln2 KO mice with the serotonin precursor 5-hydroxytryptophan or the serotonin reuptake-inhibitor fluoxetine alleviated compulsive behaviors. Conditional deletion of Cbln2 both from dorsal raphe neurons and from presynaptic neurons synapsing onto dorsal raphe neurons reproduced the compulsive behaviors of Cbln2 KO mice. Finally, injection of recombinant Cbln2 protein into the dorsal raphe of Cbln2 KO mice largely reversed their compulsive behaviors. Taken together, our results show that Cbln2 controls compulsive behaviors by regulating serotonergic circuits in the dorsal raphe.
Collapse
|
4
|
Nelson AT, Wang Y, Nelson ER. TLX, an Orphan Nuclear Receptor With Emerging Roles in Physiology and Disease. Endocrinology 2021; 162:6360449. [PMID: 34463725 PMCID: PMC8462384 DOI: 10.1210/endocr/bqab184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 12/14/2022]
Abstract
TLX (NR2E1), an orphan member of the nuclear receptor superfamily, is a transcription factor that has been described to be generally repressive in nature. It has been implicated in several aspects of physiology and disease. TLX is best known for its ability to regulate the proliferation of neural stem cells and retinal progenitor cells. Dysregulation, overexpression, or loss of TLX expression has been characterized in numerous studies focused on a diverse range of pathological conditions, including abnormal brain development, psychiatric disorders, retinopathies, metabolic disease, and malignant neoplasm. Despite the lack of an identified endogenous ligand, several studies have described putative synthetic and natural TLX ligands, suggesting that this receptor may serve as a therapeutic target. Therefore, this article aims to briefly review what is known about TLX structure and function in normal physiology, and provide an overview of TLX in regard to pathological conditions. Particular emphasis is placed on TLX and cancer, and the potential utility of this receptor as a therapeutic target.
Collapse
Affiliation(s)
- Adam T Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Yu Wang
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
- Correspondence: Erik R. Nelson, PhD, Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 407 S Goodwin Ave (MC-114), Urbana, IL 61801, USA.
| |
Collapse
|
5
|
He G, Gu J, Wang H, Cheng S, Xiong Q, Ke M, Hu Y, Feng J, Song L, Liu Z, Xu Y. Nr2e1 deficiency aggravates insulin resistance and chronic inflammation of visceral adipose tissues in a diet-induced obese mice model. Life Sci 2021; 278:119562. [PMID: 33915130 DOI: 10.1016/j.lfs.2021.119562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/11/2021] [Accepted: 04/23/2021] [Indexed: 10/21/2022]
Abstract
AIMS To investigate the nuclear receptor subfamily 2 group E member 1 (Nr2e1) expression in adipose tissues of obese mice and assess the role of Nr2e1 in insulin resistance and chronic inflammation of the adipose tissues. MAIN METHODS An obese model was established in Nr2e1 knockout (KO) mice and their wild type (WT) littermates through a long-term high-fat diet (HFD) feeding regime. The epididymal fat weight, body weight, and daily food intake were recorded. The blood lipid profile, blood inflammatory factors, and the levels of fasting blood glucose (FBG) and fasting insulin were determined. We estimated insulin resistance by the homeostasis model assessment (HOMA). The expression of inflammatory factors and F4/80 was examined by polymerase chain reaction (PCR) and western blotting to assess adipose tissues inflammation. We also determined the molecules of insulin signaling and the nuclear factor kappa B (NF-κB) pathway by western blotting. KEY FINDINGS The Nr2e1 expression was upregulated in WT obese mice when compared with that in control mice. Despite a lower body weight and epididymal fat mass in Nr2e1-/- mice, these rats showed increased inflammatory cytokines secretion, more pronounced hyperlipidemia, and impaired insulin sensitivity after HFD treatment. Further investigation revealed that Nr2e1 deletion affected the expression of insulin signaling and NF-κB pathway-related molecules in visceral adipose tissues. SIGNIFICANCE Nr2e1 may act as a potential target to improve insulin sensitivity and inflammation in obesity and related complications.
Collapse
Affiliation(s)
- Guangzhen He
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Jiaowei Gu
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Huawei Wang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Siyuan Cheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Qing Xiong
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Mengting Ke
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yong Hu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jieyuan Feng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Linyang Song
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zheng Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Wang YY, Hsu SH, Tsai HY, Cheng MC. Genetic analysis of the NR2E1 gene as a candidate gene of schizophrenia. Psychiatry Res 2020; 293:113386. [PMID: 32805587 DOI: 10.1016/j.psychres.2020.113386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/24/2020] [Accepted: 08/12/2020] [Indexed: 01/04/2023]
Abstract
NR2E1 is implicated in the regulation of neurogenesis and considered as a candidate gene for schizophrenia. We resequenced all the exons of NR2E1 in 547 patients with schizophrenia and 567 controls from Taiwan. We identified five common SNPs with no association with patients with schizophrenia. Further haplotype-based association analysis showed that two haplotypes within NR2E1 were correlated with the schizophrenia risk. Four rare mutations located at untranslated regions were identified in patients with schizophrenia but not in our control sample. The present study suggests that NR2E1 is likely to play a significant role in conferring susceptibility to schizophrenia.
Collapse
Affiliation(s)
- Yu-Yuan Wang
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, 981, Taiwan
| | - Shih-Hsin Hsu
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, 981, Taiwan
| | - Hsin-Yao Tsai
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, 981, Taiwan
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, 981, Taiwan.
| |
Collapse
|
7
|
Griffett K, Bedia-Diaz G, Hegazy L, de Vera IMS, Wanninayake US, Billon C, Koelblen T, Wilhelm ML, Burris TP. The Orphan Nuclear Receptor TLX Is a Receptor for Synthetic and Natural Retinoids. Cell Chem Biol 2020; 27:1272-1284.e4. [DOI: 10.1016/j.chembiol.2020.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/08/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
|
8
|
Bloch NI, Corral‐López A, Buechel SD, Kotrschal A, Kolm N, Mank JE. Different mating contexts lead to extensive rewiring of female brain coexpression networks in the guppy. GENES BRAIN AND BEHAVIOR 2020; 20:e12697. [DOI: 10.1111/gbb.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Natasha I. Bloch
- Department of Biomedical Engineering Universidad de Los Andes Bogotá D.C. Colombia
| | - Alberto Corral‐López
- Department of Zoology/Ethology Stockholm University Stockholm Sweden
- Department of Genetics, Evolution and Environment University College London UK
| | | | - Alexander Kotrschal
- Department of Zoology/Ethology Stockholm University Stockholm Sweden
- Wageningen University Behavioral Ecology Group Wageningen Netherlands
| | - Niclas Kolm
- Department of Zoology/Ethology Stockholm University Stockholm Sweden
| | - Judith E. Mank
- University of British Columbia Department of Zoology and Biodiversity Research Centre Vancouver Canada
- Department of Genetics, Evolution and Environment University College London UK
| |
Collapse
|
9
|
Parris TZ, Vizlin-Hodzic D, Salmela S, Funa K. Tumorigenic effects of TLX overexpression in HEK 293T cells. Cancer Rep (Hoboken) 2020; 2:e1204. [PMID: 32721119 DOI: 10.1002/cnr2.1204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The human orphan receptor TLX (NR2E1) is a key regulator of neurogenesis, adult stem cell maintenance, and tumorigenesis. However, little is known about the genetic and transcriptomic events that occur following TLX overexpression in human cell lines. AIMS Here, we used cytogenetics and RNA sequencing to investigate the effect of TLX overexpression with an inducible vector system in the HEK 293T cell line. METHODS AND RESULTS Conventional spectral karyotyping was used to identify chromosomal abnormalities, followed by fluorescence in situ hybridization (FISH) analysis on chromosome spreads to assess TLX DNA copy number. Illumina paired-end whole transcriptome sequencing was then performed to characterize recurrent genetic variants (single nucleotide polymorphisms (SNPs) and indels), expressed gene fusions, and gene expression profiles. Lastly, flow cytometry was used to analyze cell cycle distribution. Intriguingly, we show that upon transfection with a vector containing the human TLX gene (eGFP-hTLX), an isochromosome forms on the long arm of chromosome 6, thereby resulting in DNA gain of the TLX locus (6q21) and upregulation of TLX. Induction of the eGFP-hTLX vector further increased TLX expression levels, leading to G0-G1 cell cycle arrest, genetic aberrations, modulation of gene expression patterns, and crosstalk with other nuclear receptors (AR, ESR1, ESR2, NR1H4, and NR3C2). We identified a 49-gene signature associated with central nervous system (CNS) development and carcinogenesis, in addition to potentially cancer-driving gene fusions (LARP1-CNOT8 and NSL1-ZDBF2) and deleterious genetic variants (frameshift insertions in the CTSH, DBF4, POSTN, and WDR78 genes). CONCLUSION Taken together, these findings illustrate that TLX may play a pivotal role in tumorigenesis via genomic instability and perturbation of cancer-related processes.
Collapse
Affiliation(s)
- Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Dzeneta Vizlin-Hodzic
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Susanne Salmela
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Keiko Funa
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Kumari E, Velloso FJ, Nasuhidehnavi A, Somasundaram A, Savanur VH, Buono KD, Levison SW. Developmental IL-6 Exposure Favors Production of PDGF-Responsive Multipotential Progenitors at the Expense of Neural Stem Cells and Other Progenitors. Stem Cell Reports 2020; 14:861-875. [PMID: 32302560 PMCID: PMC7220986 DOI: 10.1016/j.stemcr.2020.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Interleukin-6 (IL-6) is increased in maternal serum and amniotic fluid of children subsequently diagnosed with autism spectrum disorders. However, it is not clear how increased IL-6 alters brain development. Here, we show that IL-6 increases the prevalence of a specific platelet-derived growth factor (PDGF)-responsive multipotent progenitor, with opposite effects on neural stem cells and on subsets of bipotential glial progenitors. Acutely, increasing circulating IL-6 levels 2-fold above baseline in neonatal mice specifically stimulated the proliferation of a PDGF-responsive multipotential progenitor accompanied by increased phosphorylated STAT3, increased Fbxo15 expression, and decreased Dnmt1 and Tlx expression. Fate mapping studies using a Nestin-CreERT2 driver revealed decreased astrogliogenesis in the frontal cortex. IL-6-treated mice were hyposmic; however, olfactory bulb neuronogenesis was unaffected. Altogether, these studies provide important insights into how inflammation alters neural stem cells and progenitors and provide new insights into the molecular and cellular underpinnings of neurodevelopmental disorders associated with maternal infections.
Collapse
Affiliation(s)
- Ekta Kumari
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA
| | - Fernando J Velloso
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA
| | - Azadeh Nasuhidehnavi
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA
| | - Aditya Somasundaram
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA
| | - Vibha H Savanur
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA
| | | | - Steven W Levison
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical Health Sciences of Rutgers University, 205 South Orange Ave. Newark, NJ 07103, USA.
| |
Collapse
|
11
|
A role for the orphan nuclear receptor TLX in the interaction between neural precursor cells and microglia. Neuronal Signal 2020; 3:NS20180177. [PMID: 32269832 PMCID: PMC7104222 DOI: 10.1042/ns20180177] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Microglia are an essential component of the neurogenic niche in the adult hippocampus and are involved in the control of neural precursor cell (NPC) proliferation, differentiation and the survival and integration of newborn neurons in hippocampal circuitry. Microglial and neuronal cross-talk is mediated in part by the chemokine fractalkine/chemokine (C-X3-C motif) ligand 1 (CX3CL1) released from neurons, and its receptor CX3C chemokine receptor 1 (CX3CR1) which is expressed on microglia. A disruption in this pathway has been associated with impaired neurogenesis yet the specific molecular mechanisms by which this interaction occurs remain unclear. The orphan nuclear receptor TLX (Nr2e1; homologue of the Drosophila tailless gene) is a key regulator of hippocampal neurogenesis, and we have shown that in its absence microglia exhibit a pro-inflammatory activation phenotype. However, it is unclear whether a disturbance in CX3CL1/CX3CR1 communication mediates an impairment in TLX-related pathways which may have subsequent effects on neurogenesis. To this end, we assessed miRNA expression of up- and down-stream signalling molecules of TLX in the hippocampus of mice lacking CX3CR1. Our results demonstrate that a lack of CX3CR1 is associated with altered expression of TLX and its downstream targets in the hippocampus without significantly affecting upstream regulators of TLX. Thus, TLX may be a potential participant in neural stem cell (NSC)-microglial cross-talk and may be an important target in understanding inflammatory-associated impairments in neurogenesis.
Collapse
|
12
|
Cárdenas-Tueme M, Montalvo-Martínez L, Maldonado-Ruiz R, Camacho-Morales A, Reséndez-Pérez D. Neurodegenerative Susceptibility During Maternal Nutritional Programing: Are Central and Peripheral Innate Immune Training Relevant? Front Neurosci 2020; 14:13. [PMID: 32116490 PMCID: PMC7010854 DOI: 10.3389/fnins.2020.00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/08/2020] [Indexed: 12/24/2022] Open
Abstract
Maternal overnutrition modulates body weight, development of metabolic failure and, potentially, neurodegenerative susceptibility in the offspring. Overnutrition sets a chronic pro-inflammatory profile that integrates peripheral and central immune activation nodes, damaging neuronal physiology and survival. Innate immune cells exposed to hypercaloric diets might experience trained immunity. Here, we address the role of maternal overnutrition as a trigger for central and peripheral immune training and its contribution to neurodegeneration and the molecular nodes implicated in the Nod-like receptor protein 3 (NLRP3) inflammasome pathway leading to immune training. We propose that maternal overnutrition leads to peripheral or central immune training that favor neurodegenerative susceptibility in the offspring.
Collapse
Affiliation(s)
- Marcela Cárdenas-Tueme
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Larisa Montalvo-Martínez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Roger Maldonado-Ruiz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Centro de Investigación y Desarrollo en Ciencias de la Salud, Unidad de Neurometabolismo, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Diana Reséndez-Pérez
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| |
Collapse
|
13
|
Xiong Q, Wu Y, Yang M, Wu G, Wang Y, Wang H, Feng J, Song L, Tong B, He G, Xu Y. Nr2e1 ablation impairs liver glucolipid metabolism and induces inflammation, high-fat diets amplify the damage. Biomed Pharmacother 2019; 120:109503. [DOI: 10.1016/j.biopha.2019.109503] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023] Open
|
14
|
Chistiakov DA, Chekhonin VP. Early-life adversity-induced long-term epigenetic programming associated with early onset of chronic physical aggression: Studies in humans and animals. World J Biol Psychiatry 2019; 20:258-277. [PMID: 28441915 DOI: 10.1080/15622975.2017.1322714] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objectives: To examine whether chronic physical aggression (CPA) in adulthood can be epigenetically programmed early in life due to exposure to early-life adversity. Methods: Literature search of public databases such as PubMed/MEDLINE and Scopus. Results: Children/adolescents susceptible for CPA and exposed to early-life abuse fail to efficiently cope with stress that in turn results in the development of CPA later in life. This phenomenon was observed in humans and animal models of aggression. The susceptibility to aggression is a complex trait that is regulated by the interaction between environmental and genetic factors. Epigenetic mechanisms mediate this interaction. Subjects exposed to stress early in life exhibited long-term epigenetic programming that can influence their behaviour in adulthood. This programming affects expression of many genes not only in the brain but also in other systems such as neuroendocrine and immune. Conclusions: The propensity to adult CPA behaviour in subjects experienced to early-life adversity is mediated by epigenetic programming that involves long-term systemic epigenetic alterations in a whole genome.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Vladimir P Chekhonin
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia.,b Department of Medical Nanobiotechnology , Pirogov Russian State Medical University (RSMU) , Moscow , Russia
| |
Collapse
|
15
|
Twenty-Seven Tamoxifen-Inducible iCre-Driver Mouse Strains for Eye and Brain, Including Seventeen Carrying a New Inducible-First Constitutive-Ready Allele. Genetics 2019; 211:1155-1177. [PMID: 30765420 PMCID: PMC6456315 DOI: 10.1534/genetics.119.301984] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
To understand gene function, the cre/loxP conditional system is the most powerful available for temporal and spatial control of expression in mouse. However, the research community requires more cre recombinase expressing transgenic mouse strains (cre-drivers) that restrict expression to specific cell types. To address these problems, a high-throughput method for large-scale production that produces high-quality results is necessary. Further, endogenous promoters need to be chosen that drive cell type specific expression, or we need to further focus the expression by manipulating the promoter. Here we test the suitability of using knock-ins at the docking site 5′ of Hprt for rapid development of numerous cre-driver strains focused on expression in adulthood, using an improved cre tamoxifen inducible allele (icre/ERT2), and testing a novel inducible-first, constitutive-ready allele (icre/f3/ERT2/f3). In addition, we test two types of promoters either to capture an endogenous expression pattern (MaxiPromoters), or to restrict expression further using minimal promoter element(s) designed for expression in restricted cell types (MiniPromoters). We provide new cre-driver mouse strains with applicability for brain and eye research. In addition, we demonstrate the feasibility and applicability of using the locus 5′ of Hprt for the rapid generation of substantial numbers of cre-driver strains. We also provide a new inducible-first constitutive-ready allele to further speed cre-driver generation. Finally, all these strains are available to the research community through The Jackson Laboratory.
Collapse
|
16
|
Kozareva DA, Foley T, Moloney GM, Cryan JF, Nolan YM. TLX knockdown in the dorsal dentate gyrus of juvenile rats differentially affects adolescent and adult behaviour. Behav Brain Res 2018; 360:36-50. [PMID: 30481511 DOI: 10.1016/j.bbr.2018.11.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 12/27/2022]
Abstract
The orphan nuclear receptor TLX is predominantly expressed in the central nervous system and is an important factor regulating the maintenance and self-renewal of neural stem cells from embryonic development through adulthood. In adolescence and adulthood, TLX expression is restricted to the neurogenic niches of the brain: the dentate gyrus of the hippocampus and the subventricular zone. The adolescent period is critical for maturation of the hippocampus with heightened levels of neurogenesis observed in rodents. Therefore, we investigated whether lentiviral silencing of TLX expression (TLX knockdown) in the dorsal dentate gyrus of juvenile rats incurred differential impairments in behaviour during late adolescence and adulthood. Our results showed that knockdown of TLX in the dorsal dentate gyrus led to a decrease in cell proliferation in the dorsal but not ventral dentate gyrus. At a behavioural level we observed differential effects in adolescence and adulthood across a number of parameters. A hyperactive phenotype was present in adolescent but not adult TLX knockdown rats, and an increase in immobility during adolescence and in swimming frequency during adulthood was observed in the forced swim test. There was an increased defecation frequency in the open field during adulthood but not adolescence. There were no changes in cognitive performance on hippocampus-dependent tasks or in anxiety-related behaviours. In conclusion, silencing of TLX in the dorsal dentate gyrus led to impairments in hippocampal-independent behaviours which either did not persist or were reversed during adulthood. The current data highlight the temporal importance and function of the nuclear receptor TLX during development.
Collapse
Affiliation(s)
- Danka A Kozareva
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Tara Foley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
17
|
McKenzie CW, Preston CC, Finn R, Eyster KM, Faustino RS, Lee L. Strain-specific differences in brain gene expression in a hydrocephalic mouse model with motile cilia dysfunction. Sci Rep 2018; 8:13370. [PMID: 30190587 PMCID: PMC6127338 DOI: 10.1038/s41598-018-31743-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/22/2018] [Indexed: 01/10/2023] Open
Abstract
Congenital hydrocephalus results from cerebrospinal fluid accumulation in the ventricles of the brain and causes severe neurological damage, but the underlying causes are not well understood. It is associated with several syndromes, including primary ciliary dyskinesia (PCD), which is caused by dysfunction of motile cilia. We previously demonstrated that mouse models of PCD lacking ciliary proteins CFAP221, CFAP54 and SPEF2 all have hydrocephalus with a strain-dependent severity. While morphological defects are more severe on the C57BL/6J (B6) background than 129S6/SvEvTac (129), cerebrospinal fluid flow is perturbed on both backgrounds, suggesting that abnormal cilia-driven flow is not the only factor underlying the hydrocephalus phenotype. Here, we performed a microarray analysis on brains from wild type and nm1054 mice lacking CFAP221 on the B6 and 129 backgrounds. Expression differences were observed for a number of genes that cluster into distinct groups based on expression pattern and biological function, many of them implicated in cellular and biochemical processes essential for proper brain development. These include genes known to be functionally relevant to congenital hydrocephalus, as well as formation and function of both motile and sensory cilia. Identification of these genes provides important clues to mechanisms underlying congenital hydrocephalus severity.
Collapse
Affiliation(s)
- Casey W McKenzie
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Rozzy Finn
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Kathleen M Eyster
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, 57069, USA
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA
| | - Lance Lee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA. .,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA.
| |
Collapse
|
18
|
Ó'Léime CS, Hoban AE, Hueston CM, Stilling R, Moloney G, Cryan JF, Nolan YM. The orphan nuclear receptor TLX regulates hippocampal transcriptome changes induced by IL-1β. Brain Behav Immun 2018. [PMID: 29518529 DOI: 10.1016/j.bbi.2018.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
TLX is an orphan nuclear receptor highly expressed within neural progenitor cells (NPCs) in the hippocampus where is regulates proliferation. Inflammation has been shown to have negative effects on hippocampal function as well as on NPC proliferation. Specifically, the pro-inflammatory cytokine IL-1β suppresses NPC proliferation as well as TLX expression in the hippocampus. However, it is unknown whether TLX itself is involved in regulating the inflammatory response in the hippocampus. To explore the role of TLX in inflammation, we assessed changes in the transcriptional landscape of the hippocampus of TLX knockout mice (TLX-/-) compared to wildtype (WT) littermate controls with and without intrahippocampal injection of IL-1β using a whole transcriptome RNA sequencing approach. We demonstrated that there is an increase in the transcription of genes involved in the promotion of inflammation and regulation of cell chemotaxis (Tnf, Il1b, Cxcr1, Cxcr2, Tlr4) and a decrease in the expression of genes relating to synaptic signalling (Lypd1, Syt4, Cplx2) in cannulated TLX-/- mice compared to WT controls. We demonstrate that mice lacking in TLX share a similar increase in 176 genes involved in regulating inflammation (e.g. Cxcl1, Tnf, Il1b) as WT mice injected with IL-1β into the hippocampus. Moreover, TLX-/- mice injected with IL-1β displayed a blunted transcriptional profile compared to WT mice injected with IL-1β. Thus, TLX-/- mice, which already have an exaggerated inflammatory profile after cannulation surgery, are primed to respond differently to an inflammatory stimulus such as IL-1β. Together, these results demonstrate that TLX regulates hippocampal inflammatory transcriptome response to brain injury (in this case cannulation surgery) and cytokine stimulation.
Collapse
Affiliation(s)
- Ciarán S Ó'Léime
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Alan E Hoban
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Cara M Hueston
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Roman Stilling
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Gerard Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland.
| |
Collapse
|
19
|
O'Léime CS, Cryan JF, Nolan YM. Nuclear deterrents: Intrinsic regulators of IL-1β-induced effects on hippocampal neurogenesis. Brain Behav Immun 2017; 66:394-412. [PMID: 28751020 DOI: 10.1016/j.bbi.2017.07.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/15/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
Hippocampal neurogenesis, the process by which new neurons are born and develop into the host circuitry, begins during embryonic development and persists throughout adulthood. Over the last decade considerable insights have been made into the role of hippocampal neurogenesis in cognitive function and the cellular mechanisms behind this process. Additionally, an increasing amount of evidence exists on the impact of environmental factors, such as stress and neuroinflammation on hippocampal neurogenesis and subsequent impairments in cognition. Elevated expression of the pro-inflammatory cytokine interleukin-1β (IL-1β) in the hippocampus is established as a significant contributor to the neuronal demise evident in many neurological and psychiatric disorders and is now known to negatively regulate hippocampal neurogenesis. In order to prevent the deleterious effects of IL-1β on neurogenesis it is necessary to identify signalling pathways and regulators of neurogenesis within neural progenitor cells that can interact with IL-1β. Nuclear receptors are ligand regulated transcription factors that are involved in modulating a large number of cellular processes including neurogenesis. In this review we focus on the signalling mechanisms of specific nuclear receptors involved in regulating neurogenesis (glucocorticoid receptors, peroxisome proliferator activated receptors, estrogen receptors, and nuclear receptor subfamily 2 group E member 1 (NR2E1 or TLX)). We propose that these nuclear receptors could be targeted to inhibit neuroinflammatory signalling pathways associated with IL-1β. We discuss their potential to be therapeutic targets for neuroinflammatory disorders affecting hippocampal neurogenesis and associated cognitive function.
Collapse
Affiliation(s)
- Ciarán S O'Léime
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
20
|
Kozareva DA, O'Leary OF, Cryan JF, Nolan YM. Deletion of TLX and social isolation impairs exercise-induced neurogenesis in the adolescent hippocampus. Hippocampus 2017; 28:3-11. [DOI: 10.1002/hipo.22805] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 09/07/2017] [Accepted: 09/20/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Danka A. Kozareva
- Department of Anatomy and Neuroscience; University College Cork; Ireland
- APC Microbiome Institute; University College Cork; Ireland
| | - Olivia F. O'Leary
- Department of Anatomy and Neuroscience; University College Cork; Ireland
- APC Microbiome Institute; University College Cork; Ireland
| | - John F. Cryan
- Department of Anatomy and Neuroscience; University College Cork; Ireland
- APC Microbiome Institute; University College Cork; Ireland
| | - Yvonne M. Nolan
- Department of Anatomy and Neuroscience; University College Cork; Ireland
- APC Microbiome Institute; University College Cork; Ireland
| |
Collapse
|
21
|
Maheu ME, Ressler KJ. Developmental pathway genes and neural plasticity underlying emotional learning and stress-related disorders. Learn Mem 2017; 24:492-501. [PMID: 28814475 PMCID: PMC5580529 DOI: 10.1101/lm.044271.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/18/2017] [Indexed: 11/24/2022]
Abstract
The manipulation of neural plasticity as a means of intervening in the onset and progression of stress-related disorders retains its appeal for many researchers, despite our limited success in translating such interventions from the laboratory to the clinic. Given the challenges of identifying individual genetic variants that confer increased risk for illnesses like depression and post-traumatic stress disorder, some have turned their attention instead to focusing on so-called "master regulators" of plasticity that may provide a means of controlling these potentially impaired processes in psychiatric illnesses. The mammalian homolog of Tailless (TLX), Wnt, and the homeoprotein Otx2 have all been proposed to constitute master regulators of different forms of plasticity which have, in turn, each been implicated in learning and stress-related disorders. In the present review, we provide an overview of the changing distribution of these genes and their roles both during development and in the adult brain. We further discuss how their distinct expression profiles provide clues as to their function, and may inform their suitability as candidate drug targets in the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Marissa E Maheu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts 02478, USA
| | - Kerry J Ressler
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts 02478, USA
| |
Collapse
|
22
|
Kozareva DA, Hueston CM, Ó'Léime CS, Crotty S, Dockery P, Cryan JF, Nolan YM. Absence of the neurogenesis-dependent nuclear receptor TLX induces inflammation in the hippocampus. J Neuroimmunol 2017; 331:87-96. [PMID: 28844503 DOI: 10.1016/j.jneuroim.2017.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 12/25/2022]
Abstract
The orphan nuclear receptor TLX (Nr2e1) is a key regulator of hippocampal neurogenesis. Impaired adult hippocampal neurogenesis has been reported in neurodegenerative and psychiatric conditions including dementia and stress-related depression. Neuroinflammation is also implicated in the neuropathology of these disorders, and has been shown to negatively affect hippocampal neurogenesis. To investigate a role for TLX in hippocampal neuroinflammation, we assessed microglial activation in the hippocampus of mice with a spontaneous deletion of TLX. Results from our study suggest that a lack of TLX is implicated in deregulation of microglial phenotype and that consequently, the survival and function of newborn cells in the hippocampus is impaired. TLX may be an important target in understanding inflammatory-associated impairments in neurogenesis.
Collapse
Affiliation(s)
- Danka A Kozareva
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Cara M Hueston
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Ciarán S Ó'Léime
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Suzanne Crotty
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Peter Dockery
- Department of Anatomy, National University of Ireland, Galway, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
23
|
Whole-genome DNA methylation status associated with clinical PTSD measures of OIF/OEF veterans. Transl Psychiatry 2017; 7:e1169. [PMID: 28696412 PMCID: PMC5538114 DOI: 10.1038/tp.2017.129] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
Emerging knowledge suggests that post-traumatic stress disorder (PTSD) pathophysiology is linked to the patients' epigenetic changes, but comprehensive studies examining genome-wide methylation have not been performed. In this study, we examined genome-wide DNA methylation in peripheral whole blood in combat veterans with and without PTSD to ascertain differentially methylated probes. Discovery was initially made in a training sample comprising 48 male Operation Enduring Freedom (OEF)/Operation Iraqi Freedom (OIF) veterans with PTSD and 51 age/ethnicity/gender-matched combat-exposed PTSD-negative controls. Agilent whole-genome array detected ~5600 differentially methylated CpG islands (CpGI) annotated to ~2800 differently methylated genes (DMGs). The majority (84.5%) of these CpGIs were hypermethylated in the PTSD cases. Functional analysis was performed using the DMGs encoding the promoter-bound CpGIs to identify networks related to PTSD. The identified networks were further validated by an independent test set comprising 31 PTSD+/29 PTSD- veterans. Targeted bisulfite sequencing was also used to confirm the methylation status of 20 DMGs shown to be highly perturbed in the training set. To improve the statistical power and mitigate the assay bias and batch effects, a union set combining both training and test set was assayed using a different platform from Illumina. The pathways curated from this analysis confirmed 65% of the pool of pathways mined from training and test sets. The results highlight the importance of assay methodology and use of independent samples for discovery and validation of differentially methylated genes mined from whole blood. Nonetheless, the current study demonstrates that several important epigenetically altered networks may distinguish combat-exposed veterans with and without PTSD.
Collapse
|
24
|
Fulton J, Mazumder B, Whitchurch JB, Monteiro CJ, Collins HM, Chan CM, Clemente MP, Hernandez-Quiles M, Stewart EA, Amoaku WM, Moran PM, Mongan NP, Persson JL, Ali S, Heery DM. Heterodimers of photoreceptor-specific nuclear receptor (PNR/NR2E3) and peroxisome proliferator-activated receptor-γ (PPARγ) are disrupted by retinal disease-associated mutations. Cell Death Dis 2017; 8:e2677. [PMID: 28300834 PMCID: PMC5386588 DOI: 10.1038/cddis.2017.98] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 12/30/2022]
Abstract
Photoreceptor-specific nuclear receptor (PNR/NR2E3) and Tailless homolog (TLX/NR2E1) are human orthologs of the NR2E group, a subgroup of phylogenetically related members of the nuclear receptor (NR) superfamily of transcription factors. We assessed the ability of these NRs to form heterodimers with other members of the human NRs representing all major subgroups. The TLX ligand-binding domain (LBD) did not appear to form homodimers or interact directly with any other NR tested. The PNR LBD was able to form homodimers, but also exhibited robust interactions with the LBDs of peroxisome proliferator-activated receptor-γ (PPARγ)/NR1C3 and thyroid hormone receptor b (TRb) TRβ/NR1A2. The binding of PNR to PPARγ was specific for this paralog, as no interaction was observed with the LBDs of PPARα/NR1C1 or PPARδ/NR1C2. In support of these findings, PPARγ and PNR were found to be co-expressed in human retinal tissue extracts and could be co-immunoprecipitated as a native complex. Selected sequence variants in the PNR LBD associated with human retinopathies, or a mutation in the dimerization region of PPARγ LBD associated with familial partial lipodystrophy type 3, were found to disrupt PNR/PPARγ complex formation. Wild-type PNR, but not a PNR309G mutant, was able to repress PPARγ-mediated transcription in reporter assays. In summary, our results reveal novel heterodimer interactions in the NR superfamily, suggesting previously unknown functional interactions of PNR with PPARγ and TRβ that have potential importance in retinal development and disease.
Collapse
Affiliation(s)
- Joel Fulton
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Bismoy Mazumder
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | | | | | - Chun M Chan
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | | | - Elizabeth A Stewart
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| | - Winfried M Amoaku
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| | - Paula M Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| | - Nigel P Mongan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Jenny L Persson
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - David M Heery
- School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
25
|
Putative transmembrane transporter modulates higher-level aggression in Drosophila. Proc Natl Acad Sci U S A 2017; 114:2373-2378. [PMID: 28193893 DOI: 10.1073/pnas.1618354114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
By selection of winners of dyadic fights for 35 generations, we have generated a hyperaggressive Bully line of flies that almost always win fights against the parental wild-type Canton-S stock. Maintenance of the Bully phenotype is temperature dependent during development, with the phenotype lost when flies are reared at 19 °C. No similar effect is seen with the parent line. This difference allowed us to carry out RNA-seq experiments and identify a limited number of genes that are differentially expressed by twofold or greater in the Bullies; one of these was a putative transmembrane transporter, CG13646, which showed consistent and reproducible twofold down-regulation in Bullies. We examined the causal effect of this gene on the phenotype with a mutant line for CG13646, and with an RNAi approach. In all cases, reduction in expression of CG13646 by approximately half led to a hyperaggressive phenotype partially resembling that seen in the Bully flies. This gene is a member of a very interesting family of solute carrier proteins (SLCs), some of which have been suggested as being involved in glutamine/glutamate and GABA cycles of metabolism in excitatory and inhibitory nerve terminals in mammalian systems.
Collapse
|
26
|
|
27
|
O'Leary JD, O'Leary OF, Cryan JF, Nolan YM. Regulation of behaviour by the nuclear receptor TLX. GENES BRAIN AND BEHAVIOR 2016; 17:e12357. [PMID: 27790850 DOI: 10.1111/gbb.12357] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/15/2016] [Accepted: 10/25/2016] [Indexed: 01/10/2023]
Abstract
The orphan nuclear receptor Tlx (Nr2e1) is a key regulator of both embryonic and adult hippocampal neurogenesis. Several different mouse models have been developed which target Tlx in vivo including spontaneous deletion models (from birth) and targeted and conditional knockouts. Although some conflicting findings have been reported, for the most part studies have demonstrated that Tlx is important in regulating processes that underlie neurogenesis, spatial learning, anxiety-like behaviour and interestingly, aggression. More recent data have demonstrated that disrupting Tlx during early life induces hyperactivity and that Tlx plays a role in emotional regulation. Moreover, there are sex- and age-related differences in some behaviours in Tlx knockout mice during adolescence and adulthood. Here, we discuss the role of Tlx in motor-, cognitive-, aggressive- and anxiety-related behaviours during adolescence and adulthood. We examine current evidence which provides insight into Tlx during neurodevelopment, and offer our thoughts on the function of Tlx in brain and behaviour. We further hypothesize that Tlx is a key target in understanding the emergence of neurobiological disorders during adolescence and early adulthood.
Collapse
Affiliation(s)
- J D O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - O F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - J F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Y M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
28
|
Corso-Díaz X, de Leeuw CN, Alonso V, Melchers D, Wong BKY, Houtman R, Simpson EM. Co-activator candidate interactions for orphan nuclear receptor NR2E1. BMC Genomics 2016; 17:832. [PMID: 27782803 PMCID: PMC5080790 DOI: 10.1186/s12864-016-3173-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 10/18/2016] [Indexed: 12/21/2022] Open
Abstract
Background NR2E1 (Tlx) is an orphan nuclear receptor that regulates the maintenance and self-renewal of neural stem cells, and promotes tumourigenesis. Nr2e1-null mice exhibit reduced cortical and limbic structures and pronounced retinal dystrophy. NR2E1 functions mainly as a repressor of gene transcription in association with the co-repressors atrophin-1, LSD1, HDAC and BCL11A. Recent evidence suggests that NR2E1 also acts as an activator of gene transcription. However, co-activator complexes that interact with NR2E1 have not yet been identified. In order to find potential novel co-regulators for NR2E1, we used a microarray assay for real-time analysis of co-regulator–nuclear receptor interaction (MARCoNI) that contains peptides representing interaction motifs from potential co-regulatory proteins, including known co-activator nuclear receptor box sequences (LxxLL motif). Results We found that NR2E1 binds strongly to an atrophin-1 peptide (Atro box) used as positive control and to 19 other peptides that constitute candidate NR2E1 partners. Two of these proteins, p300 and androgen receptor (AR), were further validated by reciprocal pull-down assays. The specificity of NR2E1 binding to peptides in the array was evaluated using two single amino acid variants, R274G and R276Q, which disrupted the majority of the binding interactions observed with wild-type NR2E1. The decreased binding affinity of these variants to co-regulators was further validated by pull-down assays using atrophin1 as bait. Despite the high conservation of arginine 274 in vertebrates, its reduced interactions with co-regulators were not significant in vivo as determined by retinal phenotype analysis in single-copy Nr2e1-null mice carrying the variant R274G. Conclusions We showed that MARCoNI is a specific assay to test interactions of NR2E1 with candidate co-regulators. In this way, we unveiled 19 potential co-regulator partners for NR2E1, including eight co-activators. All the candidates here identified need to be further validated using in vitro and in vivo models. This assay was sensitive to point mutations in NR2E1 ligand binding domain making it useful to identify mutations and/or small molecules that alter binding of NR2E1 to protein partners. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3173-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ximena Corso-Díaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
| | - Charles N de Leeuw
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Vivian Alonso
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | | | - Bibiana K Y Wong
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - René Houtman
- PamGene International B.V., Den Bosch, The Netherlands
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, BC, V6T 2A1, Canada. .,Department of Ophthalmology and Visual Science, University of British Columbia, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
29
|
Chen L, Chu C, Zhang YH, Zhu C, Kong X, Huang T, Cai YD. Analysis of Gene Expression Profiles in the Human Brain Stem, Cerebellum and Cerebral Cortex. PLoS One 2016; 11:e0159395. [PMID: 27434030 PMCID: PMC4951119 DOI: 10.1371/journal.pone.0159395] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 07/01/2016] [Indexed: 11/19/2022] Open
Abstract
The human brain is one of the most mysterious tissues in the body. Our knowledge of the human brain is limited due to the complexity of its structure and the microscopic nature of connections between brain regions and other tissues in the body. In this study, we analyzed the gene expression profiles of three brain regions-the brain stem, cerebellum and cerebral cortex-to identify genes that are differentially expressed among these different brain regions in humans and to obtain a list of robust, region-specific, differentially expressed genes by comparing the expression signatures from different individuals. Feature selection methods, specifically minimum redundancy maximum relevance and incremental feature selection, were employed to analyze the gene expression profiles. Sequential minimal optimization, a machine-learning algorithm, was employed to examine the utility of selected genes. We also performed a literature search, and we discuss the experimental evidence for the important physiological functions of several highly ranked genes, including NR2E1, DAO, and LRRC7, and we give our analyses on a gene (TFAP2B) that have not been investigated or experimentally validated. As a whole, the results of our study will improve our ability to predict and understand genes related to brain regionalization and function.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Chen Chu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Changming Zhu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- * E-mail: (YDC); (TH)
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- * E-mail: (YDC); (TH)
| |
Collapse
|
30
|
Freudenberg F, Carreño Gutierrez H, Post AM, Reif A, Norton WHJ. Aggression in non-human vertebrates: Genetic mechanisms and molecular pathways. Am J Med Genet B Neuropsychiatr Genet 2016; 171:603-40. [PMID: 26284957 DOI: 10.1002/ajmg.b.32358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/28/2015] [Indexed: 11/07/2022]
Abstract
Aggression is an adaptive behavioral trait that is important for the establishment of social hierarchies and competition for mating partners, food, and territories. While a certain level of aggression can be beneficial for the survival of an individual or species, abnormal aggression levels can be detrimental. Abnormal aggression is commonly found in human patients with psychiatric disorders. The predisposition to aggression is influenced by a combination of environmental and genetic factors and a large number of genes have been associated with aggression in both human and animal studies. In this review, we compare and contrast aggression studies in zebrafish and mouse. We present gene ontology and pathway analyses of genes linked to aggression and discuss the molecular pathways that underpin agonistic behavior in these species. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | | | - Antonia M Post
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| |
Collapse
|
31
|
The nuclear receptor Tlx regulates motor, cognitive and anxiety-related behaviours during adolescence and adulthood. Behav Brain Res 2016; 306:36-47. [DOI: 10.1016/j.bbr.2016.03.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 11/23/2022]
|
32
|
The TLX-miR-219 cascade regulates neural stem cell proliferation in neurodevelopment and schizophrenia iPSC model. Nat Commun 2016; 7:10965. [PMID: 26965827 PMCID: PMC4793043 DOI: 10.1038/ncomms10965] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/05/2016] [Indexed: 01/03/2023] Open
Abstract
Dysregulated expression of miR-219, a brain-specific microRNA, has been observed in neurodevelopmental disorders, such as schizophrenia (SCZ). However, its role in normal mammalian neural stem cells (NSCs) and in SCZ pathogenesis remains unknown. We show here that the nuclear receptor TLX, an essential regulator of NSC proliferation and self-renewal, inhibits miR-219 processing. miR-219 suppresses mouse NSC proliferation downstream of TLX. Moreover, we demonstrate upregulation of miR-219 and downregulation of TLX expression in NSCs derived from SCZ patient iPSCs and DISC1-mutant isogenic iPSCs. SCZ NSCs exhibit reduced cell proliferation. Overexpression of TLX or inhibition of miR-219 action rescues the proliferative defect in SCZ NSCs. Therefore, this study uncovers an important role for TLX and miR-219 in both normal neurodevelopment and in SCZ patient iPSC-derived NSCs. Moreover, this study reveals an unexpected role for TLX in regulating microRNA processing, independent of its well-characterized role in transcriptional regulation. Dysregulation of microRNAs has been implicated in neurodevelopmental disorders, including schizophrenia. Here the authors show that the TLX-miR-219 cascade regulates the proliferation of neural stem cells during normal development, and this pathway is dysregulated in a schizophrenia iPSC model.
Collapse
|
33
|
Davis SM, Thomas AL, Nomie KJ, Huang L, Dierick HA. Tailless and Atrophin control Drosophila aggression by regulating neuropeptide signalling in the pars intercerebralis. Nat Commun 2016; 5:3177. [PMID: 24495972 DOI: 10.1038/ncomms4177] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/23/2013] [Indexed: 01/21/2023] Open
Abstract
Aggressive behaviour is widespread throughout the animal kingdom. However, its mechanisms are poorly understood, and the degree of molecular conservation between distantly related species is unknown. Here we show that knockdown of tailless (tll) increases aggression in Drosophila, similar to the effect of its mouse orthologue Nr2e1. Tll localizes to the adult pars intercerebralis (PI), which shows similarity to the mammalian hypothalamus. Knockdown of tll in the PI is sufficient to increase aggression and is rescued by co-expressing human NR2E1. Knockdown of Atrophin, a Tll co-repressor, also increases aggression, and both proteins physically interact in the PI. tll knockdown-induced aggression is fully suppressed by blocking neuropeptide processing or release from the PI. In addition, genetically activating PI neurons increases aggression, mimicking the aggression-inducing effect of hypothalamic stimulation. Together, our results suggest that a transcriptional control module regulates neuropeptide signalling from the neurosecretory cells of the brain to control aggressive behaviour.
Collapse
Affiliation(s)
- Shaun M Davis
- 1] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA [2]
| | - Amanda L Thomas
- 1] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA [2]
| | - Krystle J Nomie
- 1] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA [2]
| | - Longwen Huang
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Herman A Dierick
- 1] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA [2] Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA [3] Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA [4] Program in Developmental Biology, Houston, Texas 77030, USA
| |
Collapse
|
34
|
Wang T, Xiong JQ. The Orphan Nuclear Receptor TLX/NR2E1 in Neural Stem Cells and Diseases. Neurosci Bull 2016; 32:108-14. [PMID: 26769490 DOI: 10.1007/s12264-015-0004-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/26/2015] [Indexed: 12/24/2022] Open
Abstract
The human TLX gene encodes an orphan nuclear receptor predominantly expressed in the central nervous system. Tailess and Tlx, the TLX homologues in Drosophila and mouse, play essential roles in body-pattern formation and neurogenesis during early embryogenesis and perform crucial functions in maintaining stemness and controlling the differentiation of adult neural stem cells in the central nervous system, especially the visual system. Multiple target genes and signaling pathways are regulated by TLX and its homologues in specific tissues during various developmental stages. This review aims to summarize previous studies including many recent updates from different aspects concerning TLX and its homologues in Drosophila and mouse.
Collapse
Affiliation(s)
- Tao Wang
- Department of Intensive Care, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jian-Qiong Xiong
- Department of Intensive Care, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
35
|
Shi X, Xiong X, Dai Z, Deng H, Sun L, Hu X, Zhou F, Xu Y. Nuclear orphan receptor TLX affects gene expression, proliferation and cell apoptosis in beta cells. Biochem Biophys Res Commun 2015; 468:387-93. [DOI: 10.1016/j.bbrc.2015.10.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
|
36
|
Nr2e1 Deficiency Augments Palmitate-Induced Oxidative Stress in Beta Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:9648769. [PMID: 26649147 PMCID: PMC4663339 DOI: 10.1155/2016/9648769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/12/2015] [Accepted: 07/15/2015] [Indexed: 11/19/2022]
Abstract
Nuclear receptor subfamily 2 group E member 1 (Nr2e1) has been regarded as an essential regulator of the growth of neural stem cells. However, its function elsewhere is unknown. In the present study, we generated Nr2e1 knockdown MIN6 cells and studied whether Nr2e1 knockdown affected basal beta cell functions such as proliferation, cell death, and insulin secretion. We showed that knockdown of Nr2e1 in MIN6 cells resulted in increased sensitivity to lipotoxicity, decreased proliferation, a partial G0/G1 cell-cycle arrest, and higher rates of apoptosis. Moreover, Nr2e1 deficiency exaggerates palmitate-induced impairment in insulin secretion. At the molecular level, Nr2e1 deficiency augments palmitate-induced oxidative stress. Nr2e1 deficiency also resulted in decreases in antioxidant enzymes and expression level of Nrf2. Together, this study indicated a potential protective effect of Nr2e1 on beta cells, which may serve as a target for the development of novel therapies for diabetes.
Collapse
|
37
|
Thomas AL, Davis SM, Dierick HA. Of Fighting Flies, Mice, and Men: Are Some of the Molecular and Neuronal Mechanisms of Aggression Universal in the Animal Kingdom? PLoS Genet 2015; 11:e1005416. [PMID: 26312756 PMCID: PMC4551476 DOI: 10.1371/journal.pgen.1005416] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aggressive behavior is widespread in the animal kingdom, but the degree of molecular conservation between distantly related species is still unclear. Recent reports suggest that at least some of the molecular mechanisms underlying this complex behavior in flies show remarkable similarities with such mechanisms in mice and even humans. Surprisingly, some aspects of neuronal control of aggression also show remarkable similarity between these distantly related species. We will review these recent findings, address the evolutionary implications, and discuss the potential impact for our understanding of human diseases characterized by excessive aggression.
Collapse
Affiliation(s)
- Amanda L. Thomas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shaun M. Davis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Herman A. Dierick
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Kudryavtseva NN, Markel AL, Orlov YL. Aggressive behavior: Genetic and physiological mechanisms. ACTA ACUST UNITED AC 2015. [DOI: 10.1134/s2079059715040085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
39
|
Schmouth JF, Arenillas D, Corso-Díaz X, Xie YY, Bohacec S, Banks KG, Bonaguro RJ, Wong SH, Jones SJM, Marra MA, Simpson EM, Wasserman WW. Combined serial analysis of gene expression and transcription factor binding site prediction identifies novel-candidate-target genes of Nr2e1 in neocortex development. BMC Genomics 2015. [PMID: 26204903 PMCID: PMC4512088 DOI: 10.1186/s12864-015-1770-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Nr2e1 (nuclear receptor subfamily 2, group e, member 1) encodes a transcription factor important in neocortex development. Previous work has shown that nuclear receptors can have hundreds of target genes, and bind more than 300 co-interacting proteins. However, recognition of the critical role of Nr2e1 in neural stem cells and neocortex development is relatively recent, thus the molecular mechanisms involved for this nuclear receptor are only beginning to be understood. Serial analysis of gene expression (SAGE), has given researchers both qualitative and quantitative information pertaining to biological processes. Thus, in this work, six LongSAGE mouse libraries were generated from laser microdissected tissue samples of dorsal VZ/SVZ (ventricular zone and subventricular zone) from the telencephalon of wild-type (Wt) and Nr2e1-null embryos at the critical development ages E13.5, E15.5, and E17.5. We then used a novel approach, implementing multiple computational methods followed by biological validation to further our understanding of Nr2e1 in neocortex development. Results In this work, we have generated a list of 1279 genes that are differentially expressed in response to altered Nr2e1 expression during in vivo neocortex development. We have refined this list to 64 candidate direct-targets of NR2E1. Our data suggested distinct roles for Nr2e1 during different neocortex developmental stages. Most importantly, our results suggest a possible novel pathway by which Nr2e1 regulates neurogenesis, which includes Lhx2 as one of the candidate direct-target genes, and SOX9 as a co-interactor. Conclusions In conclusion, we have provided new candidate interacting partners and numerous well-developed testable hypotheses for understanding the pathways by which Nr2e1 functions to regulate neocortex development. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1770-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean-François Schmouth
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Current address: Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, H3A 2B4, Canada.
| | - David Arenillas
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Ximena Corso-Díaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada.
| | - Yuan-Yun Xie
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Slavita Bohacec
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Kathleen G Banks
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Russell J Bonaguro
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Siaw H Wong
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Steven J M Jones
- Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, V5Z 4S6, Canada. .,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, V5Z 4S6, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, BC, V6T 2A1, Canada.
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
40
|
Genetic architecture of natural variation in Drosophila melanogaster aggressive behavior. Proc Natl Acad Sci U S A 2015; 112:E3555-63. [PMID: 26100892 DOI: 10.1073/pnas.1510104112] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aggression is an evolutionarily conserved complex behavior essential for survival and the organization of social hierarchies. With the exception of genetic variants associated with bioamine signaling, which have been implicated in aggression in many species, the genetic basis of natural variation in aggression is largely unknown. Drosophila melanogaster is a favorable model system for exploring the genetic basis of natural variation in aggression. Here, we performed genome-wide association analyses using the inbred, sequenced lines of the Drosophila melanogaster Genetic Reference Panel (DGRP) and replicate advanced intercross populations derived from the most and least aggressive DGRP lines. We identified genes that have been previously implicated in aggressive behavior as well as many novel loci, including gustatory receptor 63a (Gr63a), which encodes a subunit of the receptor for CO2, and genes associated with development and function of the nervous system. Although genes from the two association analyses were largely nonoverlapping, they mapped onto a genetic interaction network inferred from an analysis of pairwise epistasis in the DGRP. We used mutations and RNAi knock-down alleles to functionally validate 79% of the candidate genes and 75% of the candidate epistatic interactions tested. Epistasis for aggressive behavior causes cryptic genetic variation in the DGRP that is revealed by changing allele frequencies in the outbred populations derived from extreme DGRP lines. This phenomenon may pertain to other fitness traits and species, with implications for evolution, applied breeding, and human genetics.
Collapse
|
41
|
Corso-Díaz X, Simpson EM. Nr2e1 regulates retinal lamination and the development of Müller glia, S-cones, and glycineric amacrine cells during retinogenesis. Mol Brain 2015; 8:37. [PMID: 26092486 PMCID: PMC4475312 DOI: 10.1186/s13041-015-0126-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/23/2015] [Indexed: 12/25/2022] Open
Abstract
Background Nr2e1 is a nuclear receptor crucial for neural stem cell proliferation and maintenance. In the retina, lack of Nr2e1 results in premature neurogenesis, aberrant blood vessel formation and dystrophy. However, the specific role of Nr2e1 in the development of different retinal cell types and its cell-autonomous and non-cell autonomous function(s) during eye development are poorly understood. Results Here, we studied the retinas of P7 and P21 Nr2e1frc/frc mice and Nr2e1+/+ ↔ Nr2e1frc/frc chimeras. We hypothesized that Nr2e1 differentially regulates the development of various retinal cell types, and thus the cellular composition of Nr2e1frc/frc retinas does not simply reflect an overrepresentation of cells born early and underrepresentation of cells born later as a consequence of premature neurogenesis. In agreement with our hypothesis, lack of Nr2e1 resulted in increased numbers of glycinergic amacrine cells with no apparent increase in other amacrine sub-types, normal numbers of Müller glia, the last cell-type to be generated, and increased numbers of Nr2e1frc/frc S-cones in chimeras. Furthermore, Nr2e1frc/frc Müller glia were mispositioned in the retina and misexpressed the ganglion cell-specific transcription factor Brn3a. Nr2e1frc/frc retinas also displayed lamination defects including an ectopic neuropil forming an additional inner plexiform layer. In chimeric mice, retinal thickness was rescued by 34 % of wild-type cells and Nr2e1frc/frc dystrophy-related phenotypes were no longer evident. However, the formation of an ectopic neuropil, misexpression of Brn3a in Müller glia, and abnormal cell numbers in the inner and outer nuclear layers at P7 were not rescued by wild-type cells. Conclusions Together, these results show that Nr2e1, in addition to having a role in preventing premature cell cycle exit, participates in several other developmental processes during retinogenesis including neurite organization in the inner retina and development of glycinergic amacrine cells, S-cones, and Müller glia. Nr2e1 also regulates various aspects of Müller glia differentiation cell-autonomously. However, Nr2e1 does not have a cell-autonomous role in preventing retinal dystrophy. Thus, Nr2e1 regulates processes involved in neurite development and terminal retinal cell differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0126-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ximena Corso-Díaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 W 28 Ave, Vancouver, V5Z 4H4, BC, Canada.,Genetics Graduate Program, University of British Columbia, Vancouver, V6T 1Z2, BC, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 W 28 Ave, Vancouver, V5Z 4H4, BC, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, V6T 1Z2, BC, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, V6T 1Z3, BC, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, V6T 2A1, BC, Canada.
| |
Collapse
|
42
|
Sun L, Deng H, He L, Hu X, Huang Q, Xue J, Chen J, Shi X, Xu Y. The relationship between NR2E1 and subclinical inflammation in newly diagnosed type 2 diabetic patients. J Diabetes Complications 2015; 29:589-94. [PMID: 25813674 DOI: 10.1016/j.jdiacomp.2014.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/25/2014] [Accepted: 12/26/2014] [Indexed: 02/06/2023]
Abstract
AIMS To evaluate the expression level of NR2E1 (nuclear receptor subfamily 2,group E,member 1) and its correlation with type 2 diabetes (T2DM). METHODS Plasma and peripheral blood mononuclear cells (PBMCs) were collected from 54 T2DM and 88 healthy individuals. The levels of free fatty acids (FFAs), total cholesterol (TC), triglyceride (TG), high density lipoprotein (HDL-c), low density lipoprotein (LDL-c), fasting insulin (FIN), and fasting blood glucose (FBG) were measured. The insulin resistance index was calculated using the homeostasis model assessment (HOMA). NR2E1 in PBMCs were analyzed using real-time RT-PCR and Western blots. Tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) in the plasma were measured by enzyme-linked immunosorbent assay (ELISA). PBMCs isolated from healthy volunteers were treated with glucose or palmitate for 24h, followed by analysis for the expression level of NR2E1.The amount of TNF-α and IL-6 secreted into the supernatant were measured by ELISA. RESULTS FIN, FBG, HOMA and TNF-α, IL-6 were significantly higher in diabetic patients, compared to the control group. Levels of NR2E1 were significantly higher in the PBMCs isolated from the diabetic group, compared to the control group. NR2E1 expression was positively correlated with FBG, FIN, HOMA, FFAs, TNF-α and IL-6. Glucose and palmitate treatment significantly increased NR2E1 gene expression and inflammatory cytokines production in PBMCs in vitro. CONCLUSIONS Increased NR2E1 level may be closely associated with inflammation and disorder of lipid and glucose metabolism in diabetic patients.
Collapse
Affiliation(s)
- Li Sun
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Haohua Deng
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Lanjie He
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xuemei Hu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Qi Huang
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Junli Xue
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Jin Chen
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xiaoli Shi
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Yancheng Xu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China.
| |
Collapse
|
43
|
Perbal L. Early detection of criminality concerns and the social link. LIFE SCIENCES, SOCIETY AND POLICY 2014; 10:13. [PMID: 26085449 PMCID: PMC4648843 DOI: 10.1186/s40504-014-0013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 04/15/2014] [Indexed: 11/23/2022]
Abstract
In modern societies, rhetoric focused on body and health is common as biomedical sciences have taken a big place in people’s lives. They must enhance the society, health and living conditions of citizens. Solving criminality problems remains a major challenge and the early detection of antisocial children - future offenders - promises to offer a solution to criminality thanks to science and medical advances. But in a democratic society that values solidarism and pluralism and tends to preserve the social link, it is necessary to question the ethical relevance of this method of managing criminality. This paper proposes to analyze these questions in the light of debates that have troubled France for a number of years.
Collapse
|
44
|
Genes and Gene Networks Implicated in Aggression Related Behaviour. Neurogenetics 2014; 15:255-66. [DOI: 10.1007/s10048-014-0417-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 08/04/2014] [Indexed: 10/24/2022]
|
45
|
Schmouth JF, Dion PA, Rouleau GA. Genetics of essential tremor: From phenotype to genes, insights from both human and mouse studies. Prog Neurobiol 2014; 119-120:1-19. [DOI: 10.1016/j.pneurobio.2014.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/16/2014] [Accepted: 05/02/2014] [Indexed: 11/30/2022]
|
46
|
Finn R, Evans CC, Lee L. Strain-dependent brain defects in mouse models of primary ciliary dyskinesia with mutations in Pcdp1 and Spef2. Neuroscience 2014; 277:552-67. [PMID: 25073043 DOI: 10.1016/j.neuroscience.2014.07.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 10/25/2022]
Abstract
Hydrocephalus is caused by the accumulation of cerebrospinal fluid (CSF) in the cerebral ventricular system which results in an enlargement of the cranium due to increased intraventricular pressure. The increase in pressure within the brain typically results in sloughing of ciliated ependymal cells, loss of cortical gray matter, and increased gliosis. Congenital hydrocephalus is associated with several syndromes including primary ciliary dyskinesia (PCD), a rare, genetically heterogeneous, pediatric syndrome that results from defects in motile cilia and flagella. We have examined the morphological and physiological defects in the brains of two mouse models of PCD, nm1054 and bgh, which have mutations in Pcdp1 (also known as Cfap221) and Spef2, respectively. Histopathological and immunohistochemical analyses of mice with these mutations on the C57BL/6J and 129S6/SvEvTac genetic backgrounds demonstrate strain-dependent morphological brain damage. Alterations in astrocytosis, microglial activation, myelination, and the neuronal population were identified and are generally more severe on the C57BL/6J background. Analysis of ependymal ciliary clearance ex vivo and CSF flow in vivo demonstrate a physiological defect in nm1054 and bgh mice on both genetic backgrounds, indicating that abnormal cilia-driven flow is not the sole determinant of the severity of hydrocephalus in these models. These results suggest that genetic modifiers play an important role in susceptibility to severe PCD-associated hydrocephalus.
Collapse
Affiliation(s)
- R Finn
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - C C Evans
- Cancer Biology Research Center, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - L Lee
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA; Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57105, USA.
| |
Collapse
|
47
|
Islam MM, Zhang CL. TLX: A master regulator for neural stem cell maintenance and neurogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:210-6. [PMID: 24930777 DOI: 10.1016/j.bbagrm.2014.06.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/22/2014] [Accepted: 06/05/2014] [Indexed: 10/25/2022]
Abstract
The orphan nuclear receptor TLX, also known as NR2E1, is an essential regulator of neural stem cell (NSC) self-renewal, maintenance, and neurogenesis. In vertebrates, TLX is specifically localized to the neurogenic regions of the forebrain and retina throughout development and adulthood. TLX regulates the expression of genes involved in multiple pathways, such as the cell cycle, DNA replication, and cell adhesion. These roles are primarily performed through the transcriptional repression or activation of downstream target genes. Emerging evidence suggests that the misregulation of TLX might play a role in the onset and progression of human neurological disorders making this factor an ideal therapeutic target. Here, we review the current understanding of TLX function, expression, regulation, and activity significant to NSC maintenance, adult neurogenesis, and brain plasticity. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Mohammed M Islam
- Department of Molecular Biology, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, 6000 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
48
|
Nuclear receptor TLX stimulates hippocampal neurogenesis and enhances learning and memory in a transgenic mouse model. Proc Natl Acad Sci U S A 2014; 111:9115-20. [PMID: 24927526 DOI: 10.1073/pnas.1406779111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The role of the nuclear receptor TLX in hippocampal neurogenesis and cognition has just begun to be explored. In this study, we generated a transgenic mouse model that expresses TLX under the control of the promoter of nestin, a neural precursor marker. Transgenic TLX expression led to mice with enlarged brains with an elongated hippocampal dentate gyrus and increased numbers of newborn neurons. Specific expression of TLX in adult hippocampal dentate gyrus via lentiviral transduction increased the numbers of BrdU(+) cells and BrdU(+)NeuN(+) neurons. Furthermore, the neural precursor-specific expression of the TLX transgene substantially rescued the neurogenic defects of TLX-null mice. Consistent with increased neurogenesis in the hippocampus, the TLX transgenic mice exhibited enhanced cognition with increased learning and memory. These results suggest a strong association between hippocampal neurogenesis and cognition, as well as significant contributions of TLX to hippocampal neurogenesis, learning, and memory.
Collapse
|
49
|
Kim BJ, Scott DA. Mouse model reveals the role of RERE in cerebellar foliation and the migration and maturation of Purkinje cells. PLoS One 2014; 9:e87518. [PMID: 24466353 PMCID: PMC3900724 DOI: 10.1371/journal.pone.0087518] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/22/2013] [Indexed: 11/22/2022] Open
Abstract
Nuclear receptors and their coregulators play a critical role in brain development by regulating the spatiotemporal expression of their target genes. The arginine-glutamic acid dipeptide repeats gene (Rere) encodes a nuclear receptor coregulator previously known as Atrophin 2. In the developing cerebellum, RERE is expressed in the molecular layer, the Purkinje cell layer and the granule cell layer but not in granule cell precursors. To study RERE's role in cerebellar development, we used RERE-deficient embryos bearing a null allele (om) and a hypomorphic allele (eyes3) of Rere (Rereom/eyes3). In contrast to wild-type embryos, formation of the principal fissures in these RERE-deficient embryos was delayed and the proliferative activity of granule cell precursors (GCPs) was reduced at E18.5. This reduction in proliferation was accompanied by a decrease in the expression of sonic hedgehog (SHH), which is secreted from Purkinje cells and is required for normal GCP proliferation. The maturation and migration of Purkinje cells in Rereom/eyes3 embryos was also delayed with decreased numbers of post-migratory Purkinje cells in the cerebellum. During the postnatal period, RERE depletion caused incomplete division of lobules I/II and III due to truncated development of the precentral fissure in the cerebellar vermis, abnormal development of lobule crus I and lobule crus II in the cerebellar hemispheres due to attenuation of the intercrural fissure, and decreased levels of Purkinje cell dendritic branching. We conclude that RERE-deficiency leads to delayed development of the principal fissures and delayed maturation and migration of Purkinje cells during prenatal cerebellar development and abnormal cerebellar foliation and Purkinje cell maturation during postnatal cerebellar development.
Collapse
Affiliation(s)
- Bum Jun Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
50
|
Abstract
The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.
Collapse
Affiliation(s)
- Shannon E Mullican
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|