1
|
Hank EC, Sai M, Kasch T, Meijer I, Marschner JA, Merk D. Development of Tailless Homologue Receptor (TLX) Agonist Chemical Tools. J Med Chem 2024; 67:16598-16611. [PMID: 39236094 DOI: 10.1021/acs.jmedchem.4c01443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The human tailless homologue receptor (TLX) is a ligand-activated transcription factor acting as a master regulator of neural stem cell homeostasis. Despite its promising potential in neurodegenerative disease treatment, TLX ligands are rare but required to explore phenotypic effects of TLX modulation and for target validation. We have systematically studied and optimized a TLX agonist scaffold obtained by fragment fusion. Structural modification enabled the development of two TLX agonists endowed with nanomolar potency and binding affinity. Both exhibited favorable chemical tool characteristics including high selectivity and low toxicity. Most notably, the TLX agonists comprise different scaffolds and display high chemical diversity, enabling their use as a set for target identification and validation studies.
Collapse
Affiliation(s)
- Emily C Hank
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Minh Sai
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Till Kasch
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Isabelle Meijer
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Julian A Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| |
Collapse
|
2
|
Trangle SS, Rosenberg T, Parnas H, Levy G, Bar E, Marco A, Barak B. In individuals with Williams syndrome, dysregulation of methylation in non-coding regions of neuronal and oligodendrocyte DNA is associated with pathology and cortical development. Mol Psychiatry 2023; 28:1112-1127. [PMID: 36577841 DOI: 10.1038/s41380-022-01921-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder caused by a heterozygous micro-deletion in the WS critical region (WSCR) and is characterized by hyper-sociability and neurocognitive abnormalities. Nonetheless, whether and to what extent WSCR deletion leads to epigenetic modifications in the brain and induces pathological outcomes remains largely unknown. By examining DNA methylation in frontal cortex, we revealed genome-wide disruption in the methylome of individuals with WS, as compared to typically developed (TD) controls. Surprisingly, differentially methylated sites were predominantly annotated as introns and intergenic loci and were found to be highly enriched around binding sites for transcription factors that regulate neuronal development, plasticity and cognition. Moreover, by utilizing enhancer-promoter interactome data, we confirmed that most of these loci function as active enhancers in the human brain or as target genes of transcriptional networks associated with myelination, oligodendrocyte (OL) differentiation, cognition and social behavior. Cell type-specific methylation analysis revealed aberrant patterns in the methylation of active enhancers in neurons and OLs, and important neuron-glia interactions that might be impaired in individuals with WS. Finally, comparison of methylation profiles from blood samples of individuals with WS and healthy controls, along with other data collected in this study, identified putative targets of endophenotypes associated with WS, which can be used to define brain-risk loci for WS outside the WSCR locus, as well as for other associated pathologies. In conclusion, our study illuminates the brain methylome landscape of individuals with WS and sheds light on how these aberrations might be involved in social behavior and physiological abnormalities. By extension, these results may lead to better diagnostics and more refined therapeutic targets for WS.
Collapse
Affiliation(s)
- Sari Schokoroy Trangle
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tali Rosenberg
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Hadar Parnas
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Gilad Levy
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ela Bar
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel.,The School of Neurobiology, Biochemistry & Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Asaf Marco
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel.
| | - Boaz Barak
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
3
|
Faudone G, Zhubi R, Celik F, Knapp S, Chaikuad A, Heering J, Merk D. Design of a Potent TLX Agonist by Rational Fragment Fusion. J Med Chem 2022; 65:2288-2296. [PMID: 34989568 DOI: 10.1021/acs.jmedchem.1c01757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a master regulator of neurogenesis, the orphan nuclear receptor tailless homologue (TLX, NR2E1) maintains neuronal stem cell homeostasis by acting as a transcriptional repressor of tumor suppressor genes. It is hence considered as an appealing target for the treatment of neurodegenerative diseases, but a lack of potent TLX modulators as tools to probe pharmacological TLX control hinders further validation of its promising potential. Here, we report the development of a potent TLX agonist based on fragment screening, pharmacophore modeling, and fragment fusion. Pharmacophore similarity of a fragment screening hit and the TLX ligand ccrp2 provided a rational basis for fragment linkage, which resulted in several TLX activator scaffolds. Among them, the fused compound 10 evolved as a valuable TLX agonist tool with submicromolar potency and high selectivity over related nuclear receptors, rendering it suitable for functional studies on TLX.
Collapse
Affiliation(s)
- Giuseppe Faudone
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Rezart Zhubi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Fatih Celik
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, D-60596 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany.,Department of Pharmacy, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| |
Collapse
|
4
|
Faudone G, Kilu W, Ni X, Chaikuad A, Sreeramulu S, Heitel P, Schwalbe H, Knapp S, Schubert-Zsilavecz M, Heering J, Merk D. The Transcriptional Repressor Orphan Nuclear Receptor TLX Is Responsive to Xanthines. ACS Pharmacol Transl Sci 2021; 4:1794-1807. [PMID: 34927011 PMCID: PMC8669710 DOI: 10.1021/acsptsci.1c00195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Indexed: 11/28/2022]
Abstract
The orphan nuclear receptor tailless homologue (TLX) is expressed almost exclusively in neural stem cells acting as an essential factor for their survival and is hence considered as a promising drug target in neurodegeneration. However, few studies have characterized the roles of TLX due to the lack of ligands and limited functional understanding. Here, we identify xanthines including caffeine and istradefylline as TLX modulators that counteract the receptor's intrinsic repressor activity. Mutagenesis of residues lining a cavity within the TLX ligand binding domain altered the activity of these ligands, suggesting direct interactions with helix 5. Using xanthines as tool compounds, we observed a ligand-sensitive recruitment of the co-repressor silencing mediator for retinoid or thyroid-hormone receptors, TLX homodimerization, and heterodimerization with the retinoid X receptor. These protein-protein interactions evolve as factors that modulate the TLX function and suggest an unprecedented role of TLX in directly repressing other nuclear receptors.
Collapse
Affiliation(s)
- Giuseppe Faudone
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Whitney Kilu
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Xiaomin Ni
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural
Genomics Consortium, BMLS, Goethe University
Frankfurt, Max-von-Laue-Str. 15, D-60438 Frankfurt, Germany
| | - Apirat Chaikuad
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural
Genomics Consortium, BMLS, Goethe University
Frankfurt, Max-von-Laue-Str. 15, D-60438 Frankfurt, Germany
| | - Sridhar Sreeramulu
- Center
for Biomolecular Magnetic Resonance (BMRZ), Institute for Organic
Chemistry and Chemical Biology, Goethe University
Frankfurt, Max-von-Laue-Str. 7, D-60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Harald Schwalbe
- Center
for Biomolecular Magnetic Resonance (BMRZ), Institute for Organic
Chemistry and Chemical Biology, Goethe University
Frankfurt, Max-von-Laue-Str. 7, D-60438 Frankfurt, Germany
| | - Stefan Knapp
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural
Genomics Consortium, BMLS, Goethe University
Frankfurt, Max-von-Laue-Str. 15, D-60438 Frankfurt, Germany
| | - Manfred Schubert-Zsilavecz
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Jan Heering
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Daniel Merk
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Department
of Pharmacy, Ludwig-Maximilians-Universität
München, Butenandtstr.
5-13, D-81377 Munich, Germany
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|
7
|
Faudone G, Bischoff-Kont I, Rachor L, Willems S, Zhubi R, Kaiser A, Chaikuad A, Knapp S, Fürst R, Heering J, Merk D. Propranolol Activates the Orphan Nuclear Receptor TLX to Counteract Proliferation and Migration of Glioblastoma Cells. J Med Chem 2021; 64:8727-8738. [PMID: 34115934 DOI: 10.1021/acs.jmedchem.1c00733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ligand-sensing transcription factor tailless homologue (TLX, NR2E1) is an essential regulator of neuronal stem cell homeostasis with appealing therapeutic potential in neurodegenerative diseases and central nervous system tumors. However, knowledge on TLX ligands is scarce, providing an obstacle to target validation and medicinal chemistry. To discover TLX ligands, we have profiled a drug fragment collection for TLX modulation and identified several structurally diverse agonists and inverse agonists of the nuclear receptor. Propranolol evolved as the strongest TLX agonist and promoted TLX-regulated gene expression in human glioblastoma cells. Structure-activity relationship elucidation of propranolol as a TLX ligand yielded a structurally related negative control compound. In functional cellular experiments, we observed an ability of propranolol to counteract glioblastoma cell proliferation and migration, while the negative control had no effect. Our results provide a collection of TLX modulators as initial chemical tools and set of lead compounds and support therapeutic potential of TLX modulation in glioblastoma.
Collapse
Affiliation(s)
- Giuseppe Faudone
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Iris Bischoff-Kont
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Lea Rachor
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Rezart Zhubi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Astrid Kaiser
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany.,Structural Genomics Consortium, BMLS, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| |
Collapse
|
8
|
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
|
9
|
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: 0.3] [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
|
10
|
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: 1.2] [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
|
11
|
Abdolmaleky HM, Gower AC, Wong CK, Cox JW, Zhang X, Thiagalingam A, Shafa R, Sivaraman V, Zhou JR, Thiagalingam S. Aberrant transcriptomes and DNA methylomes define pathways that drive pathogenesis and loss of brain laterality/asymmetry in schizophrenia and bipolar disorder. Am J Med Genet B Neuropsychiatr Genet 2019; 180:138-149. [PMID: 30468562 PMCID: PMC6386618 DOI: 10.1002/ajmg.b.32691] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 07/23/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Abstract
Although the loss of brain laterality is one of the most consistent modalities in schizophrenia (SCZ) and bipolar disorder (BD), its molecular basis remains elusive. Our limited previous studies indicated that epigenetic modifications are key to the asymmetric transcriptomes of brain hemispheres. We used whole-genome expression microarrays to profile postmortem brain samples from subjects with SCZ, psychotic BD [BD[+]] or non-psychotic BD [BD(-)], or matched controls (10/group) and performed whole-genome DNA methylation (DNAM) profiling of the same samples (3-4/group) to identify pathways associated with SCZ or BD[+] and genes/sites susceptible to epigenetic regulation. qRT-PCR and quantitative DNAM analysis were employed to validate findings in larger sample sets (35/group). Gene Set Enrichment Analysis (GSEA) demonstrated that BMP signaling and astrocyte and cerebral cortex development are significantly (FDR q < 0.25) coordinately upregulated in both SCZ and BD[+], and glutamate signaling and TGFβ signaling are significantly coordinately upregulated in SCZ. GSEA also indicated that collagens are downregulated in right versus left brain of controls, but not in SCZ or BD[+] patients. Ingenuity Pathway Analysis predicted that TGFB2 is an upstream regulator of these genes (p = .0012). While lateralized expression of TGFB2 in controls (p = .017) is associated with a corresponding change in DNAM (p ≤ .023), lateralized expression and DNAM of TGFB2 are absent in SCZ or BD. Loss of brain laterality in SCZ and BD corresponds to aberrant epigenetic regulation of TGFB2 and changes in TGFβ signaling, indicating potential avenues for disease prevention/treatment.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA,Nutrition/Metabolism Laboratory, BIDMC, Harvard Medical School, Boston, MA,Corresponding Authors: Hamid Mostafavi Abdolmaleky () and Sam Thiagalingam ()
| | - Adam Chapin Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, MA
| | - Chen Khuan Wong
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA,Genetics & Genomics Graduate Program, Boston University School of Medicine, Boston, MA
| | - Jiayi Wu Cox
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA,Bioinformatics Graduate Program, Boston University, Boston, MA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA
| | - Arunthathi Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA
| | | | - Vadivelu Sivaraman
- Critical Care Medicine, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, BIDMC, Harvard Medical School, Boston, MA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA,Genetics & Genomics Graduate Program, Boston University School of Medicine, Boston, MA,Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA,Corresponding Authors: Hamid Mostafavi Abdolmaleky () and Sam Thiagalingam ()
| |
Collapse
|
12
|
Dueva E, Singh K, Kalyta A, LeBlanc E, Rennie PS, Cherkasov A. Computer-Aided Discovery of Small Molecule Inhibitors of Transcriptional Activity of TLX (NR2E1) Nuclear Receptor. Molecules 2018; 23:molecules23112967. [PMID: 30441799 PMCID: PMC6278398 DOI: 10.3390/molecules23112967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/01/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022] Open
Abstract
Orphan nuclear receptor TLX (NR2E1) plays a critical role in the regulation of neural stem cells (NSC) as well as in the development of NSC-derived brain tumors. In the last years, new data have emerged implicating TLX in prostate and breast cancer. Therefore, inhibitors of TLX transcriptional activity may have a significant impact on the treatment of several critical malignancies. However, the TLX protein possesses a non-canonical ligand-binding domain (LBD), which lacks a ligand-binding pocket (conventionally targeted in case of nuclear receptors) that complicates the development of small molecule inhibitors of TLX. Herein, we utilized a rational structure-based design approach to identify small molecules targeting the Atro-box binding site of human TLX LBD. As a result of virtual screening of ~7 million molecular structures, 97 compounds were identified and evaluated in the TLX-responsive luciferase reporter assay. Among those, three chemicals demonstrated 40–50% inhibition of luciferase-detected transcriptional activity of the TLX orphan nuclear receptor at a dose of 35 µM. The identified compounds represent the first class of small molecule inhibitors of TLX transcriptional activity identified via methods of computer-aided drug discovery.
Collapse
Affiliation(s)
- Evgenia Dueva
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Kriti Singh
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Anastasia Kalyta
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Eric LeBlanc
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Paul S Rennie
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
13
|
Gkikas D, Tsampoula M, Politis PK. Nuclear receptors in neural stem/progenitor cell homeostasis. Cell Mol Life Sci 2017; 74:4097-4120. [PMID: 28638936 PMCID: PMC11107725 DOI: 10.1007/s00018-017-2571-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022]
Abstract
In the central nervous system, embryonic and adult neural stem/progenitor cells (NSCs) generate the enormous variety and huge numbers of neuronal and glial cells that provide structural and functional support in the brain and spinal cord. Over the last decades, nuclear receptors and their natural ligands have emerged as critical regulators of NSC homeostasis during embryonic development and adult life. Furthermore, substantial progress has been achieved towards elucidating the molecular mechanisms of nuclear receptors action in proliferative and differentiation capacities of NSCs. Aberrant expression or function of nuclear receptors in NSCs also contributes to the pathogenesis of various nervous system diseases. Here, we review recent advances in our understanding of the regulatory roles of steroid, non-steroid, and orphan nuclear receptors in NSC fate decisions. These studies establish nuclear receptors as key therapeutic targets in brain diseases.
Collapse
Affiliation(s)
- Dimitrios Gkikas
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece.
| |
Collapse
|
14
|
Yang C, Ba H, Cao Y, Dong G, Zhang S, Gao Z, Zhao H, Zhou X. Linking Y-chromosomal short tandem repeat loci to human male impulsive aggression. Brain Behav 2017; 7:e00855. [PMID: 29201554 PMCID: PMC5698871 DOI: 10.1002/brb3.855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 04/13/2017] [Accepted: 09/17/2017] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION Men are more susceptible to impulsive behavior than women. Epidemiological studies revealed that the impulsive aggressive behavior is affected by genetic factors, and the male-specific Y chromosome plays an important role in this behavior. In this study, we investigated the association between the impulsive aggressive behavior and Y-chromosomal short tandem repeats (Y-STRs) loci. METHODS The collected biologic samples from 271 offenders with impulsive aggressive behavior and 492 healthy individuals without impulsive aggressive behavior were amplified by PowerPlexRY23 PCR System and the resultant products were separated by electrophoresis and further genotyped. Then, comparisons in allele and haplotype frequencies of the selected 22 Y-STRs were made in the two groups. RESULTS Our results showed that there were significant differences in allele frequencies at DYS448 and DYS456 between offenders and controls (p < .05). Univariate analysis further revealed significant frequency differences for alleles 18 and 22 at DYS448 (0.18 vs 0.27, compared to the controls, p = .003, OR=0.57,95% CI=0.39-0.82; 0.03 vs 0.01, compared to the controls, p = .003, OR=7.45, 95% CI=1.57-35.35, respectively) and for allele 17 at DYS456 (0.07 vs 0.14, compared to the controls, p = .006, OR=0.48, 95% CI =0.28-0.82) between two groups. Interestingly, the frequency of haploid haplotype 22-15 on the DYS448-DYS456 (DYS448-DYS456-22-15) was significantly higher in offenders than in controls (0.033 vs 0.004, compared to the control, p = .001, OR = 8.42, 95%CI =1.81-39.24). Moreover, there were no significant differences in allele frequencies of other Y-STRs loci between two groups. Furthermore, the unconditional logistic regression analysis confirmed that alleles 18 and 22 at DYS448 and allele 17 at DYS456 are associated with male impulsive aggression. However, the DYS448-DYS456-22-15 is less related to impulsive aggression. CONCLUSION Our results suggest a link between Y-chromosomal allele types and male impulsive aggression.
Collapse
Affiliation(s)
- Chun Yang
- Department of Psychiatry Psychiatry Center of Chinese People's Liberation Army No.102 Hospital of People's Liberation Army Changzhou China
| | - Huajie Ba
- DNA Laboratory Public Security Bureau of Changzhou Changzhou China
| | - Yin Cao
- Department of Neurology Laboratory of Neurological Diseases Changzhou No.2 People's Hospital The Affiliated Hospital of Nanjing Medical University Changzhou China
| | - Guoying Dong
- Department of Neurology Laboratory of Neurological Diseases Changzhou No.2 People's Hospital The Affiliated Hospital of Nanjing Medical University Changzhou China
| | - Shuyou Zhang
- Department of Psychiatry Psychiatry Center of Chinese People's Liberation Army No.102 Hospital of People's Liberation Army Changzhou China
| | - Zhiqin Gao
- Department of Psychiatry Psychiatry Center of Chinese People's Liberation Army No.102 Hospital of People's Liberation Army Changzhou China
| | - Hanqing Zhao
- Department of Psychiatry Psychiatry Center of Chinese People's Liberation Army No.102 Hospital of People's Liberation Army Changzhou China
| | - Xianju Zhou
- Department of Neurology Laboratory of Neurological Diseases Changzhou No.2 People's Hospital The Affiliated Hospital of Nanjing Medical University Changzhou China
| |
Collapse
|
15
|
Horodyska J, Hamill RM, Varley PF, Reyer H, Wimmers K. Genome-wide association analysis and functional annotation of positional candidate genes for feed conversion efficiency and growth rate in pigs. PLoS One 2017; 12:e0173482. [PMID: 28604785 PMCID: PMC5467825 DOI: 10.1371/journal.pone.0173482] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 05/24/2017] [Indexed: 01/03/2023] Open
Abstract
Feed conversion efficiency is a measure of how well an animal converts feed into live weight and it is typically expressed as feed conversion ratio (FCR). FCR and related traits like growth rate (e.g. days to 110 kg—D110) are of high interest for animal breeders, farmers and society due to implications on animal performance, feeding costs and environmental sustainability. The objective of this study was to identify genomic regions associated with FCR and D110 in pigs. A total of 952 terminal line boars, showing an individual variation in FCR, were genotyped using 60K SNP-Chips. Markers were tested for associations with estimated breeding values (EBV) for FCR and D110. For FCR, the largest number of associated SNPs was located on chromosomes 4 (30 SNPs), 1 (25 SNPs), X (15 SNPs) and 6 (12 SNPs). The most prominent genomic regions for D110 were identified on chromosomes 15 (10 SNPs), 1 and 4 (both 9 SNPs). The most significantly associated SNPs for FCR and D110 mapped 129.8 Kb from METTL11B (chromosome 4) and 32Kb from MBD5 (chromosome 15), respectively. A list of positional genes, closest to significantly associated SNPs, was used to identify enriched pathways and biological functions related to the QTL for both traits. A number of candidate genes were significantly overrepresented in pathways of immune cell trafficking, lymphoid tissue structure, organ morphology, endocrine system function, lipid metabolism, and energy production. After resequencing the coding region of selected positional and functional candidate genes, six SNPs were genotyped in a subset of boars. SNPs in PRKDC, SELL, NR2E1 and AKRIC3 showed significant associations with EBVs for FCR/D110. The study revealed a number of chromosomal regions and candidate genes affecting FCR/D110 and pointed to corresponding biological pathways related to lipid metabolism, olfactory reception, and also immunological status.
Collapse
Affiliation(s)
- Justyna Horodyska
- Teagasc, Food Research Centre, Ashtown, Dublin, Ireland
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | | | | | - Henry Reyer
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
- * E-mail:
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University Rostock, Rostock, Germany
| |
Collapse
|
16
|
|
17
|
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: 1.5] [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
|
18
|
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: 2.3] [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]
|
19
|
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: 4.1] [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
|
20
|
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: 1.4] [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
|
21
|
Veroude K, Zhang-James Y, Fernàndez-Castillo N, Bakker MJ, Cormand B, Faraone SV. Genetics of aggressive behavior: An overview. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:3-43. [PMID: 26345359 DOI: 10.1002/ajmg.b.32364] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/05/2015] [Indexed: 12/24/2022]
Abstract
The Research Domain Criteria (RDoC) address three types of aggression: frustrative non-reward, defensive aggression and offensive/proactive aggression. This review sought to present the evidence for genetic underpinnings of aggression and to determine to what degree prior studies have examined phenotypes that fit into the RDoC framework. Although the constructs of defensive and offensive aggression have been widely used in the animal genetics literature, the human literature is mostly agnostic with regard to all the RDoC constructs. We know from twin studies that about half the variance in behavior may be explained by genetic risk factors. This is true for both dimensional, trait-like, measures of aggression and categorical definitions of psychopathology. The non-shared environment seems to have a moderate influence with the effects of shared environment being unclear. Human molecular genetic studies of aggression are in an early stage. The most promising candidates are in the dopaminergic and serotonergic systems along with hormonal regulators. Genome-wide association studies have not yet achieved genome-wide significance, but current samples are too small to detect variants having the small effects one would expect for a complex disorder. The strongest molecular evidence for a genetic basis for aggression comes from animal models comparing aggressive and non-aggressive strains or documenting the effects of gene knockouts. Although we have learned much from these prior studies, future studies should improve the measurement of aggression by using a systematic method of measurement such as that proposed by the RDoC initiative.
Collapse
Affiliation(s)
- Kim Veroude
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Yanli Zhang-James
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,Departments of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - Mireille J Bakker
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Bru Cormand
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,Departments of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| |
Collapse
|
22
|
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: 94] [Impact Index Per Article: 10.4] [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
|
23
|
Lopez-Castroman J, Baca-Garcia E, Woreca Authors, Courtet P, Oquendo MA. A Cross-National Tool for Assessing and Studying Suicidal Behaviors. Arch Suicide Res 2015; 19:335-49. [PMID: 25529607 DOI: 10.1080/13811118.2014.981624] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Current methodologies employed in collecting data for suicide research present several limitations. In this article, we describe a novel method and tool to prospectively study suicidal behavior and its related risk and protective factors in different countries. This tool is a web-based database, which will initially be developed and piloted by suicide research groups from Austria, Brazil, Chile, France, Italy, Mexico, Spain, Sweden, and the US. The database will develop and integrate research tools key to collaboration, permit exploration of ethical considerations, and provide a universal interface in English, Spanish, French, Portuguese, Italian, and German that will make it possible to incorporate new groups from different countries to the network. The common database resulting of this cross-national effort will allow researchers to share and compare data across countries in a large sample of patients. Using anonymous electronic records of patients, we will: 1) determine the impact of risk and protective factors (viz. aggression and religious affiliation) in different socio-cultural environments; 2) develop a clinical model of suicidal behavior that integrates the multiple dimensions implicated; 3) try differentiating the effects of biological, environmental, and cultural factors on suicidal behavior, and 4) offer the global psychiatric research community a tool that will standardize assessment across nations, allowing for better generalization.
Collapse
|
24
|
Stergiopoulos A, Politis PK. The role of nuclear receptors in controlling the fine balance between proliferation and differentiation of neural stem cells. Arch Biochem Biophys 2013; 534:27-37. [DOI: 10.1016/j.abb.2012.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 08/23/2012] [Accepted: 09/20/2012] [Indexed: 12/22/2022]
|
25
|
Schmouth JF, Bonaguro RJ, Corso-Diaz X, Simpson EM. Modelling human regulatory variation in mouse: finding the function in genome-wide association studies and whole-genome sequencing. PLoS Genet 2012; 8:e1002544. [PMID: 22396661 PMCID: PMC3291530 DOI: 10.1371/journal.pgen.1002544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
An increasing body of literature from genome-wide association studies and human whole-genome sequencing highlights the identification of large numbers of candidate regulatory variants of potential therapeutic interest in numerous diseases. Our relatively poor understanding of the functions of non-coding genomic sequence, and the slow and laborious process of experimental validation of the functional significance of human regulatory variants, limits our ability to fully benefit from this information in our efforts to comprehend human disease. Humanized mouse models (HuMMs), in which human genes are introduced into the mouse, suggest an approach to this problem. In the past, HuMMs have been used successfully to study human disease variants; e.g., the complex genetic condition arising from Down syndrome, common monogenic disorders such as Huntington disease and β-thalassemia, and cancer susceptibility genes such as BRCA1. In this commentary, we highlight a novel method for high-throughput single-copy site-specific generation of HuMMs entitled High-throughput Human Genes on the X Chromosome (HuGX). This method can be applied to most human genes for which a bacterial artificial chromosome (BAC) construct can be derived and a mouse-null allele exists. This strategy comprises (1) the use of recombineering technology to create a human variant-harbouring BAC, (2) knock-in of this BAC into the mouse genome using Hprt docking technology, and (3) allele comparison by interspecies complementation. We demonstrate the throughput of the HuGX method by generating a series of seven different alleles for the human NR2E1 gene at Hprt. In future challenges, we consider the current limitations of experimental approaches and call for a concerted effort by the genetics community, for both human and mouse, to solve the challenge of the functional analysis of human regulatory variation.
Collapse
Affiliation(s)
- Jean-François Schmouth
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, Canada
- Genetics Graduate Program, University of British Columbia, Vancouver, Canada
| | - Russell J. Bonaguro
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | - Ximena Corso-Diaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, Canada
- Genetics Graduate Program, University of British Columbia, Vancouver, Canada
| | - Elizabeth M. Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, Canada
- Genetics Graduate Program, University of British Columbia, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
26
|
Retina restored and brain abnormalities ameliorated by single-copy knock-in of human NR2E1 in null mice. Mol Cell Biol 2012; 32:1296-311. [PMID: 22290436 DOI: 10.1128/mcb.06016-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nr2e1 encodes a stem cell fate determinant of the mouse forebrain and retina. Abnormal regulation of this gene results in retinal, brain, and behavioral abnormalities in mice. However, little is known about the functionality of human NR2E1. We investigated this functionality using a novel knock-in humanized-mouse strain carrying a single-copy bacterial artificial chromosome (BAC). We also documented, for the first time, the expression pattern of the human BAC, using an NR2E1-lacZ reporter strain. Unexpectedly, cerebrum and olfactory bulb hypoplasia, hallmarks of the Nr2e1-null phenotype, were not fully corrected in animals harboring one functional copy of human NR2E1. These results correlated with an absence of NR2E1-lacZ reporter expression in the dorsal pallium of embryos and proliferative cells of adult brains. Surprisingly, retinal histology and electroretinograms demonstrated complete correction of the retina-null phenotype. These results correlated with appropriate expression of the NR2E1-lacZ reporter in developing and adult retina. We conclude that the human BAC contained all the elements allowing correction of the mouse-null phenotype in the retina, while missing key regulatory regions important for proper spatiotemporal brain expression. This is the first time a separation of regulatory mechanisms governing NR2E1 has been demonstrated. Furthermore, candidate genomic regions controlling expression in proliferating cells during neurogenesis were identified.
Collapse
|
27
|
Shimo H, Nakamura M, Tomiyasu A, Ichiba M, Ueno SI, Sano A. Comprehensive analysis of the genes responsible for neuroacanthocytosis in mood disorder and schizophrenia. Neurosci Res 2010; 69:196-202. [PMID: 21145924 DOI: 10.1016/j.neures.2010.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 11/30/2010] [Accepted: 12/01/2010] [Indexed: 11/16/2022]
Abstract
Neuroacanthocytosis syndromes are mainly comprised of two diseases: chorea-acanthocytosis (ChAc) and McLeod syndrome (MLS). There is a high incidence of psychiatric disorders such as mood disorder and schizophrenia among neuroacanthocytosis patients. We hypothesized that neuroacanthocytosis-related-genes might be associated with susceptibility to these psychiatric disorders. We performed a comprehensive mutation screen of VPS13A and XK, the gene responsible for ChAc and MLS, respectively, in 85 mood disorder subjects and XK in 86 schizophrenia subjects and compared the variants to 100 or more control alleles. We also performed copy number variation (CNV) analysis in 72 mood disorder subjects and 86 schizophrenia subjects. We identified three non-synonymous, two synonymous and six intron variants in mood disorder subjects and a novel GAT triplet repeat polymorphism in VPS13A. By CNV analysis, we identified a heterozygous exon 60-61 deletion in VPS13A in one mood disorder subject. We identified one non-synonymous and one intron variant in mood disorder and schizophrenia subjects, respectively, in XK. The presence of a pathogenic mutation or a potentially functional variant in mood disorder or schizophrenia subjects suggests that neuroacanthocytosis-related-genes might be involved in the pathogenesis of these psychiatric disorders.
Collapse
Affiliation(s)
- Hirochika Shimo
- Department of Psychiatry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Sun G, Shi Y. Nuclear receptors in stem cells and their therapeutic potential. Adv Drug Deliv Rev 2010; 62:1299-306. [PMID: 20708051 DOI: 10.1016/j.addr.2010.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 07/28/2010] [Accepted: 08/05/2010] [Indexed: 01/02/2023]
Abstract
The core transcriptional regulatory circuitries are important for controlling stem cell self-renewal and differentiation. Nuclear receptors provide an ideal model to regulate gene expression in both ligand-dependent and ligand-independent manners. Recent studies of regulatory events by nuclear receptors in neural stem cells, embryonic stem cells, and induced pluripotent stem cells (iPSCs) provided unique insights into mechanisms of stem cell regulation and provided invaluable resources for regenerative medicine. Nuclear receptors have been shown to be key players in stem cell self-renewal, pluripotency, and reprogramming. We summarize recent progress of studies on nuclear receptors in stem cell field as well as the potential therapeutic implications of these nuclear receptors and their cognate ligands. These studies not only uncover molecular mechanisms of stem cell regulation, but also provide unique opportunities for drug discovery.
Collapse
|
29
|
Wong BKY, Hossain SM, Trinh E, Ottmann GA, Budaghzadeh S, Zheng QY, Simpson EM. Hyperactivity, startle reactivity and cell-proliferation deficits are resistant to chronic lithium treatment in adult Nr2e1(frc/frc) mice. GENES BRAIN AND BEHAVIOR 2010; 9:681-94. [PMID: 20497236 DOI: 10.1111/j.1601-183x.2010.00602.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The NR2E1 region on Chromosome 6q21-22 has been repeatedly linked to bipolar disorder (BP) and NR2E1 has been associated with BP, and more specifically bipolar I disorder (BPI). In addition, patient sequencing has shown an enrichment of rare candidate-regulatory variants. Interestingly, mice carrying either spontaneous (Nr2e1(frc) ) or targeted (Tlx(-) ) deletions of Nr2e1 (here collectively known as Nr2e1-null) show similar neurological and behavioral anomalies, including hypoplasia of the cerebrum, reduced neural stem cell proliferation, extreme aggression and deficits in fear conditioning; these are the traits that have been observed in some patients with BP. Thus, NR2E1 is a positional and functional candidate for a role in BP. However, no Nr2e1-null mice have been fully evaluated for behaviors used to model BP in rodents or pharmacological responses to drugs effective in treating BP symptoms. In this study we examine Nr2e1(frc/frc) mice, homozygous for the spontaneous deletion, for abnormalities in activity, learning and information processing, and cell proliferation; these are the phenotypes that are either affected in patients with BP or commonly assessed in rodent models of BP. The effect of lithium, a drug used to treat BP, was also evaluated for its ability to attenuate Nr2e1(frc/frc) behavioral and neural stem cell-proliferation phenotypes. We show for the first time that Nr2e1-null mice exhibit extreme hyperactivity in the open field as early as postnatal day 18 and in the home cage, deficits in open-field habituation and passive avoidance, and surprisingly, an absence of acoustic startle. We observed a reduction in neural stem/progenitor cell proliferation in Nr2e1(frc/frc) mice, similar to that seen in other Nr2e1-null strains. These behavioral and cell-proliferation phenotypes were resistant to chronic-adult-lithium treatment. Thus, Nr2e1(frc/frc) mice exhibit behavioral traits used to model BP in rodents, but our results do not support Nr2e1(frc/frc) mice as pharmacological models for BP.
Collapse
Affiliation(s)
- B K Y Wong
- Centre for Molecular Medicine and Therapeutics at the Child & Family Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Ising M, Lucae S, Binder EB, Bettecken T, Uhr M, Ripke S, Kohli MA, Hennings JM, Horstmann S, Kloiber S, Menke A, Bondy B, Rupprecht R, Domschke K, Baune BT, Arolt V, Rush AJ, Holsboer F, Müller-Myhsok B. A genomewide association study points to multiple loci that predict antidepressant drug treatment outcome in depression. ACTA ACUST UNITED AC 2009; 66:966-975. [PMID: 19736353 DOI: 10.1001/archgenpsychiatry.2009.95] [Citation(s) in RCA: 258] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CONTEXT The efficacy of antidepressant drug treatment in depression is unsatisfactory; 1 in 3 patients does not fully recover even after several treatment trials. Genetic factors and clinical characteristics contribute to the failure of a favorable treatment outcome. OBJECTIVE To identify genetic and clinical determinants of antidepressant drug treatment outcome in depression. DESIGN Genomewide pharmacogenetic association study with 2 independent replication samples. SETTING We performed a genomewide association study in patients from the Munich Antidepressant Response Signature (MARS) project and in pooled DNA from an independent German replication sample. A set of 328 single-nucleotide polymorphisms highly related to outcome in both genomewide association studies was genotyped in a sample of the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study. PARTICIPANTS A total of 339 inpatients with a depressive episode (MARS sample), a further 361 inpatients with depression (German replication sample), and 832 outpatients with major depression (STAR*D sample). MAIN OUTCOME MEASURES We generated a multilocus genetic variable that described the individual number of alleles of the selected single nucleotide polymorphisms associated with beneficial treatment outcome in the MARS sample ("response" alleles) to evaluate additive genetic effects on antidepressant drug treatment outcome. RESULTS Multilocus analysis revealed a significant contribution of a binary variable that categorized patients as carriers of a high vs low number of response alleles in the prediction of antidepressant drug treatment outcome in both samples (MARS and STAR*D). In addition, we observed that patients with a comorbid anxiety disorder combined with a low number of response alleles showed the least favorable outcome. CONCLUSION These results demonstrate the importance of multiple genetic factors combined with clinical features in the prediction of antidepressant drug treatment outcome, which underscores the multifactorial nature of this trait.
Collapse
Affiliation(s)
- Marcus Ising
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Susanne Lucae
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Elisabeth B Binder
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Thomas Bettecken
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Manfred Uhr
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Stephan Ripke
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Martin A Kohli
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Johannes M Hennings
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Sonja Horstmann
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Stefan Kloiber
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Andreas Menke
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Brigitta Bondy
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Rainer Rupprecht
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Katharina Domschke
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Bernhard T Baune
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Volker Arolt
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - A John Rush
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Florian Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany (Drs Ising, Lucae, Binder, Bettecken, Uhr, Ripke, Kohli, Hennings, Horstmann, Kloiber, Menke, Holsboer, and Müller-Myhsok), Department of Psychiatry, Ludwig Maximilians University, Munich, Germany (Drs Bondy and Rupprecht), Department of Psychiatry, Westfalian Wilhelms University Muenster, Muenster, Germany (Drs Domschke, Baune, and Arolt), Department of Psychiatry, James Cook University, Townsville, Australia (Dr Baune), Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA, and Department of Clinical Sciences, Duke-NUS, Singapore (Dr Rush)
| |
Collapse
|
31
|
Kripke DF, Nievergelt CM, Joo EJ, Shekhtman T, Kelsoe JR. Circadian polymorphisms associated with affective disorders. J Circadian Rhythms 2009; 7:2. [PMID: 19166596 PMCID: PMC2661876 DOI: 10.1186/1740-3391-7-2] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 01/23/2009] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clinical symptoms of affective disorders, their response to light treatment, and sensitivity to other circadian interventions indicate that the circadian system has a role in mood disorders. Possibly the mechanisms involve circadian seasonal and photoperiodic mechanisms. Since genetic susceptibilities contribute a strong component to affective disorders, we explored whether circadian gene polymorphisms were associated with affective disorders in four complementary studies. METHODS Four groups of subjects were recruited from several sources: 1) bipolar proband-parent trios or sib-pair-parent nuclear families, 2) unrelated bipolar participants who had completed the BALM morningness-eveningness questionnaire, 3) sib pairs from the GenRed Project having at least one sib with early-onset recurrent unipolar depression, and 4) a sleep clinic patient group who frequently suffered from depression. Working mainly with the SNPlex assay system, from 2 to 198 polymorphisms in genes related to circadian function were genotyped in the participant groups. Associations with affective disorders were examined with TDT statistics for within-family comparisons. Quantitative trait associations were examined within the unrelated samples. RESULTS In NR1D1, rs2314339 was associated with bipolar disorder (P = 0.0005). Among the unrelated bipolar participants, 3 SNPs in PER3 and CSNK1E were associated with the BALM score. A PPARGC1B coding SNP, rs7732671, was associated with affective disorder with nominal significance in bipolar family groups and independently in unipolar sib pairs. In TEF, rs738499 was associated with unipolar depression; in a replication study, rs738499 was also associated with the QIDS-SR depression scale in the sleep clinic patient sample. CONCLUSION Along with anti-manic effects of lithium and the antidepressant effects of bright light, these findings suggest that perturbations of the circadian gene network at several levels may influence mood disorders, perhaps ultimately through regulation of MAOA and its modulation of dopamine transmission. Twenty-three associations of circadian polymorphisms with affective symptoms met nominal significance criteria (P < 0.05), whereas 15 would be expected by chance, indicating that many represented false discoveries (Type II errors). Some evidence of replication has been gathered, but more studies are needed to ascertain if circadian gene polymorphisms contribute to susceptibility to affective disorders.
Collapse
Affiliation(s)
- Daniel F Kripke
- Department of Psychiatry 0939, University of California, San Diego, La Jolla, CA 92093-0939, USA
- Scripps Clinic Sleep Center W207, 10666 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Caroline M Nievergelt
- Department of Psychiatry 0939, University of California, San Diego, La Jolla, CA 92093-0939, USA
| | - EJ Joo
- Department of Neuropsychiatry, Eulji University School of Medicine, Eulji General Hospital, Nowongu Hagedong 280-1, Seoul, Korea
| | - Tatyana Shekhtman
- Department of Psychiatry 0939, University of California, San Diego, La Jolla, CA 92093-0939, USA
| | - John R Kelsoe
- Department of Psychiatry 0939, University of California, San Diego, La Jolla, CA 92093-0939, USA
| |
Collapse
|