1
|
Sun X, Zhang J, Dong B, Xiong Q, Wang X, Gu Y, Wang Z, Liu H, Zhang J, He X, Liu H, Zhong Y, Yi C, Chi X, Liu Z, Pang X, Cui Y. Targeting SLITRK4 Restrains Proliferation and Liver Metastasis in Colorectal Cancer via Regulating PI3K/AKT/NFκB Pathway and Tumor-Associated Macrophage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2400367. [PMID: 39499724 DOI: 10.1002/advs.202400367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 10/28/2024] [Indexed: 11/07/2024]
Abstract
Liver metastasis is the major cause of death in colorectal cancer (CRC) due to the lack of effective treatment. To explore novel drivers of CRC liver metastasis, the transcriptomes of primary paracancerous, colorectal tumors and metastases from human patients are profiled. It is found that SLIT- and NTRK-like family member 4 (SLITRK4) is the top upregulated gene in liver metastases and is associated with worse overall survival of CRC patients. Multiple in vitro and in vivo models suggested SLITRK4 promoted CRC tumorigenesis, invasion, migration, and angiogenesis, and inhibition of it restrained CRC tumor growth and liver metastasis with a more profound effect on the tumor microenvironment (TME). Mechanistically, SLITRK4 overexpression significantly activated the PI3K/AKT/NFκB pathway, regulated extracellular matrix organization, and multiple cytokines expression. Furthermore, the results from coculture models and single-cell RNA sequencing analyses suggested SLITRK4 promoted tumor-associated macrophages (TAMs) infiltration and polarization. In addition, macrophage depletion significantly inhibited SLITRK4-induced liver metastasis in CRC. Finally, pharmacological inhibition of SLITRK4 by using lipid-polymer hybrid nanoparticles (NPs) for systemic siRNA delivery can effectively inhibit CRC liver metastasis. Taken together, these results pinpoint that SLITRK4 regulates CRC tumorigenesis and liver metastasis, and siRNA delivering NPs agents validate the therapeutic potential of targeting SLITRK4 in CRC.
Collapse
Affiliation(s)
- Xiaojiao Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Junling Zhang
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Bingqi Dong
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Qingqing Xiong
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute, Tianjin, 300060, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Yanlun Gu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Zhiqi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Huiyu Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Xu He
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Hongjin Liu
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Yi Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chuxiao Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaowei Chi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaocong Pang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Yimin Cui
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| |
Collapse
|
2
|
Puranik N, Song M. Insight into the Association between Slitrk Protein and Neurodevelopmental and Neuropsychiatric Conditions. Biomolecules 2024; 14:1060. [PMID: 39334827 PMCID: PMC11430182 DOI: 10.3390/biom14091060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Slitrk proteins belong the leucine-rich repeat transmembrane family and share structural similarities with the Slits and tropomyosin receptor kinase families, which regulate the development of the nervous system. Slitrks are highly expressed in the developing nervous system of vertebrates, modulating neurite outgrowth and enhancing synaptogenesis; however, the expression and function of Slitrk protein members differ. Slitrk protein variations have been associated with various sensory and neuropsychiatric conditions, including myopia, deafness, obsessive-compulsive disorder, autism spectrum disorders, schizophrenia, attention-deficit/hyperactivity disorder, glioma, and Tourette syndrome; however, the underlying mechanism remains unclear. Therefore, the Slitrk family members' protein expression, roles in the signaling cascade, functions, and gene mutations need to be comprehensively studied to develop therapeutics against neurodegenerative diseases. This study presents complete and pertinent information demonstrating the relationship between Slitrk family proteins and neuropsychiatric illnesses. This review briefly discusses neurodevelopmental disorders, the leucine-rich repeat family, the Slitrk family, and the association of Slitrk with the neuropathology of representative disorders.
Collapse
Affiliation(s)
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
3
|
Van Pee T, Martens DS, Alfano R, Engelen L, Sleurs H, Rasking L, Plusquin M, Nawrot TS. Cord Blood Proteomic Profiles, Birth Weight, and Early Life Growth Trajectories. JAMA Netw Open 2024; 7:e2411246. [PMID: 38743419 PMCID: PMC11094560 DOI: 10.1001/jamanetworkopen.2024.11246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/13/2024] [Indexed: 05/16/2024] Open
Abstract
Importance The cord blood proteome, a repository of proteins derived from both mother and fetus, might offer valuable insights into the physiological and pathological state of the fetus. However, its association with birth weight and growth trajectories early in life remains unexplored. Objective To identify cord blood proteins associated with birth weight and the birth weight ratio (BWR) and to evaluate the associations of these cord blood proteins with early growth trajectories. Design, Setting, and Participants This cohort study included 288 mother-child pairs from the ongoing prospective Environmental Influence on Early Aging birth cohort study. Newborns were recruited from East-Limburg Hospital in Genk, Belgium, between February 2010 and November 2017 and followed up until ages 4 to 6 years. Data were analyzed from February 2022 to September 2023. Main Outcomes and Measures The outcome of interest was the associations of 368 inflammatory-related cord blood proteins with birth weight or BWR and with early life growth trajectories (ie, rapid growth at age 12 months and weight, body mass index [BMI] z score, waist circumference, and overweight at age 4-6 years) using multiple linear regression models. The BWR was calculated by dividing the birth weight by the median birth weight of the population-specific reference growth curve, considering parity, sex, and gestational age. Results are presented as estimates or odds ratios (ORs) for each doubling in proteins. Results The sample included 288 infants (125 [43.4%] male; mean [SD] gestation age, 277.2 [11.6] days). The mean (SD) age of the child at the follow-up examination was 4.6 (0.4) years old. After multiple testing correction, there were significant associations of birth weight and BWR with 7 proteins: 2 positive associations: afamin (birth weight: coefficient, 341.16 [95% CI, 192.76 to 489.50]) and secreted frizzled-related protein 4 (SFRP4; birth weight: coefficient, 242.60 [95% CI, 142.77 to 342.43]; BWR: coefficient, 0.07 [95% CI, 0.04 to 0.10]) and 5 negative associations: cadherin EGF LAG 7-pass G-type receptor 2 (CELSR2; birth weight: coefficient, -237.52 [95% CI, -343.15 to -131.89]), ephrin type-A receptor 4 (EPHA4; birth weight: coefficient, -342.78 [95% CI, -463.10 to -222.47]; BWR: coefficient, -0.11 [95% CI, -0.14 to -0.07]), SLIT and NTRK-like protein 1 (SLITRK1; birth weight: coefficient, -366.32 [95% CI, -476.66 to -255.97]; BWR: coefficient, -0.11 [95% CI, -0.15 to -0.08]), transcobalamin-1 (TCN1; birth weight: coefficient, -208.75 [95% CI, -305.23 to -112.26]), and unc-5 netrin receptor D (UNC5D; birth weight: coefficient, -209.27 [95% CI, -295.14 to -123.40]; BWR: coefficient, -0.07 [95% CI, -0.09 to -0.04]). Further evaluation showed that 2 proteins were still associated with rapid growth at age 12 months (afamin: OR, 0.32 [95% CI, 0.11-0.88]; TCN1: OR, 2.44 [95% CI, 1.26-4.80]). At age 4 to 6 years, CELSR2, EPHA4, SLITRK1, and UNC5D were negatively associated with weight (coefficients, -1.33 to -0.68 kg) and body mass index z score (coefficients, -0.41 to -0.23), and EPHA4, SLITRK1, and UNC5D were negatively associated with waist circumference (coefficients, -1.98 to -0.87 cm). At ages 4 to 6 years, afamin (OR, 0.19 [95% CI, 0.05-0.70]) and SLITRK1 (OR, 0.32 [95% CI, 0.10-0.99]) were associated with lower odds for overweight. Conclusions and Relevance This cohort study found 7 cord blood proteins associated with birth weight and growth trajectories early in life. Overall, these findings suggest that stressors that could affect the cord blood proteome during pregnancy might have long-lasting associations with weight and body anthropometrics.
Collapse
Affiliation(s)
- Thessa Van Pee
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Dries S. Martens
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Rossella Alfano
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Liesa Engelen
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Hanne Sleurs
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Leen Rasking
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health and Primary Care, Leuven University, Leuven, Belgium
| |
Collapse
|
4
|
Matsumoto Y, Miwa H, Katayama KI, Watanabe A, Yamada K, Ito T, Nakagawa S, Aruga J. Slitrk4 is required for the development of inhibitory neurons in the fear memory circuit of the lateral amygdala. Front Mol Neurosci 2024; 17:1386924. [PMID: 38736483 PMCID: PMC11082273 DOI: 10.3389/fnmol.2024.1386924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
The Slitrk family consists of six synaptic adhesion molecules, some of which are associated with neuropsychiatric disorders. In this study, we aimed to investigate the physiological role of Slitrk4 by analyzing Slitrk4 knockout (KO) mice. The Slitrk4 protein was widely detected in the brain and was abundant in the olfactory bulb and amygdala. In a systematic behavioral analysis, male Slitrk4 KO mice exhibited an enhanced fear memory acquisition in a cued test for classical fear conditioning, and social behavior deficits in reciprocal social interaction tests. In an electrophysiological analysis using amygdala slices, Slitrk4 KO mice showed enhanced long-term potentiation in the thalamo-amygdala afferents and reduced feedback inhibition. In the molecular marker analysis of Slitrk4 KO brains, the number of calretinin (CR)-positive interneurons was decreased in the anterior part of the lateral amygdala nuclei at the adult stage. In in vitro experiments for neuronal differentiation, Slitrk4-deficient embryonic stem cells were defective in inducing GABAergic interneurons with an altered response to sonic hedgehog signaling activation that was involved in the generation of GABAergic interneuron subsets. These results indicate that Slitrk4 function is related to the development of inhibitory neurons in the fear memory circuit and would contribute to a better understanding of osttraumatic stress disorder, in which an altered expression of Slitrk4 has been reported.
Collapse
Affiliation(s)
- Yoshifumi Matsumoto
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Hideki Miwa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kei-ichi Katayama
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Arata Watanabe
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Kazuyuki Yamada
- Support Unit for Animal Experiments, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Takashi Ito
- Department of Biochemistry, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Shinsuke Nakagawa
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Jun Aruga
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
5
|
Efthymiou S, Han W, Ilyas M, Li J, Yu Y, Scala M, Malintan NT, Ilyas M, Vavouraki N, Mankad K, Maroofian R, Rocca C, Salpietro V, Lakhani S, Mallack EJ, Palculict TB, Li H, Zhang G, Zafar F, Rana N, Takashima N, Matsunaga H, Manzoni C, Striano P, Lythgoe MF, Aruga J, Lu W, Houlden H. Human mutations in SLITRK3 implicated in GABAergic synapse development in mice. Front Mol Neurosci 2024; 17:1222935. [PMID: 38495551 PMCID: PMC10940442 DOI: 10.3389/fnmol.2024.1222935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 02/02/2024] [Indexed: 03/19/2024] Open
Abstract
This study reports on biallelic homozygous and monoallelic de novo variants in SLITRK3 in three unrelated families presenting with epileptic encephalopathy associated with a broad neurological involvement characterized by microcephaly, intellectual disability, seizures, and global developmental delay. SLITRK3 encodes for a transmembrane protein that is involved in controlling neurite outgrowth and inhibitory synapse development and that has an important role in brain function and neurological diseases. Using primary cultures of hippocampal neurons carrying patients' SLITRK3 variants and in combination with electrophysiology, we demonstrate that recessive variants are loss-of-function alleles. Immunostaining experiments in HEK-293 cells showed that human variants C566R and E606X change SLITRK3 protein expression patterns on the cell surface, resulting in highly accumulating defective proteins in the Golgi apparatus. By analyzing the development and phenotype of SLITRK3 KO (SLITRK3-/-) mice, the study shows evidence of enhanced susceptibility to pentylenetetrazole-induced seizure with the appearance of spontaneous epileptiform EEG as well as developmental deficits such as higher motor activities and reduced parvalbumin interneurons. Taken together, the results exhibit impaired development of the peripheral and central nervous system and support a conserved role of this transmembrane protein in neurological function. The study delineates an emerging spectrum of human core synaptopathies caused by variants in genes that encode SLITRK proteins and essential regulatory components of the synaptic machinery. The hallmark of these disorders is impaired postsynaptic neurotransmission at nerve terminals; an impaired neurotransmission resulting in a wide array of (often overlapping) clinical features, including neurodevelopmental impairment, weakness, seizures, and abnormal movements. The genetic synaptopathy caused by SLITRK3 mutations highlights the key roles of this gene in human brain development and function.
Collapse
Affiliation(s)
- Stephanie Efthymiou
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
- U.O.C. Genetica Medica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Muhammad Ilyas
- Department of Biological Sciences, International Islamic University Islamabad, Islamabad, Pakistan
| | - Jun Li
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Yichao Yu
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Marcello Scala
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Nancy T. Malintan
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
| | - Muhammad Ilyas
- Centre for Omic Sciences, Islamia College Peshawar, Peshawar, Pakistan
| | - Nikoleta Vavouraki
- School of Pharmacy, University of Reading, Reading, United Kingdom
- Department of Mathematics and Statistics, University of Reading, Reading, United Kingdom
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital, London, United Kingdom
- Developmental Neurosciences Department, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Reza Maroofian
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
| | - Clarissa Rocca
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
| | - Vincenzo Salpietro
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
| | - Shenela Lakhani
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Eric J. Mallack
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | | | - Hong Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Guojun Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Pediatric Neurology, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Faisal Zafar
- Department of Pediatrics, Multan Hospital, Multan, Pakistan
| | - Nuzhat Rana
- Department of Pediatrics, Multan Hospital, Multan, Pakistan
| | - Noriko Takashima
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Saitama, Japan
| | - Hayato Matsunaga
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Claudia Manzoni
- School of Pharmacy, University College London, London, United Kingdom
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Mark F. Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Jun Aruga
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Saitama, Japan
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Henry Houlden
- Department of Neuromuscular Disorders, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
6
|
Zhou YQ, Bao TS, Xie JX, Yao LL, Yu ST, Li Q, Huang PQ, Zhou WZ, Wang YY, Chen SY, Wang XQ, Zhang XL, Jiang SH, Yi SQ, Zhang ZG, Ma MZ, Hu LP, Xu J, Li J. The SLITRK4-CNPY3 axis promotes liver metastasis of gastric cancer by enhancing the endocytosis and recycling of TrkB in tumour cells. Cell Oncol (Dordr) 2023; 46:1049-1067. [PMID: 37012514 DOI: 10.1007/s13402-023-00795-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/05/2023] Open
Abstract
PURPOSE Gastric cancer (GC) is a malignant tumour with high mortality, and liver metastasis is one of the main causes of poor prognosis. SLIT- and NTRK-like family member 4 (SLITRK4) plays an important role in the nervous system, such as synapse formation. Our study aimed to explore the functional role of SLITRK4 in GC and liver metastasis. METHODS The mRNA level of SLITRK4 was evaluated using publicly available transcriptome GEO datasets and Renji cohort. The protein level of SLITRK4 in the tissue microarray of GC was observed using immunohistochemistry. Cell Counting Kit-8, colony formation, transwell migration assays in vitro and mouse model of liver metastasis in vivo was performed to investigate the functional roles of SLITRK4 in GC. Bioinformatics predictions and Co-IP experiments were applied to screen and identify SLITRK4-binding proteins. Western blot was performed to detect Tyrosine Kinase receptor B (TrkB)-related signaling molecules. RESULTS By comparing primary and liver metastases from GC, SLITRK4 was found to be upregulated in tissues of GC with liver metastasis and to be closely related to poor clinical prognosis. SLITRK4 knockdown significantly abrogated the growth, invasion, and metastasis of GC in vitro and in vivo. Further study revealed that SLITRK4 could interact with Canopy FGF Signalling Regulator 3 (CNPY3), thus enhancing TrkB- related signaling by promoting the endocytosis and recycling of the TrkB receptor. CONCLUSION In conclusion, the CNPY3-SLITRK4 axis contributes to liver metastasis of GC according to the TrkB-related signaling pathway. which may be a therapeutic target for the treatment of GC with liver metastasis.
Collapse
Affiliation(s)
- Yao-Qi Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Tian-Shang Bao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jia-Xuan Xie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Si-Te Yu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Pei-Qi Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Wan-Zhen Zhou
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang-Yang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Su-Yuan Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xiao-Qi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Ming-Ze Ma
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
7
|
Yu F, Zhao X, Li M, Meng M. SLITRK6 promotes the progression of lung adenocarcinoma by regulating PI3K/AKT/mTOR signaling and Warburg effect. Apoptosis 2023:10.1007/s10495-023-01838-0. [PMID: 37219677 DOI: 10.1007/s10495-023-01838-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/24/2023]
Abstract
To investigate the role of SLITRK6 in lung adenocarcinoma (LUAD) and the underlying mechanism in it, clinical tissues and tissue microarray of LUAD were used to detect the expression of SLITRK6. In vitro cell viability assay and colony formation assay in LUAD cells were conducted to investigate SLITRK6 related biological functions. In vivo subcutaneous model was used to determine the role of SLITRK6 in LUAD growth. It was found that the expression of SLITRK6 was significantly upregulated in LUAD tissues compared with that in para-cancerous tissues. Knockdown of SLITRK6 suppressed the proliferation and colony formation of LUAD cells in vitro. Meanwhile, the growth of LUAD cells was also inhibited by SLITRK6 knockdown in vivo. Furthermore, we found that SLITRK6 knockdown could suppress the glycolysis of LUAD cells by regulating the phosphorylation of AKT and mTOR. All results suggest that SLITRK6 promotes LUAD cell proliferation and colony formation by regulating PI3K/AKT/mTOR signaling and Warburg effect. SLITRK6 may serve as a potential therapeutic target for LUAD in future.
Collapse
Affiliation(s)
- Fangyuan Yu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong Province, China
| | - Xinya Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mingtao Li
- Huake Medical Technology Co., Ltd, Shenzhen, China
| | - Min Meng
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong Province, China.
| |
Collapse
|
8
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538959. [PMID: 37205520 PMCID: PMC10187231 DOI: 10.1101/2023.05.01.538959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data obtained from fetal mouse brains 3 hr after VPA administration revealed that VPA significantly [p(FDR) ≤ 0.025] increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of genes associated with neurodevelopmental disorders such as autism as well as neurogenesis, axon growth and synaptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 400 autism risk genes was significantly altered by VPA as was expression of 247 genes that have been reported to play fundamental roles in the development of the nervous system, but are not linked to autism by GWAS. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria provides potential targets for future hypothesis-driven approaches to elucidating the proximal underlying causes of defective brain connectivity in neurodevelopmental disorders such as autism.
Collapse
|
9
|
Hatayama M, Aruga J. Developmental control of noradrenergic system by SLITRK1 and its implications in the pathophysiology of neuropsychiatric disorders. Front Mol Neurosci 2023; 15:1080739. [PMID: 36683853 PMCID: PMC9846221 DOI: 10.3389/fnmol.2022.1080739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
SLITRK1 is a neuronal transmembrane protein with neurite development-and synaptic formation-controlling abilities. Several rare variants of SLITRK1 have been identified and implicated in the pathogenesis of Tourette's syndrome, trichotillomania, and obsessive-compulsive disorder, which can be collectively referred to as obsessive-compulsive-spectrum disorders. Recent studies have reported a possible association between bipolar disorder and schizophrenia, including a revertant of modern human-specific amino acid residues. Although the mechanisms underlying SLITRK1-associated neuropsychiatric disorders are yet to be fully clarified, rodent studies may provide some noteworthy clues. Slitrk1-deficient mice show neonatal dysregulation of the noradrenergic system, and later, anxiety-like behaviors that can be attenuated by an alpha 2 noradrenergic receptor agonist. The noradrenergic abnormality is characterized by the excessive growth of noradrenergic fibers and increased noradrenaline content in the medial prefrontal cortex, concomitant with enlarged serotonergic varicosities. Slitrk1 has both cell-autonomous and cell-non-autonomous functions in controlling noradrenergic fiber development, and partly alters Sema3a-mediated neurite control. These findings suggest that transiently enhanced noradrenergic signaling during the neonatal stage could cause neuroplasticity associated with neuropsychiatric disorders. Studies adopting noradrenergic signal perturbation via pharmacological or genetic means support this hypothesis. Thus, Slitrk1 is a potential candidate genetic linkage between the neonatal noradrenergic signaling and the pathophysiology of neuropsychiatric disorders involving anxiety-like or depression-like behaviors.
Collapse
|
10
|
Shultz SR, Shah AD, Huang C, Dill LK, Schittenhelm RB, Morganti-Kossmann MC, Semple BD. Temporal proteomics of human cerebrospinal fluid after severe traumatic brain injury. J Neuroinflammation 2022; 19:291. [PMID: 36482407 PMCID: PMC9730674 DOI: 10.1186/s12974-022-02654-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
The pathophysiology of traumatic brain injury (TBI) requires further characterization to fully elucidate changes in molecular pathways. Cerebrospinal fluid (CSF) provides a rich repository of brain-associated proteins. In this retrospective observational study, we implemented high-resolution mass spectrometry to evaluate changes to the CSF proteome after severe TBI. 91 CSF samples were analyzed with mass spectrometry, collected from 16 patients with severe TBI (mean 32 yrs; 81% male) on day 0, 1, 2, 4, 7 and/or 10 post-injury (8-16 samples/timepoint) and compared to CSF obtained from 11 non-injured controls. We quantified 1152 proteins with mass spectrometry, of which approximately 80% were associated with CSF. 1083 proteins were differentially regulated after TBI compared to control samples. The most highly-upregulated proteins at each timepoint included neutrophil elastase, myeloperoxidase, cathepsin G, matrix metalloproteinase-8, and S100 calcium-binding proteins A8, A9 and A12-all proteins involved in neutrophil activation, recruitment, and degranulation. Pathway enrichment analysis confirmed the robust upregulation of proteins associated with innate immune responses. Conversely, downregulated pathways included those involved in nervous system development, and several proteins not previously identified after TBI such as testican-1 and latrophilin-1. We also identified 7 proteins (GM2A, Calsyntenin 1, FAT2, GANAB, Lumican, NPTX1, SFRP2) positively associated with an unfavorable outcome at 6 months post-injury. Together, these findings highlight the robust innate immune response that occurs after severe TBI, supporting future studies to target neutrophil-related processes. In addition, the novel proteins we identified to be differentially regulated by severe TBI warrant further investigation as potential biomarkers of brain damage or therapeutic targets.
Collapse
Affiliation(s)
- Sandy R. Shultz
- grid.1002.30000 0004 1936 7857Department of Neuroscience, Monash University, Melbourne, VIC Australia ,grid.267362.40000 0004 0432 5259Alfred Health, Prahran, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC Australia ,grid.267756.70000 0001 2183 6550Health and Human Services, Vancouver Island University, Nanaimo, Canada
| | - Anup D. Shah
- grid.1002.30000 0004 1936 7857Monash Proteomics and Metabolomics Facility, Monash University, Clayton, VIC Australia ,grid.1002.30000 0004 1936 7857Monash Bioinformatics Platform, Monash University, Clayton, VIC Australia
| | - Cheng Huang
- grid.1002.30000 0004 1936 7857Monash Proteomics and Metabolomics Facility, Monash University, Clayton, VIC Australia
| | - Larissa K. Dill
- grid.1002.30000 0004 1936 7857Department of Neuroscience, Monash University, Melbourne, VIC Australia ,grid.267362.40000 0004 0432 5259Alfred Health, Prahran, VIC Australia ,grid.482226.80000 0004 0437 5686The Perron Institute for Neurological and Translational Science, Nedlands, WA 6009 Australia
| | - Ralf B. Schittenhelm
- grid.1002.30000 0004 1936 7857Monash Proteomics and Metabolomics Facility, Monash University, Clayton, VIC Australia
| | - M. Cristina Morganti-Kossmann
- grid.1002.30000 0004 1936 7857Department of Epidemiology & Preventive Medicine, Monash University, Prahran, VIC Australia ,grid.427785.b0000 0001 0664 3531Department of Child Health, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ USA ,grid.134563.60000 0001 2168 186XUniversity of Arizona College of Medicine, Phoenix, AZ USA
| | - Bridgette D. Semple
- grid.1002.30000 0004 1936 7857Department of Neuroscience, Monash University, Melbourne, VIC Australia ,grid.267362.40000 0004 0432 5259Alfred Health, Prahran, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
11
|
Implications of Genetic Factors and Modifiers in Autism Spectrum Disorders: a Systematic Review. REVIEW JOURNAL OF AUTISM AND DEVELOPMENTAL DISORDERS 2022. [DOI: 10.1007/s40489-022-00333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
12
|
Liu Y, Zhang L, Mei R, Ai M, Pang R, Xia D, Chen L, Zhong L. The Role of SliTrk5 in Central Nervous System. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4678026. [PMID: 35872846 PMCID: PMC9303146 DOI: 10.1155/2022/4678026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 06/06/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022]
Abstract
SLIT and NTRK-like protein-5 (SliTrk5) is one of the six members of SliTrk protein family, which is widely expressed in the central nervous system (CNS), regulating and participating in many essential steps of central nervous system development, including axon and dendritic growth, neuron differentiation, and synaptogenesis. SliTrk5, as a neuron transmembrane protein, contains two important conservative domains consisting of leucine repeats (LRRs) located at the amino terminal in the extracellular region and tyrosine residues (Tyr) located at the carboxyl terminal in the intracellular domains. These special structures make SliTrk5 play an important role in the pathological process of the CNS. A large number of studies have shown that SliTrk5 may be involved in the pathogenesis of CNS diseases, such as obsessive-compulsive-disorder (OCD), attention deficit/hyperactivity disorder (ADHD), glioma, autism spectrum disorders (ASDs), and Parkinson's disease (PD). Targeting SliTrk5 is expected to become a new target for the treatment of CNS diseases, promoting the functional recovery of CNS. The purpose of this article is to review the current research progression of the role of SliTrk5 in CNS and its potential mechanisms in CNS diseases.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Linming Zhang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan 650032, China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China
| | - Mingda Ai
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Ruijing Pang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Di Xia
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Ling Chen
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan 650032, China
| | - Lianmei Zhong
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China
| |
Collapse
|
13
|
Kajs BL, van Roessel PJ, Davis GL, Williams LM, Rodriguez CI, Gunaydin LA. Valence processing alterations in SAPAP3 knockout mice and human OCD. J Psychiatr Res 2022; 151:657-666. [PMID: 35661523 DOI: 10.1016/j.jpsychires.2022.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 10/18/2022]
Abstract
Abnormalities in valence processing - the processing of aversive or appetitive stimuli - may be an underrecognized component of obsessive-compulsive disorder (OCD). Preclinical rodent models have been critical in furthering pathophysiological understanding of OCD, yet there is a dearth of investigations examining whether rodent models of compulsive behavior show alterations in valence systems congruent with those seen in individuals with OCD. In this study, we sought to assess valence processing in a preclinical rodent model of compulsive behavior, the SAPAP3 knockout (KO) mouse model, and compare our preclinical findings to similar behavioral phenomena in OCD patients. In SAPAP3 KO mice, we used auditory fear conditioning and extinction to examine alterations in negative valence processing and reward-based operant conditioning to examine alterations in positive valence processing. We find that SAPAP3 KO mice show evidence of heightened negative valence processing through enhanced fear learning and impaired fear extinction. SAPAP3 KO mice also show deficits in reward acquisition and goal-directed behavior, suggesting impaired positive valence processing. In OCD patients, we used validated behavioral tests to assess explicit and implicit processing of fear-related facial expressions (negative valence) and socially-rewarding happy expressions (positive valence). We find similar trends towards enhanced negative and impaired positive valence processing in OCD patients. Overall, our results reveal valence processing abnormalities in a preclinical rodent model of compulsive behavior similar to those seen in OCD patients, with implications for valence processing alterations as novel therapeutic targets across a translational research spectrum.
Collapse
Affiliation(s)
- Bridget L Kajs
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94158, USA
| | - Peter J van Roessel
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Sierra Pacific Mental Illness Research, Education, and Clinical Center (MIRECC), VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Gwynne L Davis
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94158, USA
| | - Leanne M Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Sierra Pacific Mental Illness Research, Education, and Clinical Center (MIRECC), VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Carolyn I Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| | - Lisa A Gunaydin
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
14
|
New Insights on Gene by Environmental Effects of Drugs of Abuse in Animal Models Using GeneNetwork. Genes (Basel) 2022; 13:genes13040614. [PMID: 35456420 PMCID: PMC9024903 DOI: 10.3390/genes13040614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/18/2022] Open
Abstract
Gene-by-environment interactions are important for all facets of biology, especially behaviour. Families of isogenic strains of mice, such as the BXD strains, are excellently placed to study these interactions, as the same genome can be tested in multiple environments. BXD strains are recombinant inbred mouse strains derived from crossing two inbred strains—C57BL/6J and DBA/2J mice. Many reproducible genometypes can be leveraged, and old data can be reanalysed with new tools to produce novel insights. We obtained drug and behavioural phenotypes from Philip et al. Genes, Brain and Behaviour 2010, and reanalysed their data with new genotypes from sequencing, as well as new models (Genome-wide Efficient Mixed Model Association (GEMMA) and R/qtl2). We discovered QTLs on chromosomes 3, 5, 9, 11, and 14, not found in the original study. We reduced the candidate genes based on their ability to alter gene expression or protein function. Candidate genes included Slitrk6 and Cdk14. Slitrk6, in a Chromosome14 QTL for locomotion, was found to be part of a co-expression network involved in voluntary movement and associated with neuropsychiatric phenotypes. Cdk14, one of only three genes in a Chromosome5 QTL, is associated with handling induced convulsions after ethanol treatment, that is regulated by the anticonvulsant drug valproic acid. By using families of isogenic strains, we can reanalyse data to discover novel candidate genes involved in response to drugs of abuse.
Collapse
|
15
|
Kuo FC, Huang YC, Yen MR, Lee CH, Hsu KF, Yang HY, Wu LW, Lu CH, Hsu YJ, Chen PY. Aberrant overexpression of HOTAIR inhibits abdominal adipogenesis through remodelling of genome-wide DNA methylation and transcription. Mol Metab 2022; 60:101473. [PMID: 35292404 PMCID: PMC9034304 DOI: 10.1016/j.molmet.2022.101473] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Objective Abdominal adiposity is strongly associated with diabetic and cardiovascular comorbidities. The long noncoding RNA HOTAIR (HOX Transcript Antisense Intergenic RNA) is an important epigenetic regulator with fat depot-specific expression. Its functional roles and epigenetic regulation in abdominal adipogenesis remain uncertain. Methods We collected different fat depots from healthy, severely obese, and uraemic subjects to measure fat-depot specific gene expression and quantify regional adiposity via dual-energy X-ray absorptiometry (DXA). HOTAIR was overexpressed to evaluate its functional roles. Reduced representation bisulfite sequencing (RRBS), RNA-sequencing, real-time qPCR and RNA/chromatin immunoprecipitation were performed to analyse HOTAIR-mediated epigenetic regulation. Results A negative correlation between adipose tissue HOTAIR expression (arm or abdominal subcutaneous fat depots) and regional adiposity under the status of severe obesity or uraemia was observed. HOTAIR overexpression using human immortalized abdominal preadipocytes further revealed its anti-adipogenic effects. Integrative analysis of genome-wide DNA methylation by reduced representation bisulfite sequencing (RRBS) and gene expression was performed. Overall, the differentially methylated genes were functionally enriched for nervous system development, suggesting that HOTAIR may be epigenetically associated with cell lineage commitment. We specifically found that HOTAIR-mediated genes showed strong changes in both DNA methylation and gene expression during abdominal adipogenesis. We observed that two HOTAIR-repressed genes, SLITRK4 and PITPNC1, present an obesity-driven fat-depot specific expression pattern that is positively correlated with the central body fat distribution. Conclusions Our study indicated that HOTAIR is a key regulator of abdominal adipogenesis via intricate DNA methylation and is likely to be associated with the transcriptional regulation of genes involved in nervous system development and lipid metabolism, such as SLITRK4 and PITPNC1. HOTAIR was lowly expressed in abdominal and arm fats compared to the gluteal fat. Fat-depot-specific HOTAIR expression could be altered in the obese or uraemic status. HOTAIR overexpression suppressed abdominal adipogenesis and modulated methylome. HOTAIR-suppressed genes were associated with neural development and lipid metabolism.
Collapse
|
16
|
Identification of Novel Gene Variants for Autism Spectrum Disorders in the Lebanese Population Using Whole-Exome Sequencing. Genes (Basel) 2022; 13:genes13020186. [PMID: 35205231 PMCID: PMC8871811 DOI: 10.3390/genes13020186] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
In our previous study, in which array CGH was used on 19 Lebanese ASD subjects and their parents, we identified rare copy number variants (CNVs) in 14 subjects. The five remaining subjects did not show any CNVs related to autism spectrum disorders (ASD). In the present complementary study, we applied whole-exome sequencing (WES), which allows the identification of rare genetic variations such as single nucleotide variations and small insertions/deletions, to the five negative CNV subjects. After stringent filtering of initial data on the five families, three novel genes potentially related to neurodevelopment were identified, including a de novo mutation in the MIS18BP1 gene. In addition, genes already known to be related to ASD contained sequence variations. Our findings outline the potential involvement of the novel de novo mutation in the MIS18BP1 gene in the genetic etiology and pathophysiology of ASD and highlights the genetic complexity of these disorders. Further studies with larger cohorts of subjects are needed to confirm these observations, and functional analyses need to be performed to understand the precise pathophysiology in these cases.
Collapse
|
17
|
Deng M, Wang Y, Yu S, Fan Q, Qiu J, Wang Z, Xiao Z. Exploring Association Between Serotonin and Neurogenesis Related Genes in Obsessive-Compulsive Disorder in Chinese Han People: Promising Association Between DMRT2, miR-30a-5p, and Early-Onset Patients. Front Psychiatry 2022; 13:857574. [PMID: 35633798 PMCID: PMC9137639 DOI: 10.3389/fpsyt.2022.857574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Obsessive-compulsive disorder (OCD) is a deliberating disorder with complex genetic and environmental etiologies. Hypotheses about OCD mainly include dysregulated neurotransmitters, especially serotonin, and disturbed neurodevelopment. Single nucleotide polymorphism (SNP) association studies regarding OCD are often met with inconsistent results. However, stratification by age of onset may sometimes help to limit the heterogenicity of OCD patients. Therefore, we conducted a stratified SNP association study enrolling 636 patients and 612 healthy controls. Patients were stratified by age of onset as early-onset (EO-OCD) and late-onset (LO-OCD). Blood extracted from the patients was used to genotype 18 loci, including serotonin system genes, Slitrk1, Slitrk5, and DMRT2 and related miRNA genes. Logistic regression was used to compare allele and genotype frequencies of variants. A general linear model was used to evaluate the association between variants and trait anxiety. In our study, rs3824419 in DMRT2 was associated with EO-OCD, G allele was the risk allele. Rs2222722 in miR-30a-5p was associated with EO-OCD, with the C allele being the risk allele. Rs1000952 in HTR3D was found associated with trait anxiety in OCD patients. The significance disappeared after FDR correction. Our results supported neurodevelopment-related genes, DMRT2 and miR-30a-5p, to be related to EO-OCD. However, we cannot prove serotonin genes to be directly associated with EO-OCD. While an association between HTR3D and trait anxiety was discovered, comparisons based on biological or clinical traits may be helpful in future studies. As our detective powers were limited, more large-scale studies will be needed to confirm our conclusion.
Collapse
Affiliation(s)
- Miaohan Deng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunying Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Fan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyin Qiu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeping Xiao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Hay M, Kumar V, Ricaño-Ponce I. The role of the X chromosome in infectious diseases. Brief Funct Genomics 2021; 21:143-158. [PMID: 34651167 DOI: 10.1093/bfgp/elab039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023] Open
Abstract
Many infectious diseases in humans present with a sex bias. This bias arises from a combination of environmental factors, hormones and genetics. In this study, we review the contribution of the X chromosome to the genetic factor associated with infectious diseases. First, we give an overview of the X-linked genes that have been described in the context of infectious diseases and group them in four main pathways that seem to be dysregulated in infectious diseases: nuclear factor kappa-B, interleukin 2 and interferon γ cascade, toll-like receptors and programmed death ligand 1. Then, we review the infectious disease associations in existing genome-wide association studies (GWAS) from the GWAS Catalog and the Pan-UK Biobank, describing the main associations and their possible implications for the disease. Finally, we highlight the importance of including the X chromosome in GWAS analysis and the importance of sex-specific analysis.
Collapse
|
19
|
Levy AM, Paschou P, Tümer Z. Candidate Genes and Pathways Associated with Gilles de la Tourette Syndrome-Where Are We? Genes (Basel) 2021; 12:1321. [PMID: 34573303 PMCID: PMC8468358 DOI: 10.3390/genes12091321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/05/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022] Open
Abstract
Gilles de la Tourette syndrome (GTS) is a childhood-onset neurodevelopmental and -psychiatric tic-disorder of complex etiology which is often comorbid with obsessive-compulsive disorder (OCD) and/or attention deficit hyperactivity disorder (ADHD). Twin and family studies of GTS individuals have shown a high level of heritability suggesting, that genetic risk factors play an important role in disease etiology. However, the identification of major GTS susceptibility genes has been challenging, presumably due to the complex interplay between several genetic factors and environmental influences, low penetrance of each individual factor, genetic diversity in populations, and the presence of comorbid disorders. To understand the genetic components of GTS etiopathology, we conducted an extensive review of the literature, compiling the candidate susceptibility genes identified through various genetic approaches. Even though several strong candidate genes have hitherto been identified, none of these have turned out to be major susceptibility genes yet.
Collapse
Affiliation(s)
- Amanda M. Levy
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark;
| | - Peristera Paschou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Zeynep Tümer
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
20
|
Ueda K, Black KJ. A Comprehensive Review of Tic Disorders in Children. J Clin Med 2021; 10:2479. [PMID: 34204991 PMCID: PMC8199885 DOI: 10.3390/jcm10112479] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 01/13/2023] Open
Abstract
Tics are characterized by sudden, rapid, recurrent, nonrhythmic movement or vocalization, and are the most common movement disorders in children. Their onset is usually in childhood and tics often will diminish within one year. However, some of the tics can persist and cause various problems such as social embarrassment, physical discomfort, or emotional impairments, which could interfere with daily activities and school performance. Furthermore, tic disorders are frequently associated with comorbid neuropsychiatric symptoms, which can become more problematic than tic symptoms. Unfortunately, misunderstanding and misconceptions of tic disorders still exist among the general population. Understanding tic disorders and their comorbidities is important to deliver appropriate care to patients with tics. Several studies have been conducted to elucidate the clinical course, epidemiology, and pathophysiology of tics, but they are still not well understood. This article aims to provide an overview about tics and tic disorders, and recent findings on tic disorders including history, definition, diagnosis, epidemiology, etiology, diagnostic approach, comorbidities, treatment and management, and differential diagnosis.
Collapse
Affiliation(s)
- Keisuke Ueda
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Kevin J. Black
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
MiR-139-5p influences hepatocellular carcinoma cell invasion and proliferation capacities via decreasing SLITRK4 expression. Biosci Rep 2021; 40:222640. [PMID: 32285917 PMCID: PMC7199452 DOI: 10.1042/bsr20193295] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 02/29/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
The microRNA, miR-139-5p, has been proved to play important roles in regulating tumor progression, including prostate cancer, osteosarcoma, esophageal cancer, and so on, but its correlation of hepatocellular carcinoma (HCC) still remains unclear. Here we found that hsa-miR-139-5p (miR-139-5p) was decreased in HCC samples compared with normal liver tissues, and a lower expression of miR-139-5p was connected to a poorer prognosis. Mechanism study indicated that a decreased/increased miR-139-5p could increase/decrease HCC cells invasion and proliferation capacities via increasing SLITRK4 expression, what’s more, the reverse assays also confirmed the conclusion when we knocked down SLITRK4 in the miR-139-5p low-expression cells. Luciferase assay confirmed that miR-139-5p could directly bind to the 3′UTR of SLITRK4 mRNA to regulate its expression. Together, these findings show the importance of miR-139-5p/SLITRK4 pathway in HCC growth and progression and may provide new targets for us to better arrange the progression of HCC.
Collapse
|
22
|
Salesse C, Charest J, Doucet-Beaupré H, Castonguay AM, Labrecque S, De Koninck P, Lévesque M. Opposite Control of Excitatory and Inhibitory Synapse Formation by Slitrk2 and Slitrk5 on Dopamine Neurons Modulates Hyperactivity Behavior. Cell Rep 2021; 30:2374-2386.e5. [PMID: 32075770 DOI: 10.1016/j.celrep.2020.01.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 12/03/2019] [Accepted: 01/24/2020] [Indexed: 11/26/2022] Open
Abstract
The neurodevelopmental origin of hyperactivity disorder has been suggested to involve the dopaminergic system, but the underlying mechanisms are still unknown. Here, transcription factors Lmx1a and Lmx1b are shown to be essential for midbrain dopaminergic (mDA) neuron excitatory synaptic inputs and dendritic development. Strikingly, conditional knockout (cKO) of Lmx1a/b in postmitotic mDA neurons results in marked hyperactivity. In seeking Lmx1a/b target genes, we identify positively regulated Slitrk2 and negatively regulated Slitrk5. These two synaptic adhesion proteins promote excitatory and inhibitory synapses on mDA neurons, respectively. Knocking down Slitrk2 reproduces some of the Lmx1a/b cKO cellular and behavioral phenotypes, whereas Slitrk5 knockdown has opposite effects. The hyperactivity caused by this imbalance in excitatory/inhibitory synaptic inputs on dopamine neurons is reproduced by chronically inhibiting the ventral tegmental area during development using pharmacogenetics. Our study shows that alterations in developing dopaminergic circuits strongly impact locomotor activity, shedding light on mechanisms causing hyperactivity behaviors.
Collapse
Affiliation(s)
- Charleen Salesse
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Julien Charest
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | | | | | - Simon Labrecque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Paul De Koninck
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC G1V 0A6, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
23
|
Urh K, Žlajpah M, Zidar N, Boštjančič E. Identification and Validation of New Cancer Stem Cell-Related Genes and Their Regulatory microRNAs in Colorectal Cancerogenesis. Biomedicines 2021; 9:biomedicines9020179. [PMID: 33670246 PMCID: PMC7916981 DOI: 10.3390/biomedicines9020179] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Significant progress has been made in the last decade in our understanding of the pathogenetic mechanisms of colorectal cancer (CRC). Cancer stem cells (CSC) have gained much attention and are now believed to play a crucial role in the pathogenesis of various cancers, including CRC. In the current study, we validated gene expression of four genes related to CSC, L1TD1, SLITRK6, ST6GALNAC1 and TCEA3, identified in a previous bioinformatics analysis. Using bioinformatics, potential miRNA-target gene correlations were prioritized. In total, 70 formalin-fixed paraffin-embedded biopsy samples from 47 patients with adenoma, adenoma with early carcinoma and CRC without and with lymph node metastases were included. The expression of selected genes and microRNAs (miRNAs) was evaluated using quantitative PCR. Differential expression of all investigated genes and four of six prioritized miRNAs (hsa-miR-199a-3p, hsa-miR-335-5p, hsa-miR-425-5p, hsa-miR-1225-3p, hsa-miR-1233-3p and hsa-miR-1303) was found in at least one group of CRC cancerogenesis. L1TD1, SLITRK6, miR-1233-3p and miR-1225-3p were correlated to the level of malignancy. A negative correlation between miR-199a-3p and its predicted target SLITRK6 was observed, showing potential for further experimental validation in CRC. Our results provide further evidence that CSC-related genes and their regulatory miRNAs are involved in CRC development and progression and suggest that some them, particularly miR-199a-3p and its SLITRK6 target gene, are promising for further validation in CRC.
Collapse
|
24
|
Kim HY, Um JW, Ko J. Proper synaptic adhesion signaling in the control of neural circuit architecture and brain function. Prog Neurobiol 2021; 200:101983. [PMID: 33422662 DOI: 10.1016/j.pneurobio.2020.101983] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Trans-synaptic cell-adhesion molecules are critical for governing various stages of synapse development and specifying neural circuit properties via the formation of multifarious signaling pathways. Recent studies have pinpointed the putative roles of trans-synaptic cell-adhesion molecules in mediating various cognitive functions. Here, we review the literature on the roles of a diverse group of central synaptic organizers, including neurexins (Nrxns), leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs), and their associated binding proteins, in regulating properties of specific type of synapses and neural circuits. In addition, we highlight the findings that aberrant synaptic adhesion signaling leads to alterations in the structures, transmission, and plasticity of specific synapses across diverse brain areas. These results seem to suggest that proper trans-synaptic signaling pathways by Nrxns, LAR-RPTPs, and their interacting network is likely to constitute central molecular complexes that form the basis for cognitive functions, and that these complexes are heterogeneously and complexly disrupted in many neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea; Core Protein Resources Center, DGIST, Daegu, 42988, South Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea.
| |
Collapse
|
25
|
Teo YYA, Danilevsky A, Shomron N. Overcoming Interpretability in Deep Learning Cancer Classification. Methods Mol Biol 2021; 2243:297-309. [PMID: 33606264 DOI: 10.1007/978-1-0716-1103-6_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since its inception, deep learning has revolutionized the field of machine learning and data-driven science. One such data-driven science to be transformed by deep learning is genomics. In the past decade, numerous genomics studies have adopted deep learning and its applications range from predicting regulatory elements to cancer classification. Despite its dominating efficacy in these applications, deep learning is not without drawbacks. A prominent shortcoming of deep learning is the lack of interpretability. Hence, the main objective of this study is to address this obstacle in the deep learning cancer classification. Here we adopt a feature importance scoring methodology (Gradient-based class activation mapping or Grad-CAM) on a quasi-recurrent neural network model that classify cancer based on FASTA sequencing data. In this study, we managed to formulate a nucleotide-to-genomic-region Grad-CAM scoring methodology, as well as, validate the use this methodology for the chosen model. Consequently, this allows for the utilization of the Grad-CAM scoring methodology for feature importance in deep learning cancer classification. The results from our study identify potential novel candidate genes, genomic elements, and mechanisms for future cancer research.
Collapse
Affiliation(s)
| | | | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
26
|
Loomis C, Stephens A, Janicot R, Baqai U, Drebushenko L, Round J. Identification of MAGUK scaffold proteins as intracellular binding partners of synaptic adhesion protein Slitrk2. Mol Cell Neurosci 2020; 103:103465. [DOI: 10.1016/j.mcn.2019.103465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 01/10/2023] Open
|
27
|
Abstract
Movement disorders in women during pregnancy are uncommon. Therefore, high quality studies are limited, and guidelines are lacking for the treatment of movement disorders in pregnancy, thus posing a significant therapeutic challenge for the treating physicians. In this chapter, we discuss movement disorders that arise during pregnancy and the preexisting movement disorders during pregnancy. Common conditions encountered in pregnancy include but are not limited to restless legs syndrome, chorea gravidarum, Parkinson disease, essential tremor, and Huntington disease as well as more rare movement disorders (Wilson's disease, dystonia, etc.). This chapter summarizes the published literature on movement disorders and pharmacologic and surgical considerations for neurologists and physicians in other specialties caring for patients who are pregnant or considering pregnancy.
Collapse
Affiliation(s)
- Fang Ba
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Janis M Miyasaki
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
28
|
Depienne C, Ciura S, Trouillard O, Bouteiller D, Leitão E, Nava C, Keren B, Marie Y, Guegan J, Forlani S, Brice A, Anheim M, Agid Y, Krack P, Damier P, Viallet F, Houeto JL, Durif F, Vidailhet M, Worbe Y, Roze E, Kabashi E, Hartmann A. Association of Rare Genetic Variants in Opioid Receptors with Tourette Syndrome. TREMOR AND OTHER HYPERKINETIC MOVEMENTS (NEW YORK, N.Y.) 2019; 9:tre-09-693. [PMID: 31824749 PMCID: PMC6878848 DOI: 10.7916/tohm.v0.693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/15/2019] [Indexed: 01/07/2023]
Abstract
Background Genes involved in Tourette syndrome (TS) remain largely unknown. We aimed to identify genetic factors contributing to TS in a French cohort of 120 individuals using a combination of hypothesis-driven and exome-sequencing approaches. Methods We first sequenced exons of SLITRK1-6 and HDC in the TS cohort and subsequently sequenced the exome of 12 individuals harboring rare variants in these genes to find additional rare variants contributing to the disorder under the hypothesis of oligogenic inheritance. We further screened three candidate genes (OPRK1, PCDH10, and NTSR2) preferentially expressed in the basal ganglia, and three additional genes involved in neurotensin and opioid signaling (OPRM1, NTS, and NTSR1), and compared variant frequencies in TS patients and 788 matched control individuals. We also investigated the impact of altering the expression of Oprk1 in zebrafish. Results Thirteen ultrarare missense variants of SLITRK1-6 and HDC were identified in 12 patients. Exome sequencing in these patients revealed rare possibly deleterious variants in 3,041 genes, 54 of which were preferentially expressed in the basal ganglia. Comparison of variant frequencies altering selected candidate genes in TS and control individuals revealed an excess of potentially disrupting variants in OPRK1, encoding the opioid kappa receptor, in TS patients. Accordingly, we show that downregulation of the Oprk1 orthologue in zebrafish induces a hyperkinetic phenotype in early development. Discussion These results support a heterogeneous and complex genetic etiology of TS, possibly involving rare variants altering the opioid pathway in some individuals, which could represent a novel therapeutic target in this disorder.
Collapse
Affiliation(s)
- Christel Depienne
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, DE
| | - Sorana Ciura
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Oriane Trouillard
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Delphine Bouteiller
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Elsa Leitão
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, DE
| | - Caroline Nava
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département de Génétique, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Boris Keren
- Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département de Génétique, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Yannick Marie
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Justine Guegan
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Sylvie Forlani
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Alexis Brice
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Mathieu Anheim
- Service de neurologie, CHU de Strasbourg, Hôpital de Hautepierre, Avenue Molière, 67200 Strasbourg Strasbourg, FR
| | - Yves Agid
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Paul Krack
- Service de Neurologie, CHU de Grenoble, Avenue Maquis du Grésivaudan, 38700 La Tronche, FR.,Center for Movement Disorders, Inselspital, University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Philippe Damier
- Service de Neurologie, CHU de Nantes, 5 Allée de l'Île Gloriette, 44093 Nantes, FR
| | - François Viallet
- Service de Neurologie, CRHU d'Aix-en-Provence, Avenue des Tamaris, 13100 Aix-en-Provence, FR
| | - Jean-Luc Houeto
- Service de Neurologie, CHU de Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, FR
| | - Franck Durif
- Service de Neurologie, CHU de Clermont-Ferrand, CHU de Clermont-Ferrand, Hôpital Gabriel Montpied, 58 rue Montalembert, 63003 Clermont-Ferrand, FR
| | - Marie Vidailhet
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Yulia Worbe
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Emmanuel Roze
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Edor Kabashi
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Andreas Hartmann
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| |
Collapse
|
29
|
Han KA, Kim J, Kim H, Kim D, Lim D, Ko J, Um JW. Slitrk2 controls excitatory synapse development via PDZ-mediated protein interactions. Sci Rep 2019; 9:17094. [PMID: 31745231 PMCID: PMC6863843 DOI: 10.1038/s41598-019-53519-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/31/2019] [Indexed: 01/09/2023] Open
Abstract
Members of the Slitrk (Slit- and Trk-like protein) family of synaptic cell-adhesion molecules control excitatory and inhibitory synapse development through isoform-dependent extracellular interactions with leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs). However, how Slitrks participate in activation of intracellular signaling pathways in postsynaptic neurons remains largely unknown. Here we report that, among the six members of the Slitrk family, only Slitrk2 directly interacts with the PDZ domain-containing excitatory scaffolds, PSD-95 and Shank3. The interaction of Slitrk2 with PDZ proteins is mediated by the cytoplasmic COOH-terminal PDZ domain-binding motif (Ile-Ser-Glu-Leu), which is not found in other Slitrks. Mapping analyses further revealed that a single PDZ domain of Shank3 is responsible for binding to Slitrk2. Slitrk2 forms in vivo complexes with membrane-associated guanylate kinase (MAGUK) family proteins in addition to PSD-95 and Shank3. Intriguingly, in addition to its role in synaptic targeting in cultured hippocampal neurons, the PDZ domain-binding motif of Slitrk2 is required for Slitrk2 promotion of excitatory synapse formation, transmission, and spine development in the CA1 hippocampal region. Collectively, our data suggest a new molecular mechanism for conferring isoform-specific regulatory actions of the Slitrk family in orchestrating intracellular signal transduction pathways in postsynaptic neurons.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Hyeonho Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Dongwook Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Dongseok Lim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-eup, Dalseong-gun, Daegu, 42988, Korea.
| |
Collapse
|
30
|
Mukai J, Cannavò E, Crabtree GW, Sun Z, Diamantopoulou A, Thakur P, Chang CY, Cai Y, Lomvardas S, Takata A, Xu B, Gogos JA. Recapitulation and Reversal of Schizophrenia-Related Phenotypes in Setd1a-Deficient Mice. Neuron 2019; 104:471-487.e12. [PMID: 31606247 DOI: 10.1016/j.neuron.2019.09.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/28/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022]
Abstract
SETD1A, a lysine-methyltransferase, is a key schizophrenia susceptibility gene. Mice carrying a heterozygous loss-of-function mutation of the orthologous gene exhibit alterations in axonal branching and cortical synaptic dynamics accompanied by working memory deficits. We show that Setd1a binds both promoters and enhancers with a striking overlap between Setd1a and Mef2 on enhancers. Setd1a targets are highly expressed in pyramidal neurons and display a complex pattern of transcriptional up- and downregulations shaped by presumed opposing functions of Setd1a on promoters and Mef2-bound enhancers. Notably, evolutionarily conserved Setd1a targets are associated with neuropsychiatric genetic risk burden. Reinstating Setd1a expression in adulthood rescues cognitive deficits. Finally, we identify LSD1 as a major counteracting demethylase for Setd1a and show that its pharmacological antagonism results in a full rescue of the behavioral and morphological deficits in Setd1a-deficient mice. Our findings advance understanding of how SETD1A mutations predispose to schizophrenia (SCZ) and point to novel therapeutic interventions.
Collapse
Affiliation(s)
- Jun Mukai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Enrico Cannavò
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Gregg W Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Ziyi Sun
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Pratibha Thakur
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Chia-Yuan Chang
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yifei Cai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Stavros Lomvardas
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
31
|
Li J, Han W, Wu K, Li YD, Liu Q, Lu W. A Conserved Tyrosine Residue in Slitrk3 Carboxyl-Terminus Is Critical for GABAergic Synapse Development. Front Mol Neurosci 2019; 12:213. [PMID: 31551708 PMCID: PMC6746929 DOI: 10.3389/fnmol.2019.00213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Single-passing transmembrane protein, Slitrk3 (Slit and Trk-like family member 3, ST3), is a synaptic cell adhesion molecule highly expressed at inhibitory synapses. Recent studies have shown that ST3, through its extracellular domain, selectively regulates inhibitory synapse development via the trans-synaptic interaction with presynaptic cell adhesion molecule, receptor protein tyrosine phosphatase δ (PTPδ) and the cis-interaction with postsynaptic cell adhesion molecule, Neuroligin 2 (NL2). However, little is known about the physiological function of ST3 intracellular, carboxyl (C)-terminal region. Here we report that in heterologous cells, ST3 C-terminus is not required for ST3 homo-dimerization and trafficking to the cell surface. In contrast, in hippocampal neurons, ST3 C-terminus, more specifically, the conserved tyrosine Y969 (in mice), is critical for GABAergic synapse development. Indeed, overexpression of ST3 Y969A mutant markedly reduced the gephyrin puncta density and GABAergic transmission in hippocampal neurons. In addition, single-cell genetic deletion of ST3 strongly impaired GABAergic transmission. Importantly, wild-type (WT) ST3, but not the ST3 Y969A mutant, could fully rescue GABAergic transmission deficits in neurons lacking endogenous ST3, confirming a critical role of Y969 in the regulation of inhibitory synapses. Taken together, our data identify a single critical residue in ST3 C-terminus that is important for GABAergic synapse development and function.
Collapse
Affiliation(s)
- Jun Li
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Wenyan Han
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Kunwei Wu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Yuping Derek Li
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Qun Liu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
32
|
Choufani S, Turinsky AL, Melamed N, Greenblatt E, Brudno M, Bérard A, Fraser WD, Weksberg R, Trasler J, Monnier P. Impact of assisted reproduction, infertility, sex and paternal factors on the placental DNA methylome. Hum Mol Genet 2019; 28:372-385. [PMID: 30239726 DOI: 10.1093/hmg/ddy321] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/29/2018] [Indexed: 12/28/2022] Open
Abstract
Children conceived using Assisted Reproductive Technologies (ART) have a higher incidence of growth and birth defects, attributable in part to epigenetic perturbations. Both ART and germline defects associated with parental infertility could interfere with epigenetic reprogramming events in germ cells or early embryos. Mouse models indicate that the placenta is more susceptible to the induction of epigenetic abnormalities than the embryo, and thus the placental methylome may provide a sensitive indicator of 'at risk' conceptuses. Our goal was to use genome-wide profiling to examine the extent of epigenetic abnormalities in matched placentas from an ART/infertility group and control singleton pregnancies (n = 44/group) from a human prospective longitudinal birth cohort, the Design, Develop, Discover (3D) Study. Principal component analysis revealed a group of ART outliers. The ART outlier group was enriched for females and a subset of placentas showing loss of methylation of several imprinted genes including GNAS, SGCE, KCNQT1OT1 and BLCAP/NNAT. Within the ART group, placentas from pregnancies conceived with in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) showed distinct epigenetic profiles as compared to those conceived with less invasive procedures (ovulation induction, intrauterine insemination). Male factor infertility and paternal age further differentiated the IVF/ICSI group, suggesting an interaction of infertility and techniques in perturbing the placental epigenome. Together, the results suggest that the human placenta is sensitive to the induction of epigenetic defects by ART and/or infertility, and we stress the importance of considering both sex and paternal factors and that some but not all ART conceptuses will be susceptible.
Collapse
Affiliation(s)
- Sanaa Choufani
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrei L Turinsky
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nir Melamed
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynaecology Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Ellen Greenblatt
- Mount Sinai Centre for Fertility and Reproductive Health, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michael Brudno
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Anick Bérard
- Research Unit on Medications and Pregnancy, Research Centre, CHU Sainte-Justine, and Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - William D Fraser
- Department of Obstetrics and Gynecology, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Rosanna Weksberg
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jacquetta Trasler
- Departments of Pediatrics, Human Genetics and Pharmacology & Therapeutics, and The Montreal Children's Hospital and Research Institute of the McGill University Health Centre
| | - Patricia Monnier
- MUHC Reproductive Centre, Department of Obstetrics and Gynecology, Royal Victoria Hospital and Research Institute of McGill University Health Centre, Quebec, Canada
| | | |
Collapse
|
33
|
Won SY, Lee P, Kim HM. Synaptic organizer: Slitrks and type IIa receptor protein tyrosine phosphatases. Curr Opin Struct Biol 2019; 54:95-103. [PMID: 30822649 DOI: 10.1016/j.sbi.2019.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
Abstract
Slit-like and Trk-like (Slitrk) family members are leucine-rich repeat (LRR)-containing neuronal transmembrane proteins. Slitrks have been highlighted as key synapse organizers at neuronal synapses through interactions with specific members of the presynaptic type IIa receptor protein tyrosine phosphatase (RPTP) family. Recent structural studies on type IIa RPTP/Slitrk1 complexes have unveiled molecular insights into their binding selectivity and have established the role of higher-order receptor clustering in their synaptogenic activity. Here, we will discuss key structural aspects of Slitrk interactions with type IIa RPTP family members, the biological roles of Slitrks in neurons, and our current knowledge of SLITRK mutations in human diseases.
Collapse
Affiliation(s)
- Seoung Youn Won
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Pedro Lee
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ho Min Kim
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea; Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| |
Collapse
|
34
|
Basavanhally T, Fonseca R, Uversky VN. Born This Way: Using Intrinsic Disorder to Map the Connections between SLITRKs, TSHR, and Male Sexual Orientation. Proteomics 2018; 18:e1800307. [PMID: 30156382 DOI: 10.1002/pmic.201800307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/03/2018] [Indexed: 12/15/2022]
Abstract
Recently, genome-wide association study reveals a significant association between specific single nucleotide polymorphisms (SNPs) in men and their sexual orientation. These SNPs (rs9547443 and rs1035144) reside in the intergenic region between the SLITRK5 and SLITRK6 genes and in the intronic region of the TSHR gene and might affect functionality of SLITRK5, SLITRK6, and TSHR proteins that are engaged in tight control of key developmental processes, such as neurite outgrowth and modulation, cellular differentiation, and hormonal regulation. SLITRK5 and SLITRK6 are single-pass transmembrane proteins, whereas TSHR is a heptahelical G protein-coupled receptor (GPCR). Mutations in these proteins are associated with various diseases and are linked to phenotypes found at a higher rate in homosexual men. A bioinformatics analysis of SLITRK5, SLITRK6, and TSHR proteins is conducted to look at their structure, protein interaction networks, and propensity for intrinsic disorder. It is assumed that this information might improve understanding of the roles that SLITRK5, SLITRK6, and TSHR play within neuronal and thyroidal tissues and give insight into the phenotypes associated with male homosexuality.
Collapse
Affiliation(s)
- Tara Basavanhally
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Renée Fonseca
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.,USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.,Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, 142290, Pushchino, Moscow, Russia
| |
Collapse
|
35
|
Bansal V, Mitjans M, Burik CAP, Linnér RK, Okbay A, Rietveld CA, Begemann M, Bonn S, Ripke S, de Vlaming R, Nivard MG, Ehrenreich H, Koellinger PD. Genome-wide association study results for educational attainment aid in identifying genetic heterogeneity of schizophrenia. Nat Commun 2018; 9:3078. [PMID: 30082721 PMCID: PMC6079028 DOI: 10.1038/s41467-018-05510-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 07/09/2018] [Indexed: 01/03/2023] Open
Abstract
Higher educational attainment (EA) is negatively associated with schizophrenia (SZ). However, recent studies found a positive genetic correlation between EA and SZ. We investigate possible causes of this counterintuitive finding using genome-wide association study results for EA and SZ (N = 443,581) and a replication cohort (1169 controls; 1067 cases) with deeply phenotyped SZ patients. We find strong genetic dependence between EA and SZ that cannot be explained by chance, linkage disequilibrium, or assortative mating. Instead, several genes seem to have pleiotropic effects on EA and SZ, but without a clear pattern of sign concordance. Using EA as a proxy phenotype, we isolate FOXO6 and SLITRK1 as novel candidate genes for SZ. Our results reveal that current SZ diagnoses aggregate over at least two disease subtypes: one part resembles high intelligence and bipolar disorder (BIP), while the other part is a cognitive disorder that is independent of BIP.
Collapse
Affiliation(s)
- V Bansal
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- Research Group for Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Straße 3A, 37075, Göttingen, Germany
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Clinic Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - M Mitjans
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Humboldtallee 23, 30703, Göttingen, Germany
| | - C A P Burik
- Complex Trait Genetics, Vrije Universiteit Amsterdam, De Boelelaan 1085 B-631, 1081 HV, Amsterdam, Netherlands
- Institute for Behavior and Biology, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, Netherlands
- School of Business and Economics, Department of Economics, De Boelelaan 1105, 1081 HV, Amsterdam, Netherlands
| | - R K Linnér
- Complex Trait Genetics, Vrije Universiteit Amsterdam, De Boelelaan 1085 B-631, 1081 HV, Amsterdam, Netherlands
- Institute for Behavior and Biology, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, Netherlands
- School of Business and Economics, Department of Economics, De Boelelaan 1105, 1081 HV, Amsterdam, Netherlands
| | - A Okbay
- Complex Trait Genetics, Vrije Universiteit Amsterdam, De Boelelaan 1085 B-631, 1081 HV, Amsterdam, Netherlands
- School of Business and Economics, Department of Economics, De Boelelaan 1105, 1081 HV, Amsterdam, Netherlands
| | - C A Rietveld
- Institute for Behavior and Biology, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, Netherlands
- Erasmus School of Economics, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, Netherlands
| | - M Begemann
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- Department of Psychiatry & Psychotherapy, University of Göttingen, Von-Siebold-Straße 5, 37075, Göttingen, Germany
| | - S Bonn
- Research Group for Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Straße 3A, 37075, Göttingen, Germany
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Clinic Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - S Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, 02114 MA, Boston, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 02142 MA, Cambridge, USA
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, 10117, Germany
| | - R de Vlaming
- Complex Trait Genetics, Vrije Universiteit Amsterdam, De Boelelaan 1085 B-631, 1081 HV, Amsterdam, Netherlands
- School of Business and Economics, Department of Economics, De Boelelaan 1105, 1081 HV, Amsterdam, Netherlands
| | - M G Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, van der Boechorststraat 1, 1081 BT, Amsterdam, Netherlands
| | - H Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Humboldtallee 23, 30703, Göttingen, Germany
| | - P D Koellinger
- Complex Trait Genetics, Vrije Universiteit Amsterdam, De Boelelaan 1085 B-631, 1081 HV, Amsterdam, Netherlands.
- Institute for Behavior and Biology, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, Netherlands.
- School of Business and Economics, Department of Economics, De Boelelaan 1105, 1081 HV, Amsterdam, Netherlands.
| |
Collapse
|
36
|
Leucine-rich repeat-containing synaptic adhesion molecules as organizers of synaptic specificity and diversity. Exp Mol Med 2018; 50:1-9. [PMID: 29628503 PMCID: PMC5938020 DOI: 10.1038/s12276-017-0023-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
The brain harbors billions of neurons that form distinct neural circuits with exquisite specificity. Specific patterns of connectivity between distinct neuronal cell types permit the transfer and computation of information. The molecular correlates that give rise to synaptic specificity are incompletely understood. Recent studies indicate that cell-surface molecules are important determinants of cell type identity and suggest that these are essential players in the specification of synaptic connectivity. Leucine-rich repeat (LRR)-containing adhesion molecules in particular have emerged as key organizers of excitatory and inhibitory synapses. Here, we discuss emerging evidence that LRR proteins regulate the assembly of specific connectivity patterns across neural circuits, and contribute to the diverse structural and functional properties of synapses, two key features that are critical for the proper formation and function of neural circuits. Further analysis of synaptic proteins will provide insights into the functioning of neural circuits and associated brain disorders. The brain houses numerous highly specialized neuron types, which transfer and process information via a complex network of synaptic connections. Every neuron develops its own distinctive synapses with specific functions, but exactly how this is achieved is not clear. Joris de Wit and Anna Schroeder at the VIB Center for Brain and Disease Research in Leuven, Belgium, reviewed recent research into the leucine-rich repeat-containing (LRR) proteins, which are thought to be major organizers of synaptic connectivity and key regulators of healthy neural circuit development. Further investigations into the functionality of LRR proteins in the brain will not only improve understanding of neural circuitry but also provide insights into synaptic impairments in brain disorders like schizophrenia.
Collapse
|
37
|
Sanders AR, Beecham GW, Guo S, Dawood K, Rieger G, Badner JA, Gershon ES, Krishnappa RS, Kolundzija AB, Duan J, Gejman PV, Bailey JM, Martin ER. Genome-Wide Association Study of Male Sexual Orientation. Sci Rep 2017; 7:16950. [PMID: 29217827 PMCID: PMC5721098 DOI: 10.1038/s41598-017-15736-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/31/2017] [Indexed: 11/11/2022] Open
Abstract
Family and twin studies suggest that genes play a role in male sexual orientation. We conducted a genome-wide association study (GWAS) of male sexual orientation on a primarily European ancestry sample of 1,077 homosexual men and 1,231 heterosexual men using Affymetrix single nucleotide polymorphism (SNP) arrays. We identified several SNPs with p < 10-5, including regions of multiple supporting SNPs on chromosomes 13 (minimum p = 7.5 × 10-7) and 14 (p = 4.7 × 10-7). The genes nearest to these peaks have functions plausibly relevant to the development of sexual orientation. On chromosome 13, SLITRK6 is a neurodevelopmental gene mostly expressed in the diencephalon, which contains a region previously reported as differing in size in men by sexual orientation. On chromosome 14, TSHR genetic variants in intron 1 could conceivably help explain past findings relating familial atypical thyroid function and male homosexuality. Furthermore, skewed X chromosome inactivation has been found in the thyroid condition, Graves' disease, as well as in mothers of homosexual men. On pericentromeric chromosome 8 within our previously reported linkage peak, we found support (p = 4.1 × 10-3) for a SNP association previously reported (rs77013977, p = 7.1 × 10-8), with the combined analysis yielding p = 6.7 × 10-9, i.e., a genome-wide significant association.
Collapse
Affiliation(s)
- Alan R Sanders
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem Research Institute, Evanston, Illinois, 60201, United States of America.
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, 60637, United States of America.
| | - Gary W Beecham
- Department of Human Genetics, University of Miami, Miami, Florida, 33136, United States of America
| | - Shengru Guo
- Department of Human Genetics, University of Miami, Miami, Florida, 33136, United States of America
| | - Khytam Dawood
- Department of Psychology, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Gerulf Rieger
- Department of Psychology, University of Essex, Colchester, England, CO4 3SQ, United Kingdom
| | - Judith A Badner
- Department of Psychiatry, Rush University Medical Center, Chicago, Illinois, 60612, United States of America
| | - Elliot S Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Ritesha S Krishnappa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Elmhurst, New York, 11373, United States of America
| | - Alana B Kolundzija
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, New York, 10027, United States of America
| | - Jubao Duan
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem Research Institute, Evanston, Illinois, 60201, United States of America
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Pablo V Gejman
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem Research Institute, Evanston, Illinois, 60201, United States of America
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - J Michael Bailey
- Department of Psychology, Northwestern University, Evanston, Illinois, 60208, United States of America
| | - Eden R Martin
- Department of Human Genetics, University of Miami, Miami, Florida, 33136, United States of America
| |
Collapse
|
38
|
Qi Y, Zheng Y, Li Z, Xiong L. Progress in Genetic Studies of Tourette's Syndrome. Brain Sci 2017; 7:E134. [PMID: 29053637 PMCID: PMC5664061 DOI: 10.3390/brainsci7100134] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 10/17/2017] [Indexed: 12/23/2022] Open
Abstract
Tourette's Syndrome (TS) is a complex disorder characterized by repetitive, sudden, and involuntary movements or vocalizations, called tics. Tics usually appear in childhood, and their severity varies over time. In addition to frequent tics, people with TS are at risk for associated problems including attention deficit hyperactivity disorder (ADHD), obsessive-compulsive disorder (OCD), anxiety, depression, and problems with sleep. TS occurs in most populations and ethnic groups worldwide, and it is more common in males than in females. Previous family and twin studies have shown that the majority of cases of TS are inherited. TS was previously thought to have an autosomal dominant pattern of inheritance. However, several decades of research have shown that this is unlikely the case. Instead TS most likely results from a variety of genetic and environmental factors, not changes in a single gene. In the past decade, there has been a rapid development of innovative genetic technologies and methodologies, as well as significant progresses in genetic studies of psychiatric disorders. In this review, we will briefly summarize previous genetic epidemiological studies of TS and related disorders. We will also review previous genetic studies based on genome-wide linkage analyses and candidate gene association studies to comment on problems of previous methodological and strategic issues. Our main purpose for this review will be to summarize the new genetic discoveries of TS based on novel genetic methods and strategies, such as genome-wide association studies (GWASs), whole exome sequencing (WES) and whole genome sequencing (WGS). We will also compare the new genetic discoveries of TS with other major psychiatric disorders in order to understand the current status of TS genetics and its relationship with other psychiatric disorders.
Collapse
Affiliation(s)
- Yanjie Qi
- Laboratoire de Neurogénétique, Centre de Recherche, Institut Universitaire en Santé Mentale de Montréal, Montreal, QC H1N 3V2, Canada.
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China.
| | - Yi Zheng
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China.
- Center of Schizophrenia, Beijing Institute for Brain Disorders, Beijing 100088, China.
| | - Zhanjiang Li
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China.
- Center of Schizophrenia, Beijing Institute for Brain Disorders, Beijing 100088, China.
| | - Lan Xiong
- Laboratoire de Neurogénétique, Centre de Recherche, Institut Universitaire en Santé Mentale de Montréal, Montreal, QC H1N 3V2, Canada.
- Département de Psychiatrie, Faculté de Médecine, Université de Montréal, Montreal, QC H3C 3J7, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
39
|
Sirchia F, Di Gregorio E, Restagno G, Grosso E, Pappi P, Talarico F, Savin E, Cavalieri S, Giorgio E, Mancini C, Pasini B, Mehta JS, Brusco A. A case of Feingold type 2 syndrome associated with keratoconus refines keratoconus type 7 locus on chromosome 13q. Eur J Med Genet 2017; 60:224-227. [DOI: 10.1016/j.ejmg.2017.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/21/2016] [Accepted: 01/29/2017] [Indexed: 01/21/2023]
|
40
|
Chen Y, Bartanus J, Liang D, Zhu H, Breman AM, Smith JL, Wang H, Ren Z, Patel A, Stankiewicz P, Cram DS, Cheung SW, Wu L, Yu F. Characterization of chromosomal abnormalities in pregnancy losses reveals critical genes and loci for human early development. Hum Mutat 2017; 38:669-677. [PMID: 28247551 DOI: 10.1002/humu.23207] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/19/2017] [Accepted: 02/21/2017] [Indexed: 11/09/2022]
Abstract
Detailed characterization of chromosomal abnormalities, a common cause for congenital abnormalities and pregnancy loss, is critical for elucidating genes for human fetal development. Here, 2,186 product-of-conception samples were tested for copy-number variations (CNVs) at two clinical diagnostic centers using whole-genome sequencing and high-resolution chromosomal microarray analysis. We developed a new gene discovery approach to predict potential developmental genes and identified 275 candidate genes from CNVs detected from both datasets. Based on Mouse Genome Informatics (MGI) and Zebrafish model organism database (ZFIN), 75% of identified genes could lead to developmental defects when mutated. Genes involved in embryonic development, gene transcription, and regulation of biological processes were significantly enriched. Especially, transcription factors and gene families sharing specific protein domains predominated, which included known developmental genes such as HOX, NKX homeodomain genes, and helix-loop-helix containing HAND2, NEUROG2, and NEUROD1 as well as potential novel developmental genes. We observed that developmental genes were denser in certain chromosomal regions, enabling identification of 31 potential genomic loci with clustered genes associated with development.
Collapse
Affiliation(s)
- Yiyun Chen
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Justin Bartanus
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Desheng Liang
- State Key Lab of Medical Genetics of China Central South University, Changsha, Hunan, China
| | | | - Amy M Breman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Cytogenetics Laboratory, Baylor Miraca Genetics Laboratories, Houston, Texas
| | - Janice L Smith
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Cytogenetics Laboratory, Baylor Miraca Genetics Laboratories, Houston, Texas
| | - Hua Wang
- Hunan Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Zhilin Ren
- Berry Genomics Corporation, Beijing, China
| | - Ankita Patel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Cytogenetics Laboratory, Baylor Miraca Genetics Laboratories, Houston, Texas
| | - Pawel Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Cytogenetics Laboratory, Baylor Miraca Genetics Laboratories, Houston, Texas
| | | | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Cytogenetics Laboratory, Baylor Miraca Genetics Laboratories, Houston, Texas
| | - Lingqian Wu
- State Key Lab of Medical Genetics of China Central South University, Changsha, Hunan, China
| | - Fuli Yu
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Berry Genomics Corporation, Beijing, China
| |
Collapse
|
41
|
Song M, Mathews CA, Stewart SE, Shmelkov SV, Mezey JG, Rodriguez-Flores JL, Rasmussen SA, Britton JC, Oh YS, Walkup JT, Lee FS, Glatt CE. Rare Synaptogenesis-Impairing Mutations in SLITRK5 Are Associated with Obsessive Compulsive Disorder. PLoS One 2017; 12:e0169994. [PMID: 28085938 PMCID: PMC5234816 DOI: 10.1371/journal.pone.0169994] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/27/2016] [Indexed: 01/02/2023] Open
Abstract
Obsessive compulsive disorder (OCD) is substantially heritable, but few molecular genetic risk factors have been identified. Knockout mice lacking SLIT and NTRK-Like Family, Member 5 (SLITRK5) display OCD-like phenotypes including serotonin reuptake inhibitor-sensitive pathologic grooming, and corticostriatal dysfunction. Thus, mutations that impair SLITRK5 function may contribute to the genetic risk for OCD. We re-sequenced the protein-coding sequence of the human SLITRK5 gene (SLITRK5) in three hundred and seventy seven OCD subjects and compared rare non-synonymous mutations (RNMs) in that sample with similar mutations in the 1000 Genomes database. We also performed in silico assessments and in vitro functional synaptogenesis assays on the Slitrk5 mutations identified. We identified four RNM's among these OCD subjects. There were no significant differences in the prevalence or in silico effects of rare non-synonymous mutations in the OCD sample versus controls. Direct functional testing of recombinant SLITRK5 proteins found that all mutations identified in OCD subjects impaired synaptogenic activity whereas none of the pseudo-matched mutations identified in 1000 Genomes controls had significant effects on SLITRK5 function (Fisher's exact test P = 0.028). These results demonstrate that rare functional mutations in SLITRK5 contribute to the genetic risk for OCD in human populations. They also highlight the importance of biological characterization of allelic effects in understanding genotype-phenotype relationships as there were no statistical differences in overall prevalence or bioinformatically predicted effects of OCD case versus control mutations. Finally, these results converge with others to highlight the role of aberrant synaptic function in corticostriatal neurons in the pathophysiology of OCD.
Collapse
Affiliation(s)
- Minseok Song
- Synaptic Circuit Plasticity Laboratory, Department of Structure & Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, Korea
| | - Carol A. Mathews
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
| | - S. Evelyn Stewart
- Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Sergey V. Shmelkov
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, United States of America
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Jason G. Mezey
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY United States of America
- Department of Genetic Medicine, Weill Cornell Medical College, NY, NY United States of America
| | | | - Steven A. Rasmussen
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, United States of America
| | - Jennifer C. Britton
- Department of Psychology, University of Miami, Miami, FL, United States of America
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - John T. Walkup
- Department of Psychiatry, Weill Cornell Medical College, New York, New York, United States of America
| | - Francis S. Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, New York, United States of America
- Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, New York, United States of America
| | - Charles E. Glatt
- Department of Psychiatry, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
42
|
Hashemiyoon R, Kuhn J, Visser-Vandewalle V. Putting the Pieces Together in Gilles de la Tourette Syndrome: Exploring the Link Between Clinical Observations and the Biological Basis of Dysfunction. Brain Topogr 2017; 30:3-29. [PMID: 27783238 PMCID: PMC5219042 DOI: 10.1007/s10548-016-0525-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022]
Abstract
Gilles de la Tourette syndrome is a complex, idiopathic neuropsychiatric disorder whose pathophysiological mechanisms have yet to be elucidated. It is phenotypically heterogeneous and manifests more often than not with both motor and behavioral impairment, although tics are its clinical hallmark. Tics themselves present with a complex profile as they characteristically wax and wane and are often preceded by premonitory somatosensory sensations to which it is said a tic is the response. Highly comorbid with obsessive-compulsive disorder and attention deficit-hyperactivity disorder, it is purported to be an epigenetic, neurodevelopmental spectrum disorder with a complex genetic profile. It has a childhood onset, occurs disproportionately in males, and shows spontaneous symptomatic attenuation by adulthood in the majority of those afflicted. Although not fully understood, its neurobiological basis is linked to dysfunction in the cortico-basal ganglia-thalamo-cortical network. Treatment modalities for Tourette syndrome include behavioral, pharmacological and surgical interventions, but there is presently no cure for the disorder. For those severely affected, deep brain stimulation (DBS) has recently become a viable therapeutic option. A key factor to attaining optimal results from this surgery is target selection, a topic still under debate due to the complex clinical profile presented by GTS patients. Depending on its phenotypic expression and the most problematic aspect of the disorder for the individual, one of three brain regions is most commonly chosen for stimulation: the thalamus, globus pallidus, or nucleus accumbens. Neurophysiological analyses of intra- and post-operative human electrophysiological recordings from clinical DBS studies suggest a link between tic behavior and activity in both the thalamus and globus pallidus. In particular, chronic recordings from the thalamus have shown a correlation between symptomatology and (1) spectral activity in gamma band power and (2) theta/gamma cross frequency coherence. These results suggest gamma oscillations and theta/gamma cross correlation dynamics may serve as biomarkers for dysfunction. While acute and chronic recordings from human subjects undergoing DBS have provided better insight into tic genesis and the neuropathophysiological mechanisms underlying Tourette syndrome, these studies are still sparse and the field would greatly benefit from further investigations. This review reports data and discoveries of scientific and clinical relevance from a wide variety of methods and provides up-to-date information about our current understanding of the pathomechanisms underlying Tourette syndrome. It gives a comprehensive overview of the current state of knowledge and addresses open questions in the field.
Collapse
Affiliation(s)
- Rowshanak Hashemiyoon
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany.
| | - Jens Kuhn
- Department of Psychiatry and Psychotherapy, University Hospital of Cologne, Cologne, Germany
- Johanniter Hospital, EVKLN, Oberhausen, Germany
| | - Veerle Visser-Vandewalle
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| |
Collapse
|
43
|
Disruption of Ninjurin1 Leads to Repetitive and Anxiety-Like Behaviors in Mice. Mol Neurobiol 2016; 54:7353-7368. [PMID: 27815839 DOI: 10.1007/s12035-016-0207-6] [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: 06/08/2016] [Accepted: 10/11/2016] [Indexed: 01/28/2023]
Abstract
Over the last few decades, molecular neurobiology has uncovered many genes whose deficiency in mice results in behavioral traits associated with human neuropsychiatric disorders such as autism, obsessive-compulsive disorder (OCD), and schizophrenia. However, the etiology of these common diseases remains enigmatic with the potential involvement of a battery of genes. Here, we report abnormal behavioral phenotypes of mice deficient in a cell adhesion molecule Ninjurin 1 (Ninj1), which are relevant to repetitive and anxiety behaviors of neuropsychiatric disorders. Ninj1 knockout (KO) mice exhibit compulsive grooming-induced hair loss and self-made lesions as well as increased anxiety-like behaviors. Histological analysis reveals that Ninj1 is predominantly expressed in cortico-thalamic circuits, and neuron-specific Ninj1 conditional KO mice manifest aberrant phenotypes similar to the global Ninj1 KO mice. Notably, the brains of Ninj1 KO mice display altered synaptic transmission in thalamic neurons as well as a reduced number of functional synapses. Moreover, the disruption of Ninj1 leads to glutamatergic abnormalities, including increased ionotropic glutamate receptors but reduced glutamate levels. Furthermore, chronic treatment with fluoxetine, a drug reportedly ameliorates compulsive behaviors in mice, prevents progression of hair loss and alleviates the compulsive grooming and anxiety-like behavior of Ninj1 KO mice. Collectively, our results suggest that Ninj1 could be involved in neuropsychiatric disorders associated with impairments of repetitive and anxiety behaviors.
Collapse
|
44
|
Kang H, Han KA, Won SY, Kim HM, Lee YH, Ko J, Um JW. Slitrk Missense Mutations Associated with Neuropsychiatric Disorders Distinctively Impair Slitrk Trafficking and Synapse Formation. Front Mol Neurosci 2016; 9:104. [PMID: 27812321 PMCID: PMC5071332 DOI: 10.3389/fnmol.2016.00104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/04/2016] [Indexed: 12/29/2022] Open
Abstract
Slit- and Trk-like (Slitrks) are a six-member family of synapse organizers that control excitatory and inhibitory synapse formation by forming trans-synaptic adhesions with LAR receptor protein tyrosine phosphatases (PTPs). Intriguingly, genetic mutations of Slitrks have been associated with a multitude of neuropsychiatric disorders. However, nothing is known about the neuronal and synaptic consequences of these mutations. Here, we report the structural and functional effects on synapses of various rare de novo mutations identified in patients with schizophrenia or Tourette syndrome. A number of single amino acid substitutions in Slitrk1 (N400I or T418S) or Slitrk4 (V206I or I578V) reduced their surface expression levels. These substitutions impaired glycosylation of Slitrks expressed in HEK293T cells, caused retention of Slitrks in the endoplasmic reticulum and cis-Golgi compartment in COS-7 cells and neurons, and abolished Slitrk binding to PTPδ. Furthermore, these substitutions eliminated the synapse-inducing activity of Slitrks, abolishing their functional effects on synapse density in cultured neurons. Strikingly, a valine-to-methionine mutation in Slitrk2 (V89M) compromised synapse formation activity in cultured neuron, without affecting surface transport, expression, or synapse-inducing activity in coculture assays. Similar deleterious effects were observed upon introduction of the corresponding valine-to-methionine mutation into Slitrk1 (V85M), suggesting that this conserved valine residue plays a key role in maintaining the synaptic functions of Slitrks. Collectively, these data indicate that inactivation of distinct cellular mechanisms caused by specific Slitrk dysfunctions may underlie Slitrk-associated neuropsychiatric disorders in humans, and provide a robust cellular readout for the development of knowledge-based therapies.
Collapse
Affiliation(s)
- Hyeyeon Kang
- Department of Physiology and BK21 PLUS Project for Medical Science, Yonsei University College of Medicine Seoul, Korea
| | - Kyung Ah Han
- Department of Physiology and BK21 PLUS Project for Medical Science, Yonsei University College of Medicine Seoul, Korea
| | - Seoung Youn Won
- Department of Chemistry, Korea Advanced Institute of Science and Technology Daejeon, Korea
| | - Ho Min Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology Daejeon, Korea
| | - Young-Ho Lee
- Department of Physiology and BK21 PLUS Project for Medical Science, Yonsei University College of Medicine Seoul, Korea
| | - Jaewon Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University Seoul, Korea
| | - Ji Won Um
- Department of Physiology and BK21 PLUS Project for Medical Science, Yonsei University College of Medicine Seoul, Korea
| |
Collapse
|
45
|
Alexander J, Potamianou H, Xing J, Deng L, Karagiannidis I, Tsetsos F, Drineas P, Tarnok Z, Rizzo R, Wolanczyk T, Farkas L, Nagy P, Szymanska U, Androutsos C, Tsironi V, Koumoula A, Barta C, Sandor P, Barr CL, Tischfield J, Paschou P, Heiman GA, Georgitsi M. Targeted Re-Sequencing Approach of Candidate Genes Implicates Rare Potentially Functional Variants in Tourette Syndrome Etiology. Front Neurosci 2016; 10:428. [PMID: 27708560 PMCID: PMC5030307 DOI: 10.3389/fnins.2016.00428] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
Although the genetic basis of Tourette Syndrome (TS) remains unclear, several candidate genes have been implicated. Using a set of 382 TS individuals of European ancestry we investigated four candidate genes for TS (HDC, SLITRK1, BTBD9, and SLC6A4) in an effort to identify possibly causal variants using a targeted re-sequencing approach by next generation sequencing technology. Identification of possible disease causing variants under different modes of inheritance was performed using the algorithms implemented in VAAST. We prioritized variants using Variant ranker and validated five rare variants via Sanger sequencing in HDC and SLITRK1, all of which are predicted to be deleterious. Intriguingly, one of the identified variants is in linkage disequilibrium with a variant that is included among the top hits of a genome-wide association study for response to citalopram treatment, an antidepressant drug with off-label use also in obsessive compulsive disorder. Our findings provide additional evidence for the implication of these two genes in TS susceptibility and the possible role of these proteins in the pathobiology of TS should be revisited.
Collapse
Affiliation(s)
- John Alexander
- Department of Molecular Biology and Genetics, Democritus University of Thrace Alexandroupoli, Greece
| | - Hera Potamianou
- Department of Molecular Biology and Genetics, Democritus University of Thrace Alexandroupoli, Greece
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New JerseyPiscataway, NJ, USA; Human Genetics Institute of New Jersey, Rutgers, The State University of New JerseyPiscataway, NJ, USA
| | - Li Deng
- Department of Genetics, Rutgers, The State University of New JerseyPiscataway, NJ, USA; Human Genetics Institute of New Jersey, Rutgers, The State University of New JerseyPiscataway, NJ, USA
| | - Iordanis Karagiannidis
- Department of Molecular Biology and Genetics, Democritus University of Thrace Alexandroupoli, Greece
| | - Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace Alexandroupoli, Greece
| | - Petros Drineas
- Computer Science Department, Purdue University West Lafayette, USA
| | - Zsanett Tarnok
- Vadaskert Clinic for Child and Adolescent Psychiatry Budapest, Hungary
| | - Renata Rizzo
- Department of Clinical and Experimental Medicine, University of Catania Catania, Italy
| | - Tomasz Wolanczyk
- Department of Child Psychiatry, Medical University of Warsaw Warsaw, Poland
| | - Luca Farkas
- Vadaskert Clinic for Child and Adolescent Psychiatry Budapest, Hungary
| | - Peter Nagy
- Vadaskert Clinic for Child and Adolescent Psychiatry Budapest, Hungary
| | - Urszula Szymanska
- Department of Child Psychiatry, Medical University of Warsaw Warsaw, Poland
| | - Christos Androutsos
- Child and Adolescent Psychiatry Clinic, Sismanoglio General Hospital of Attica Athens, Greece
| | - Vaia Tsironi
- Child and Adolescent Psychiatry Clinic, Sismanoglio General Hospital of Attica Athens, Greece
| | - Anastasia Koumoula
- Child and Adolescent Psychiatry Clinic, Sismanoglio General Hospital of Attica Athens, Greece
| | - Csaba Barta
- Molecular Biology and Pathobiochemistry, Institute of Medical Chemistry, Semmelweis University Budapest, Hungary
| | | | - Paul Sandor
- Department of Psychiatry, University of Toronto Toronto, ON, Canada
| | - Cathy L Barr
- Genetics and Development Division, Krembil Research Institute, University Health NetworkToronto, ON, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick ChildrenToronto, ON, Canada
| | - Jay Tischfield
- Department of Genetics, Rutgers, The State University of New JerseyPiscataway, NJ, USA; Human Genetics Institute of New Jersey, Rutgers, The State University of New JerseyPiscataway, NJ, USA
| | - Peristera Paschou
- Department of Molecular Biology and Genetics, Democritus University of Thrace Alexandroupoli, Greece
| | - Gary A Heiman
- Department of Genetics, Rutgers, The State University of New JerseyPiscataway, NJ, USA; Human Genetics Institute of New Jersey, Rutgers, The State University of New JerseyPiscataway, NJ, USA
| | - Marianthi Georgitsi
- Department of Molecular Biology and Genetics, Democritus University of ThraceAlexandroupoli, Greece; Laboratory of General Biology, Department of Medicine, Aristotle University of ThessalonikiThessaloniki, Greece
| |
Collapse
|
46
|
Rodent models of obsessive compulsive disorder: Evaluating validity to interpret emerging neurobiology. Neuroscience 2016; 345:256-273. [PMID: 27646291 DOI: 10.1016/j.neuroscience.2016.09.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/03/2016] [Accepted: 09/08/2016] [Indexed: 11/21/2022]
Abstract
Obsessive Compulsive Disorder (OCD) is a common neuropsychiatric disorder with unknown molecular underpinnings. Identification of genetic and non-genetic risk factors has largely been elusive, primarily because of a lack of power. In contrast, neuroimaging has consistently implicated the cortico-striatal-thalamo-cortical circuits in OCD. Pharmacological treatment studies also show specificity, with consistent response of OCD symptoms to chronic treatment with serotonin reuptake inhibitors; although most patients are left with residual impairment. In theory, animal models could provide a bridge from the neuroimaging and pharmacology data to an understanding of pathophysiology at the cellular and molecular level. Several mouse models have been proposed using genetic, immunological, pharmacological, and optogenetic tools. These experimental model systems allow testing of hypotheses about the origins of compulsive behavior. Several models have generated behavior that appears compulsive-like, particularly excessive grooming, and some have demonstrated response to chronic serotonin reuptake inhibitors, establishing both face validity and predictive validity. Construct validity is more difficult to establish in the context of a limited understanding of OCD risk factors. Our current models may help us to dissect the circuits and molecular pathways that can elicit OCD-relevant behavior in rodents. We can hope that this growing understanding, coupled with developing technology, will prepare us when robust OCD risk factors are better understood.
Collapse
|
47
|
Doucet-Beaupré H, Gilbert C, Profes MS, Chabrat A, Pacelli C, Giguère N, Rioux V, Charest J, Deng Q, Laguna A, Ericson J, Perlmann T, Ang SL, Cicchetti F, Parent M, Trudeau LE, Lévesque M. Lmx1a and Lmx1b regulate mitochondrial functions and survival of adult midbrain dopaminergic neurons. Proc Natl Acad Sci U S A 2016; 113:E4387-96. [PMID: 27407143 PMCID: PMC4968767 DOI: 10.1073/pnas.1520387113] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The LIM-homeodomain transcription factors Lmx1a and Lmx1b play critical roles during the development of midbrain dopaminergic progenitors, but their functions in the adult brain remain poorly understood. We show here that sustained expression of Lmx1a and Lmx1b is required for the survival of adult midbrain dopaminergic neurons. Strikingly, inactivation of Lmx1a and Lmx1b recreates cellular features observed in Parkinson's disease. We found that Lmx1a/b control the expression of key genes involved in mitochondrial functions, and their ablation results in impaired respiratory chain activity, increased oxidative stress, and mitochondrial DNA damage. Lmx1a/b deficiency caused axonal pathology characterized by α-synuclein(+) inclusions, followed by a progressive loss of dopaminergic neurons. These results reveal the key role of these transcription factors beyond the early developmental stages and provide mechanistic links between mitochondrial dysfunctions, α-synuclein aggregation, and the survival of dopaminergic neurons.
Collapse
Affiliation(s)
- Hélène Doucet-Beaupré
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Catherine Gilbert
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Marcos Schaan Profes
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Audrey Chabrat
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Consiglia Pacelli
- Department of Pharmacology, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Department of Neurosciences, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Nicolas Giguère
- Department of Pharmacology, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Department of Neurosciences, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Véronique Rioux
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Julien Charest
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Qiaolin Deng
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ariadna Laguna
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; The Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; The Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden
| | - Siew-Lan Ang
- The Francis Crick Institute, London, NW1 2BE, United Kingdom
| | - Francesca Cicchetti
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de recherche du Centre Hospitalier Universitaire de Québec, Quebec, QC G1V 4G2, Canada
| | - Martin Parent
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada
| | - Louis-Eric Trudeau
- Department of Pharmacology, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Department of Neurosciences, Central Nervous System Research Group, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec QC G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec, QC G1J 2G3, Canada;
| |
Collapse
|
48
|
Pagliaroli L, Vető B, Arányi T, Barta C. From Genetics to Epigenetics: New Perspectives in Tourette Syndrome Research. Front Neurosci 2016; 10:277. [PMID: 27462201 PMCID: PMC4940402 DOI: 10.3389/fnins.2016.00277] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/06/2016] [Indexed: 11/13/2022] Open
Abstract
Gilles de la Tourette Syndrome (TS) is a neurodevelopmental disorder marked by the appearance of multiple involuntary motor and vocal tics. TS presents high comorbidity rates with other disorders such as attention deficit hyperactivity disorder (ADHD) and obsessive compulsive disorder (OCD). TS is highly heritable and has a complex polygenic background. However, environmental factors also play a role in the manifestation of symptoms. Different epigenetic mechanisms may represent the link between these two causalities. Epigenetic regulation has been shown to have an impact in the development of many neuropsychiatric disorders, however very little is known about its effects on Tourette Syndrome. This review provides a summary of the recent findings in genetic background of TS, followed by an overview on different epigenetic mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs in the regulation of gene expression. Epigenetic studies in other neurological and psychiatric disorders are discussed along with the TS-related epigenetic findings available in the literature to date. Moreover, we are proposing that some general epigenetic mechanisms seen in other neuropsychiatric disorders may also play a role in the pathogenesis of TS.
Collapse
Affiliation(s)
- Luca Pagliaroli
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis UniversityBudapest, Hungary; Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of SciencesBudapest, Hungary
| | - Borbála Vető
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences Budapest, Hungary
| | - Tamás Arányi
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of SciencesBudapest, Hungary; Centre National de la Recherche Scientifique UMR 6214, Institut National de la Santé et de la Recherche Médicale U1083, University of AngersAngers, France
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Hungary
| |
Collapse
|
49
|
Spinello C, Laviola G, Macrì S. Pediatric Autoimmune Disorders Associated with Streptococcal Infections and Tourette's Syndrome in Preclinical Studies. Front Neurosci 2016; 10:310. [PMID: 27445678 PMCID: PMC4928151 DOI: 10.3389/fnins.2016.00310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/20/2016] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence suggests that Tourette's Syndrome (TS) - a multifactorial pediatric disorder characterized by the recurrent exhibition of motor tics and/or vocal utterances - can partly depend on immune dysregulation provoked by early repeated streptococcal infections. The natural and adaptive antibody-mediated reaction to streptococcus has been proposed to potentially turn into a pathological autoimmune response in vulnerable individuals. Specifically, in conditions of increased permeability of the blood brain barrier (BBB), streptococcus-induced antibodies have been proposed to: (i) reach neuronal targets located in brain areas responsible for motion control; and (ii) contribute to the exhibition of symptoms. This theoretical framework is supported by indirect evidence indicating that a subset of TS patients exhibit elevated streptococcal antibody titers upon tic relapses. A systematic evaluation of this hypothesis entails preclinical studies providing a proof of concept of the aforementioned pathological sequelae. These studies shall rest upon individuals characterized by a vulnerable immune system, repeatedly exposed to streptococcus, and carefully screened for phenotypes isomorphic to the pathological signs of TS observed in patients. Preclinical animal models may thus constitute an informative, useful tool upon which conducting targeted, hypothesis-driven experiments. In the present review we discuss the available evidence in preclinical models in support of the link between TS and pediatric autoimmune neuropsychiatric disorders associated with streptococcus infections (PANDAS), and the existing gaps that future research shall bridge. Specifically, we report recent preclinical evidence indicating that the immune responses to repeated streptococcal immunizations relate to the occurrence of behavioral and neurological phenotypes reminiscent of TS. By the same token, we discuss the limitations of these studies: limited evidence of behavioral phenotypes isomorphic to tics and scarce knowledge about the immunological phenomena favoring the transition from natural adaptive immunity to pathological outcomes.
Collapse
Affiliation(s)
- Chiara Spinello
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Giovanni Laviola
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Simone Macrì
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| |
Collapse
|
50
|
Alsina FC, Hita FJ, Fontanet PA, Irala D, Hedman H, Ledda F, Paratcha G. Lrig1 is a cell-intrinsic modulator of hippocampal dendrite complexity and BDNF signaling. EMBO Rep 2016; 17:601-16. [PMID: 26935556 DOI: 10.15252/embr.201541218] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 11/09/2022] Open
Abstract
Even though many extracellular factors have been identified as promoters of general dendritic growth and branching, little is known about the cell-intrinsic modulators that allow neurons to sculpt distinctive patterns of dendrite arborization. Here, we identify Lrig1, a nervous system-enriched LRR protein, as a key physiological regulator of dendrite complexity of hippocampal pyramidal neurons. Lrig1-deficient mice display morphological changes in proximal dendrite arborization and defects in social interaction. Specifically, knockdown of Lrig1 enhances both primary dendrite formation and proximal dendritic branching of hippocampal neurons, two phenotypes that resemble the effect of BDNF on these neurons. In addition, we show that Lrig1 physically interacts with TrkB and attenuates BDNF signaling. Gain and loss of function assays indicate that Lrig1 restricts BDNF-induced dendrite morphology. Together, our findings reveal a novel and essential role of Lrig1 in regulating morphogenic events that shape the hippocampal circuits and establish that the assembly of TrkB with Lrig1 represents a key mechanism for understanding how specific neuronal populations expand the repertoire of responses to BDNF during brain development.
Collapse
Affiliation(s)
- Fernando Cruz Alsina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Francisco Javier Hita
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Dolores Irala
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Håkan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|