1
|
Liu Z, Pan C, Huang H. The role of axon guidance molecules in the pathogenesis of epilepsy. Neural Regen Res 2025; 20:1244-1257. [PMID: 39075893 PMCID: PMC11624883 DOI: 10.4103/nrr.nrr-d-23-01620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 07/31/2024] Open
Abstract
Current treatments for epilepsy can only manage the symptoms of the condition but cannot alter the initial onset or halt the progression of the disease. Consequently, it is crucial to identify drugs that can target novel cellular and molecular mechanisms and mechanisms of action. Increasing evidence suggests that axon guidance molecules play a role in the structural and functional modifications of neural networks and that the dysregulation of these molecules is associated with epilepsy susceptibility. In this review, we discuss the essential role of axon guidance molecules in neuronal activity in patients with epilepsy as well as the impact of these molecules on synaptic plasticity and brain tissue remodeling. Furthermore, we examine the relationship between axon guidance molecules and neuroinflammation, as well as the structural changes in specific brain regions that contribute to the development of epilepsy. Ample evidence indicates that axon guidance molecules, including semaphorins and ephrins, play a fundamental role in guiding axon growth and the establishment of synaptic connections. Deviations in their expression or function can disrupt neuronal connections, ultimately leading to epileptic seizures. The remodeling of neural networks is a significant characteristic of epilepsy, with axon guidance molecules playing a role in the dynamic reorganization of neural circuits. This, in turn, affects synapse formation and elimination. Dysregulation of these molecules can upset the delicate balance between excitation and inhibition within a neural network, thereby increasing the risk of overexcitation and the development of epilepsy. Inflammatory signals can regulate the expression and function of axon guidance molecules, thus influencing axonal growth, axon orientation, and synaptic plasticity. The dysregulation of neuroinflammation can intensify neuronal dysfunction and contribute to the occurrence of epilepsy. This review delves into the mechanisms associated with the pathogenicity of axon guidance molecules in epilepsy, offering a valuable reference for the exploration of therapeutic targets and presenting a fresh perspective on treatment strategies for this condition.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Chunhua Pan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
Zhu K, Wang H, Ye K, Chen G, Zhang Z. Netrin-1 signaling pathway mechanisms in neurodegenerative diseases. Neural Regen Res 2025; 20:960-972. [PMID: 38989931 PMCID: PMC11438344 DOI: 10.4103/nrr.nrr-d-23-01573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/16/2024] [Indexed: 07/12/2024] Open
Abstract
Netrin-1 and its receptors play crucial roles in inducing axonal growth and neuronal migration during neuronal development. Their profound impacts then extend into adulthood to encompass the maintenance of neuronal survival and synaptic function. Increasing amounts of evidence highlight several key points: (1) Diminished Netrin-1 levels exacerbate pathological progression in animal models of Alzheimer's disease and Parkinson's disease, and potentially, similar alterations occur in humans. (2) Genetic mutations of Netrin-1 receptors increase an individuals' susceptibility to neurodegenerative disorders. (3) Therapeutic approaches targeting Netrin-1 and its receptors offer the benefits of enhancing memory and motor function. (4) Netrin-1 and its receptors show genetic and epigenetic alterations in a variety of cancers. These findings provide compelling evidence that Netrin-1 and its receptors are crucial targets in neurodegenerative diseases. Through a comprehensive review of Netrin-1 signaling pathways, our objective is to uncover potential therapeutic avenues for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kedong Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Hualong Wang
- Department of Neurology, The First Hospital of Hebei Medical University; Brain Aging and Cognitive Neuroscience Laboratory of Heibei Province, Shijiazhuang, Hebei Province, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Guiqin Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Lee S, McAfee JC, Lee J, Gomez A, Ledford AT, Clarke D, Min H, Gerstein MB, Boyle AP, Sullivan PF, Beltran AS, Won H. Massively parallel reporter assay investigates shared genetic variants of eight psychiatric disorders. Cell 2025:S0092-8674(24)01435-1. [PMID: 39848247 DOI: 10.1016/j.cell.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 07/08/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025]
Abstract
A meta-genome-wide association study across eight psychiatric disorders has highlighted the genetic architecture of pleiotropy in major psychiatric disorders. However, mechanisms underlying pleiotropic effects of the associated variants remain to be explored. We conducted a massively parallel reporter assay to decode the regulatory logic of variants with pleiotropic and disorder-specific effects. Pleiotropic variants differ from disorder-specific variants by exhibiting chromatin accessibility that extends across diverse cell types in the neuronal lineage and by altering motifs for transcription factors with higher connectivity in protein-protein interaction networks. We mapped pleiotropic and disorder-specific variants to putative target genes using functional genomics approaches and CRISPR perturbation. In vivo CRISPR perturbation of a pleiotropic and a disorder-specific gene suggests that pleiotropy may involve the regulation of genes expressed broadly across neuronal cell types and with higher network connectivity.
Collapse
Affiliation(s)
- Sool Lee
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica C McAfee
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiseok Lee
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro Gomez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Austin T Ledford
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Declan Clarke
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520, USA; Department of Computer Science, Yale University, New Haven, CT 06520, USA; Department of Statistics & Data Science, Yale University, New Haven, CT 06520, USA; Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT 06520, USA
| | - Hyunggyu Min
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark B Gerstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520, USA; Department of Computer Science, Yale University, New Haven, CT 06520, USA; Department of Statistics & Data Science, Yale University, New Haven, CT 06520, USA; Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT 06520, USA
| | - Alan P Boyle
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Adriana S Beltran
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Human Pluripotent Cell Core, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Göteson A, Holmén-Larsson J, Celik H, Pelanis A, Sellgren CM, Sparding T, Pålsson E, Zetterberg H, Blennow K, Jonsson L, Gobom J, Landén M. MAPPING THE CEREBROSPINAL FLUID PROTEOME IN BIPOLAR DISORDER. Biol Psychiatry 2025:S0006-3223(25)00029-0. [PMID: 39827936 DOI: 10.1016/j.biopsych.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Bipolar disorder (BD) is a severe psychiatric condition with unclear etiology and no established biomarkers. Here, we aimed to characterize the cerebrospinal fluid (CSF) proteome in euthymic BD individuals to identify potential protein biomarkers. METHODS We employed nano-flow liquid chromatography coupled to high-resolution mass spectrometry to quantify over 2,000 CSF proteins in 374 individuals from two independent clinical cohorts (n=164+89 and 66+55 cases and controls, respectively). A subset of the cases was followed longitudinally and reexamined after a median of 6.5 years. RESULTS Differential abundance analysis revealed 41 proteins with robust case-control association in both cohorts. These included lower levels of synaptic proteins (e.g., APP, CLSTN1, NPTX2, NRXN1), axon guidance and cell adhesion molecules (e.g., NEO1, NCAM1, SEMA7A), higher levels of blood-brain-barrier integrity proteins (e.g., VTN, SERPIN3), and complement components (e.g., C1RL, C3, C5). The findings were consistently driven by the BD type 1 subtype. Comparing BD type 1 with controls increased discoverability, revealing 86 replicated associations despite a loss of statistical power. Moreover, longitudinal analyses of co-expression modules revealed dynamic changes in the CSF proteome composition that correlated with clinical outcomes, including disease severity, future manic episodes, and symptom improvement. Finally, we conducted association analyses of CSF proteins with genetic risk loci for bipolar disorder and schizophrenia. CONCLUSIONS This study represents the first large-scale untargeted profiling of the CSF proteome in BD, unveiling potential biomarkers and providing in vivo support for altered synaptic and brain connectivity processes, impaired neurovascular integrity, and complement activation in the pathology of BD.
Collapse
Affiliation(s)
- Andreas Göteson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Jessica Holmén-Larsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Hatice Celik
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Aurimantas Pelanis
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Timea Sparding
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Pålsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Lina Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mikael Landén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Khosroshahi PA, Ghanbari M. MicroRNA dysregulation in glutamate and dopamine pathways of schizophrenia: From molecular pathways to diagnostic and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111081. [PMID: 39002925 DOI: 10.1016/j.pnpbp.2024.111081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
Schizophrenia is a complex psychiatric disorder, and genetic and environmental factors have been implicated in its development. Dysregulated glutamatergic and dopaminergic transmission pathways are involved in schizophrenia development. Besides genetic mutations, epigenetic dysregulation has a considerable role in dysregulating molecular pathways involved in schizophrenia. MicroRNAs (miRNAs) are small, non-coding RNAs that target specific mRNAs and inhibit their translation into proteins. As epigenetic factors, miRNAs regulate many genes involved in glutamate and dopamine signaling pathways; thereby, their dysregulation can contribute to the development of schizophrenia. Secretion of specific miRNAs from damaged cells into body fluids can make them one of the ideal non-invasive biomarkers in the early diagnosis of schizophrenia. Also, understanding the molecular mechanisms of miRNAs in schizophrenia pathogenesis can pave the way for developing novel treatments for patients with schizophrenia. In this study, we reviewed the glutamatergic and dopaminergic pathophysiology and highlighted the role of miRNA dysregulation in schizophrenia development. Besides, we shed light on the significance of circulating miRNAs for schizophrenia diagnosis and the recent findings on the miRNA-based treatment for schizophrenia.
Collapse
Affiliation(s)
| | - Mohammad Ghanbari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
6
|
Wu YL, Hu T, Zheng H, Feng J, Huang C, Zhou X, Wang W, Jiang CL. Pinocembrin alleviates LPS-induced depressive-like behavior in mice via the NLRP3/DCC signaling pathway. Biochem Biophys Res Commun 2024; 736:150870. [PMID: 39476759 DOI: 10.1016/j.bbrc.2024.150870] [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: 08/21/2024] [Revised: 10/01/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
OBJECTIVE Depression, a prevalent and severe mental disorder, continues to be a significant area of research concerning its pathogenesis and therapeutic approaches. Conventional antidepressants are often limited by delayed therapeutic effects and notable adverse reactions, necessitating the development of innovative and efficacious treatment modalities. Multiple lines of evidence suggest that peripheral and central inflammation play a role in depression, and that anti-inflammatory drugs can ameliorate depressive symptoms in patients with inflammation-related depression. Pinocembrin (PB), a natural bioactive compound, is renowned for its anti-inflammatory and antioxidant properties, while the effect and mechanism of PB are still unclear. Consequently, this study employs PB as an intervention to investigate its effects on depression in mice model, with the objective of establishing a novel therapeutic strategy and foundational data for the treatment of depression. METHODS (1) The acute inflammation model used lipopolysaccharide (LPS) to induce depression-like behavior in mice by injecting LPS intraperitoneally at a dose of 0.83 mg/kg. The effects of PB (20 mg/kg, i.p.) and the NLRP3 inflammasome inhibitor MCC950 (10 mg/kg, i.p.) on improving depression behavior in mice were evaluated. (2) To explore the specific mechanism of PB in improving depression-like behavior in LPS mice by regulating NLRP3 and Netrin-1/DCC pathway. RESULTS The results showed that after intraperitoneal injection of LPS, the mice exhibited a significant decrease in body weight, sucrose preference score, and a significant increase in tail suspension immobility time. Treatment with PB and MCC950 increased the sucrose preference score and decreased the tail suspension immobility time. Besides, PB and MCC950 could inhibit the expression of NLRP3 related neuroinflammation, down-regulated the Netrin-1/DCC signaling pathway, and improved hippocampal neuroplasticity in mice. CONCLUSION In conclusion, PB significantly improved LPS-induced depression-like behavior in mice by reducing the expression of hippocampal NLRP3 inflammasome and down-regulating the Netrin-1/DCC signaling pathway. Additionally, PB was found to regulate α-amino-3-hydroxy-5-methyl-4 isoxazole receptor (AMPAR) and postsynaptic density 95 (PSD95), protecting excitatory synaptic transmission and enhancing synaptic plasticity. This study demonstrates the effectiveness of PB in improving depressive symptoms induced by LPS and provides a new strategy for the clinical treatment of depression.
Collapse
Affiliation(s)
- Yi-Lin Wu
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Ting Hu
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Hong Zheng
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Jifeng Feng
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Chenwei Huang
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Xiaona Zhou
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Wei Wang
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| | - Chun-Lei Jiang
- Laboratory of Stress Medicine, Faculty of Psychology, Navy Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| |
Collapse
|
7
|
Wang C, Zeng Y, Wang J, Wang T, Li X, Shen Z, Meng J, Yao X. A genome-wide association study of the racing performance traits in Yili horses based on Blink and FarmCPU models. Sci Rep 2024; 14:27648. [PMID: 39532956 PMCID: PMC11557848 DOI: 10.1038/s41598-024-79014-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Racing performance traits are the main indicators for evaluating the performance and value of sport horses. The aim of this study was to identify the key genes for racing performance traits in Yili horses by performing a genome-wide association study (GWAS). Breeding values for racing performance traits were calculated for Yili horses (n = 827) using an animal model. Genome-wide association analysis of racing performance traits in horses (n = 236) was carried out using the Blink, and FarmCPU models in GAPIT software, and genes within the significant regions were functionally annotated. The results of GWAS showed that a total of 24 significant SNP markers (P < 6.05 × 10- 9) and 22 suggestive SNP markers (P < 1.21 × 10- 7) were identified. Among them, the Blink associated 16 significant SNP loci and FarmCPU associated 12 significant SNP loci. A total of 127 candidate genes (50 significant) were annotated. Among these, CNTN6 (motor coordination), NIPA1 (neuronal development), and DCC (dopamine pathway maturation) may be the main candidate genes affecting speed traits. SHANK2 (neuronal synaptic regulation), ISCA1 (mitochondrial protein assembly), and KCNIP4 (neuronal excitability) may be the main candidate genes affecting ranking score traits. A common locus (ECA1: 22698579) was significantly associated with racing performance traits, and the function of the genes at this locus needs to be studied in depth. These findings will provide new insights into the detection and selection of genetic variants for racing performance and will help to accelerate the genetic improvement of Yili horses.
Collapse
Affiliation(s)
- Chuankun Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Yaqi Zeng
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
- Xinjiang Key Laboratory of Horse Breeding and Exercise Physiology, Urumqi, 830052, China
| | - Jianwen Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
- Xinjiang Key Laboratory of Horse Breeding and Exercise Physiology, Urumqi, 830052, China
| | - Tongliang Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Xueyan Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Zhehong Shen
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Jun Meng
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China.
- Xinjiang Key Laboratory of Horse Breeding and Exercise Physiology, Urumqi, 830052, China.
| | - Xinkui Yao
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China.
- Xinjiang Key Laboratory of Horse Breeding and Exercise Physiology, Urumqi, 830052, China.
| |
Collapse
|
8
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
9
|
Hu YQ, Liu WT, Wu Y, Hu ZB, Tao YC, Zhang Q, Chen JY, Li M, Hu L, Ding YQ. DCC in the cerebral cortex is required for cognitive functions in mouse. Brain Pathol 2024:e13306. [PMID: 39293934 DOI: 10.1111/bpa.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024] Open
Abstract
Schizophrenia (SZ) is a highly heritable mental disorder, and genome-wide association studies have identified the association between deleted in colorectal cancer (DCC) and SZ. Previous study has shown a lowered expression of DCC in the cerebral cortex of SZ patient. In this study, we identified novel single nucleotide polymorphisms (SNPs) of DCC statistically correlated with SZ. Based on these, we generated DCC conditional knockout (CKO) mice and explored behavioral phenotypes in these mice. We observed that deletion of DCC in cortical layer VI but not layer V led to deficits in fear and spatial memory, as well as defective sensorimotor gating revealed by the prepulse inhibition test (PPI). Critically, the defective sensorimotor gating could be restored by olanzapine, an antipsychotic drug. Furthermore, we found that the levels of p-AKT and p-GSK3α/β were decreased, which was responsible for impaired PPI in the DCC-deficient mice. Finally, the DCC-deficient mice also displayed reduced spine density of pyramidal neurons and disturbed delta-oscillations. Our data, for the first time, identified and explored downstream substrates and signaling pathway of DCC which supports the hypothesis that DCC is a SZ-related risky gene and when defective, may promote SZ-like pathogenesis and behavioral phenotypes in mice.
Collapse
Affiliation(s)
- Yun-Qing Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wei-Tang Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Jianghan University, Wuhan, China
| | - Zhi-Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yun-Chao Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qiong Zhang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Hua Y, Wang M, Yao Q, Hu B, Lu F, Fan Y, Lu W. Association between plasma Netrin-1 levels and motor and nonmotor symptoms in Parkinson's disease. CNS Neurosci Ther 2024; 30:e70022. [PMID: 39215401 PMCID: PMC11364512 DOI: 10.1111/cns.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/31/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by dopaminergic neuron degeneration and diverse motor and nonmotor symptoms. Early diagnosis and intervention are crucial but challenging due to reliance on clinical presentation. Recent research suggests potential biomarkers for early detection, including plasma netrin-1 (NTN-1), a protein implicated in neuronal survival. METHODS This cross-sectional study recruited 105 PD patients and 65 healthy controls, assessing plasma NTN-1 levels and correlating them with clinical characteristics. Statistical analyses explored associations between NTN-1 levels and PD symptoms, considering demographic factors. RESULTS PD patients exhibited significantly lower plasma NTN-1 levels compared to controls. NTN-1 demonstrated moderate potential as a PD biomarker. Positive correlations were found between NTN-1 levels and motor, depression, and cognitive symptoms. Multiple regression analysis revealed disease duration and NTN-1 levels as key factors influencing symptom severity. Gender also impacted symptom scores. CONCLUSION Reduced plasma NTN-1 levels correlate with PD severity, suggesting its potential as a biomarker. However, further research is needed to elucidate the roles of NTN-1 in PD pathophysiology and validate its diagnostic and therapeutic implications. Understanding the involvement of NTN-1 may lead to personalized management strategies for PD.
Collapse
Affiliation(s)
- Ye Hua
- Department of NeurologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of NeurologyWuxi No. 2 People's Hospital, Jiangnan University Medical CenterWuxiChina
| | - Min Wang
- Department of PharmacologyNeuroprotective Drug Discovery Center of Nanjing Medical University, Nanjing Medical UniversityNanjingChina
| | - Qingyu Yao
- Department of NeurologyWuxi No. 2 People's Hospital, Jiangnan University Medical CenterWuxiChina
| | - Bin Hu
- Department of NeurologyWuxi No. 2 People's Hospital, Jiangnan University Medical CenterWuxiChina
| | - Feng Lu
- Department of NeurologyWuxi No. 2 People's Hospital, Jiangnan University Medical CenterWuxiChina
| | - Yi Fan
- Department of PharmacologyNeuroprotective Drug Discovery Center of Nanjing Medical University, Nanjing Medical UniversityNanjingChina
| | - Weifeng Lu
- Department of NeurologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
11
|
Wei B, Liu W, Jin L, Huang Y, Cheng W, Fan H, Su S, Jin F, Zhang X, Yang Z, Liang S, Li L, Wu Y, Liu Y, Duan C, Li X. Hepcidin depending on astrocytic NEO1 ameliorates blood-brain barrier dysfunction after subarachnoid hemorrhage. Cell Death Dis 2024; 15:569. [PMID: 39107268 PMCID: PMC11303805 DOI: 10.1038/s41419-024-06909-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 08/09/2024]
Abstract
Subarachnoid hemorrhage (SAH) significantly compromises the blood-brain barrier (BBB) and impairs patient recovery. This study elucidates the critical role of astrocytic Neogenin-1 (NEO1) in BBB integrity post-SAH and examines the regulatory effects of hepcidin on endothelial cell (EC) function amid NEO1-mediated disruptions in iron homeostasis. Proteomic analyses of cerebrospinal fluid (CSF) from SAH patients revealed a substantial decrease in NEO1 expression, identifying it as a key factor in BBB integrity. 111 CSF proteins were significantly reduced in early SAH stages (days 1-3), with NEO1 among the most significantly altered. This dysregulation was linked to poorer patient outcomes, as indicated by a negative correlation between NEO1 levels and Modified Rankin Scale scores six months post-SAH (R = -0.4743, P < 0.0001). Experimental models further highlighted the importance of NEO1: SAH model and NEO1GFAP-Cre mice exhibited exacerbated EC dysfunction and increased BBB permeability, evidenced by significant Evans Blue retention and dextran leakage in the parietal cortex, effects that were mitigated by hepcidin administration. Our findings highlight the complex interplay between astrocytic signaling and endothelial function in SAH pathophysiology. The loss of astrocytic NEO1 led to increased EC proliferation and altered BBB structure, as confirmed by transmission electron microscopy and immunostaining for PECAM-1, indicating heightened blood vessel density in the affected cortex. Hepcidin treatment effectively reversed the EC dysfunction and BBB disruption in both NEO1-cKO mice and the SAH model, highlighting its potential as a therapeutic agent to enhance recovery and improve prognosis following SAH.
Collapse
Affiliation(s)
- Boyang Wei
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wenchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Lei Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yaxian Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wenping Cheng
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haiyan Fan
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Shixing Su
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Fa Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xin Zhang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zeyu Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shuyin Liang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Longxiang Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yu Wu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yanchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Xifeng Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
12
|
Sokolov AV, Schiöth HB. Decoding depression: a comprehensive multi-cohort exploration of blood DNA methylation using machine learning and deep learning approaches. Transl Psychiatry 2024; 14:287. [PMID: 39009577 PMCID: PMC11250806 DOI: 10.1038/s41398-024-02992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
The causes of depression are complex, and the current diagnosis methods rely solely on psychiatric evaluations with no incorporation of laboratory biomarkers in clinical practices. We investigated the stability of blood DNA methylation depression signatures in six different populations using six public and two domestic cohorts (n = 1942) conducting mega-analysis and meta-analysis of the individual studies. We evaluated 12 machine learning and deep learning strategies for depression classification both in cross-validation (CV) and in hold-out tests using merged data from 8 separate batches, constructing models with both biased and unbiased feature selection. We found 1987 CpG sites related to depression in both mega- and meta-analysis at the nominal level, and the associated genes were nominally related to axon guidance and immune pathways based on enrichment analysis and eQTM data. Random forest classifiers achieved the highest performance (AUC 0.73 and 0.76) in CV and hold-out tests respectively on the batch-level processed data. In contrast, the methylation showed low predictive power (all AUCs < 0.57) for all classifiers in CV and no predictive power in hold-out tests when used with harmonized data. All models achieved significantly better performance (>14% gain in AUCs) with pre-selected features (selection bias), with some of the models (joint autoencoder-classifier) reaching AUCs of up to 0.91 in the final testing regardless of data preparation. Different algorithmic feature selection approaches may outperform limma, however, random forest models perform well regardless of the strategy. The results provide an overview over potential future biomarkers for depression and highlight many important methodological aspects for DNA methylation-based depression profiling including the use of machine learning strategies.
Collapse
Affiliation(s)
- Aleksandr V Sokolov
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
13
|
Bastawy EM, Eraslan IM, Voglsanger L, Suphioglu C, Walker AJ, Dean OM, Read JL, Ziemann M, Smith CM. Novel Insights into Changes in Gene Expression within the Hypothalamus in Two Asthma Mouse Models: A Transcriptomic Lung-Brain Axis Study. Int J Mol Sci 2024; 25:7391. [PMID: 39000495 PMCID: PMC11242700 DOI: 10.3390/ijms25137391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Patients with asthma experience elevated rates of mental illness. However, the molecular links underlying such lung-brain crosstalk remain ambiguous. Hypothalamic dysfunction is observed in many psychiatric disorders, particularly those with an inflammatory component due to many hypothalamic regions being unprotected by the blood-brain barrier. To gain a better insight into such neuropsychiatric sequelae, this study investigated gene expression differences in the hypothalamus following lung inflammation (asthma) induction in mice, using RNA transcriptome profiling. BALB/c mice were challenged with either bacterial lipopolysaccharide (LPS, E. coli) or ovalbumin (OVA) allergens or saline control (n = 7 per group), and lung inflammation was confirmed via histological examination of postmortem lung tissue. The majority of the hypothalamus was micro-dissected, and total RNA was extracted for sequencing. Differential expression analysis identified 31 statistically significant single genes (false discovery rate FDR5%) altered in expression following LPS exposure compared to controls; however, none were significantly changed following OVA treatment, suggesting a milder hypothalamic response. When gene sets were examined, 48 were upregulated and 8 were downregulated in both asthma groups relative to controls. REACTOME enrichment analysis suggests these gene sets are involved in signal transduction metabolism, immune response and neuroplasticity. Interestingly, we identified five altered gene sets directly associated with neurotransmitter signaling. Intriguingly, many of these altered gene sets can influence mental health and or/neuroinflammation in humans. These findings help characterize the links between asthma-induced lung inflammation and the brain and may assist in identifying relevant pathways and therapeutic targets for future intervention.
Collapse
Affiliation(s)
- Eslam M Bastawy
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Izel M Eraslan
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Lara Voglsanger
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Cenk Suphioglu
- Faculty of Science, Engineering and Built Environment, School of Life and Environmental Sciences, Deakin University, Geelong 3216, Australia
| | - Adam J Walker
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Olivia M Dean
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne 3052, Australia
| | - Justin L Read
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Mark Ziemann
- Faculty of Science, Engineering and Built Environment, School of Life and Environmental Sciences, Deakin University, Geelong 3216, Australia
- Burnet Institute, Melbourne 3004, Australia
| | - Craig M Smith
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| |
Collapse
|
14
|
Hoops D, Yee Y, Hammill C, Wong S, Manitt C, Bedell BJ, Cahill L, Lerch JP, Flores C, Sled JG. Disproportionate neuroanatomical effects of DCC haploinsufficiency in adolescence compared with adulthood: links to dopamine, connectivity, covariance, and gene expression brain maps in mice. J Psychiatry Neurosci 2024; 49:E157-E171. [PMID: 38692693 PMCID: PMC11068426 DOI: 10.1503/jpn.230106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/23/2024] [Accepted: 03/06/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Critical adolescent neural refinement is controlled by the DCC (deleted in colorectal cancer) protein, a receptor for the netrin-1 guidance cue. We sought to describe the effects of reduced DCC on neuroanatomy in the adolescent and adult mouse brain. METHODS We examined neuronal connectivity, structural covariance, and molecular processes in a DCC-haploinsufficient mouse model, compared with wild-type mice, using new, custom analytical tools designed to leverage publicly available databases from the Allen Institute. RESULTS We included 11 DCC-haploinsufficient mice and 16 wild-type littermates. Neuroanatomical effects of DCC haploinsufficiency were more severe in adolescence than adulthood and were largely restricted to the mesocorticolimbic dopamine system. The latter finding was consistent whether we identified the regions of the mesocorticolimbic dopamine system a priori or used connectivity data from the Allen Brain Atlas to determine de novo where these dopamine axons terminated. Covariance analyses found that DCC haploinsufficiency disrupted the coordinated development of the brain regions that make up the mesocorticolimbic dopamine system. Gene expression maps pointed to molecular processes involving the expression of DCC, UNC5C (encoding DCC's co-receptor), and NTN1 (encoding its ligand, netrin-1) as underlying our structural findings. LIMITATIONS Our study involved a single sex (males) at only 2 ages. CONCLUSION The neuroanatomical phenotype of DCC haploinsufficiency described in mice parallels that observed in DCC-haploinsufficient humans. It is critical to understand the DCC-haploinsufficient mouse as a clinically relevant model system.
Collapse
Affiliation(s)
- Daniel Hoops
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Yohan Yee
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Christopher Hammill
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Sammi Wong
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Colleen Manitt
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Barry J Bedell
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Lindsay Cahill
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Jason P Lerch
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Cecilia Flores
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - John G Sled
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| |
Collapse
|
15
|
Cai M, Zheng Q, Chen Y, Liu S, Zhu H, Bai B. Insights from the neural guidance factor Netrin-1 into neurodegeneration and other diseases. Front Mol Neurosci 2024; 17:1379726. [PMID: 38638604 PMCID: PMC11024333 DOI: 10.3389/fnmol.2024.1379726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Netrin-1 was initially discovered as a neuronal growth cue for axonal guidance, and its functions have later been identified in inflammation, tumorigenesis, neurodegeneration, and other disorders. We have recently found its alterations in the brains with Alzheimer's disease, which might provide important clues to the mechanisms of some unique pathologies. To provide better understanding of this promising molecule, we here summarize research progresses in genetics, pathology, biochemistry, cell biology and other studies of Netrin-1 about its mechanistic roles and biomarker potentials with an emphasis on clinical neurodegenerative disorders in order to expand understanding of this promising molecular player in human diseases.
Collapse
Affiliation(s)
- Minqi Cai
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Qian Zheng
- Health Management Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yiqiang Chen
- Center for Precision Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Siyuan Liu
- Center for Precision Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huimin Zhu
- Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Bing Bai
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Baranoglu Kilinc Y, Torun IE, Kilinc E. D2 dopamine receptor-mediated mechanisms of dopaminergic system modulation in in vivo and in vitro experimental models of migraine. Eur J Neurosci 2024; 59:1177-1193. [PMID: 37539658 DOI: 10.1111/ejn.16106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
The dopaminergic system is implicated in the pathophysiology of migraine. However, the underlying mechanisms remain unclear. We explored the effects and mechanisms of dopaminergic system modulation in the in vivo and in vitro rat models of migraine. Dopaminergic agonist apomorphine, D2 receptor antagonists metoclopramide and haloperidol and 5-HT3 receptor antagonist ondansetron alone and together were tested in nitroglycerin-induced migraine model, in vivo. Likewise, the combinations of drugs were also tested on basal calcitonin gene-related peptide (CGRP) release in vitro hemiskull preparations. Mechanical allodynia was tested by von Frey filaments. CGRP concentrations in trigeminovascular structures and in vitro superfusates and c-Fos levels in the brainstem were determined by enzyme-linked immunosorbent assay. Meningeal mast cells were evaluated with toluidine blue staining. Apomorphine further enhanced nitroglycerin-induced mechanical allodynia, brainstem c-fos expression, trigeminal ganglion and brainstem CGRP concentrations and meningeal mast cell degranulation, in vivo. Haloperidol completely antagonised all apomorphine-induced effects and also alleviated changes induced by nitroglycerin without apomorphine. Metoclopramide and ondansetron partially attenuated apomorphine- or nitroglycerin-induced effects. A combination of haloperidol and ondansetron decreased basal CGRP release, in vitro, whereas the other administrations were ineffective. Apomorphine-mediated dopaminergic activation exacerbated nitroglycerin-stimulated nociceptive reactions by further enhancing c-fos expression, CGRP release and mast cell degranulation in strategical structures associated with migraine pain. Metoclopramide partially attenuated the effects of apomorphine, most likely because it is also a 5-HT3 receptor antagonist. Haloperidol with pure D2 receptor antagonism feature appears to be more effective than metoclopramide in reducing migraine-related parameters in dopaminergic activation- and/or NTG-induced migraine-like conditions.
Collapse
Affiliation(s)
| | - Ibrahim Ethem Torun
- Department of Physiology, Medical School, University of Bolu Abant Izzet Baysal, Bolu, Turkey
| | - Erkan Kilinc
- Department of Physiology, Medical School, University of Bolu Abant Izzet Baysal, Bolu, Turkey
| |
Collapse
|
17
|
Haroon H, Ho AMC, Gupta VK, Dasari S, Sellgren CM, Cervenka S, Engberg G, Eren F, Erhardt S, Sung J, Choi DS. Cerebrospinal fluid proteomic signatures are associated with symptom severity of first-episode psychosis. J Psychiatr Res 2024; 171:306-315. [PMID: 38340697 PMCID: PMC10995989 DOI: 10.1016/j.jpsychires.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Apart from their diagnostic, monitoring, or prognostic utility in clinical settings, molecular biomarkers may be instrumental in understanding the pathophysiology of psychiatric disorders, including schizophrenia. Using untargeted metabolomics, we recently identified eight cerebrospinal fluid (CSF) metabolites unique to first-episode psychosis (FEP) subjects compared to healthy controls (HC). In this study, we sought to investigate the CSF proteomic signatures associated with FEP. We employed 16-plex tandem mass tag (TMT) mass spectrometry (MS) to examine the relative protein abundance in CSF samples of 15 individuals diagnosed with FEP and 15 age-and-sex-matched healthy controls (HC). Multiple linear regression model (MLRM) identified 16 differentially abundant CSF proteins between FEP and HC at p < 0.01. Among them, the two most significant CSF proteins were collagen alpha-2 (IV) chain (COL4A2: standard mean difference [SMD] = -1.12, p = 1.64 × 10-4) and neuron-derived neurotrophic factor (NDNF: SMD = -1.03, p = 4.52 × 10-4) both of which were down-regulated in FEP subjects compared to HC. We also identified several potential CSF proteins associated with the pathophysiology and the symptom profile and severity in FEP subjects, including COL4A2, NDNF, hornerin (HRNR), contactin-6 (CNTN6), voltage-dependent calcium channel subunit alpha-2/delta-3 (CACNA2D3), tropomyosin alpha-3 chain (TPM3 and TPM4). Moreover, several protein signatures were associated with cognitive performance. Although the results need replication, our exploratory study suggests that CSF protein signatures can be used to increase the understanding of the pathophysiology of psychosis.
Collapse
Affiliation(s)
- Humza Haroon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Ada Man-Choi Ho
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Vinod K Gupta
- Division of Surgery Research, Department of Surgery, Rochester, MN, USA; Microbiome Program, Center for Individualized Medicine, Rochester, MN, USA
| | - Surendra Dasari
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden; Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Feride Eren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jaeyun Sung
- Division of Surgery Research, Department of Surgery, Rochester, MN, USA; Microbiome Program, Center for Individualized Medicine, Rochester, MN, USA; Division of Rheumatology, Department of Internal Medicine, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| |
Collapse
|
18
|
Treccarichi S, Failla P, Vinci M, Musumeci A, Gloria A, Vasta A, Calabrese G, Papa C, Federico C, Saccone S, Calì F. UNC5C: Novel Gene Associated with Psychiatric Disorders Impacts Dysregulation of Axon Guidance Pathways. Genes (Basel) 2024; 15:306. [PMID: 38540364 PMCID: PMC10970690 DOI: 10.3390/genes15030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 06/14/2024] Open
Abstract
The UNC-5 family of netrin receptor genes, predominantly expressed in brain tissues, plays a pivotal role in various neuronal processes. Mutations in genes involved in axon development contribute to a wide spectrum of human diseases, including developmental, neuropsychiatric, and neurodegenerative disorders. The NTN1/DCC signaling pathway, interacting with UNC5C, plays a crucial role in central nervous system axon guidance and has been associated with psychiatric disorders during adolescence in humans. Whole-exome sequencing analysis unveiled two compound heterozygous causative mutations within the UNC5C gene in a patient diagnosed with psychiatric disorders. In silico analysis demonstrated that neither of the observed variants affected the allosteric linkage between UNC5C and NTN1. In fact, these mutations are located within crucial cytoplasmic domains, specifically ZU5 and the region required for the netrin-mediated axon repulsion of neuronal growth cones. These domains play a critical role in forming the supramodular protein structure and directly interact with microtubules, thereby ensuring the functionality of the axon repulsion process. We emphasize that these mutations disrupt the aforementioned processes, thereby associating the UNC5C gene with psychiatric disorders for the first time and expanding the number of genes related to psychiatric disorders. Further research is required to validate the correlation of the UNC5C gene with psychiatric disorders, but we suggest including it in the genetic analysis of patients with psychiatric disorders.
Collapse
Affiliation(s)
- Simone Treccarichi
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Pinella Failla
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Mirella Vinci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Antonino Musumeci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Angelo Gloria
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Anna Vasta
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Giuseppe Calabrese
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Carla Papa
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| | - Concetta Federico
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy;
| | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy;
| | - Francesco Calì
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (S.T.); (P.F.); (M.V.); (A.M.); (A.G.); (A.V.); (G.C.); (C.P.); (F.C.)
| |
Collapse
|
19
|
Begni V, Marizzoni M, Creutzberg KC, Silipo DM, Papp M, Cattaneo A, Riva MA. Transcriptomic analyses of rats exposed to chronic mild stress: Modulation by chronic treatment with the antipsychotic drug lurasidone. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110885. [PMID: 37865392 DOI: 10.1016/j.pnpbp.2023.110885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Exposure to stressful experiences accounts for almost half of the risk for mental disorders. Hence, stress-induced alterations represent a key target for pharmacological interventions aimed at restoring brain function in affected individuals. We have previously demonstrated that lurasidone, a multi-receptor antipsychotic drug approved for the treatment of schizophrenia and bipolar depression, can normalize the functional and molecular impairments induced by stress exposure, representing a valuable tool for the treatment of stress-induced mental illnesses. However, the mechanisms that may contribute to the therapeutic effects of lurasidone are still poorly understood. Here, we performed a transcriptomic analysis on the prefrontal cortex (PFC) of adult male rats exposed to the chronic mild stress (CMS) paradigm and we investigated the impact of chronic lurasidone treatment on such changes. We found that CMS exposure leads to an anhedonic phenotype associated with a down-regulation of different pathways associated to neuronal guidance and synaptic plasticity within the PFC. Interestingly, a significant part of these alterations (around 25%) were counteracted by lurasidone treatment. In summary, we provided new insights on the transcriptional changes relevant for the therapeutic intervention with lurasidone, which may ultimately promote resilience.
Collapse
Affiliation(s)
- Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy; Lab of Neuroimaging and Alzheimer's Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Kerstin Camile Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Diana Morena Silipo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Mariusz Papp
- Institute of Pharmacology and Polish Academy of Sciences, Smętna Street 12, Kraków 31-343, Poland
| | - Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy.
| |
Collapse
|
20
|
Wang M, Fan J, Shao Z. Cellular and Molecular Mechanisms Underlying Synaptic Subcellular Specificity. Brain Sci 2024; 14:155. [PMID: 38391729 PMCID: PMC10886843 DOI: 10.3390/brainsci14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 02/24/2024] Open
Abstract
Chemical synapses are essential for neuronal information storage and relay. The synaptic signal received or sent from spatially distinct subcellular compartments often generates different outcomes due to the distance or physical property difference. Therefore, the final output of postsynaptic neurons is determined not only by the type and intensity of synaptic inputs but also by the synaptic subcellular location. How synaptic subcellular specificity is determined has long been the focus of study in the neurodevelopment field. Genetic studies from invertebrates such as Caenorhabditis elegans (C. elegans) have uncovered important molecular and cellular mechanisms required for subcellular specificity. Interestingly, similar molecular mechanisms were found in the mammalian cerebellum, hippocampus, and cerebral cortex. This review summarizes the comprehensive advances in the cellular and molecular mechanisms underlying synaptic subcellular specificity, focusing on studies from C. elegans and rodents.
Collapse
Affiliation(s)
- Mengqing Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| | - Jiale Fan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| | - Zhiyong Shao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| |
Collapse
|
21
|
Panoyan MA, Shi Y, Abbatangelo CL, Adler N, Moo-Choy A, Parra EJ, Polimanti R, Hu P, Wendt FR. Exome-wide tandem repeats confer large effects on subcortical volumes in UK Biobank participants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.11.23299818. [PMID: 38168307 PMCID: PMC10760277 DOI: 10.1101/2023.12.11.23299818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The human subcortex is involved in memory and cognition. Structural and functional changes in subcortical regions is implicated in psychiatric conditions. We performed an association study of subcortical volumes using 15,941 tandem repeats (TRs) derived from whole exome sequencing (WES) data in 16,527 unrelated European ancestry participants. We identified 17 loci, most of which were associated with accumbens volume, and nine of which had fine-mapping probability supporting their causal effect on subcortical volume independent of surrounding variation. The most significant association involved NTN1 -[GCGG] N and increased accumbens volume (β=5.93, P=8.16x10 -9 ). Three exonic TRs had large effects on thalamus volume ( LAT2 -[CATC] N β=-949, P=3.84x10 -6 and SLC39A4 -[CAG] N β=-1599, P=2.42x10 -8 ) and pallidum volume ( MCM2 -[AGG] N β=-404.9, P=147x10 -7 ). These genetic effects were consistent measurements of per-repeat expansion/contraction effects on organism fitness. With 3-dimensional modeling, we reinforced these effects to show that the expanded and contracted LAT2 -[CATC] N repeat causes a frameshift mutation that prevents appropriate protein folding. These TRs also exhibited independent effects on several psychiatric symptoms, including LAT2 -[CATC] N and the tiredness/low energy symptom of depression (β=0.340, P=0.003). These findings link genetic variation to tractable biology in the brain and relevant psychiatric symptoms. We also chart one pathway for TR prioritization in future complex trait genetic studies.
Collapse
|
22
|
Ma Z, Wei Y, Liao T, Jie L, Yang N, Yu L, Wang P. Activation of vascular endothelial cells by synovial fibrosis promotes Netrin-1-induced sensory nerve sprouting and exacerbates pain sensitivity. J Cell Mol Med 2023; 27:3773-3785. [PMID: 37702437 PMCID: PMC10718133 DOI: 10.1111/jcmm.17950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Synovial fibrosis is one of the most dominant histopathological changes in osteoarthritis of the knee (KOA), and activation of vascular endothelial cells in synovial fibrosis is both an important factor in mediating pain in KOA and a major contributor to the generation of pain signals. At the same time, angiogenesis and nerve fibres are more likely to underlie the pathology of pain induced by synovial fibrosis. In the present study, we established a co-culture model of human umbilical vein endothelial cells (HUVECs) with dorsal root ganglion (DRG) and detected tissue and cellular Netrin-1, vascular cell adhesion molecule-1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), growth-associated protein-43 (GAP43), colorectal cancer deleted (DCC), uncoordinated 5 (UNC5), and the related expression of calcitonin gene-related peptide (CGRP), substance P (SP) and nerve growth factor (NGF) in supernatant by ELISA to investigate the intervention of vascular endothelial cell activation on sensory nerve sprouting exacerbating peripheral pain sensitivity and to investigate the effect of Netrin-1 from the perspective of Netrin-1 secretion to illustrate its effector mechanism.
Collapse
Affiliation(s)
- Zhenyuan Ma
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Yibao Wei
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Taiyang Liao
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Lishi Jie
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Nan Yang
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Likai Yu
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Peimin Wang
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and ApplicationNanjingChina
| |
Collapse
|
23
|
Managò F, Scheggia D, Pontillo M, Mereu M, Mastrogiacomo R, Udayan G, Valentini P, Tata MC, Weinberger DR, Weickert CS, Pompa PP, De Luca MA, Vicari S, Papaleo F. Dopaminergic signalling and behavioural alterations by Comt-Dtnbp1 genetic interaction and their clinical relevance. Br J Pharmacol 2023; 180:2514-2531. [PMID: 37218669 DOI: 10.1111/bph.16147] [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: 06/13/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Cognitive and motor functions are modulated by dopaminergic signalling, which is shaped by several genetic factors. The biological effects of single genetic variants might differ depending on epistatic interactions that can be functionally multi-directional and non-linear. EXPERIMENTAL APPROACH We performed behavioural and neurochemical assessments in genetically modified mice and behavioural assessments and genetic screening in human patients with 22q11.2 deletion syndrome (22q11.2DS). KEY RESULTS Here, we confirm a genetic interaction between the Comt (catechol-O-methyltransferase, human orthologue: COMT) and Dtnbp1 (dystrobrevin binding protein 1, alias dysbindin, human orthologue: DTNBP1) genes that modulate cortical and striatal dopaminergic signalling in a manner not predictable by the effects of each single gene. In mice, Comt-by-Dtnbp1 concomitant reduction leads to a hypoactive mesocortical and a hyperactive mesostriatal dopamine pathway, associated with specific cognitive abnormalities. Like mice, in subjects with the 22q11.2DS (characterized by COMT hemideletion and dopamine alterations), COMT-by-DTNBP1 concomitant reduction was associated with analogous cognitive disturbances. We then developed an easy and inexpensive colourimetric kit for the genetic screening of common COMT and DTNBP1 functional genetic variants for clinical application. CONCLUSIONS AND IMPLICATIONS These findings illustrate an epistatic interaction of two dopamine-related genes and their functional effects, supporting the need to address genetic interaction mechanisms at the base of complex behavioural traits.
Collapse
Affiliation(s)
- Francesca Managò
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Diego Scheggia
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Pontillo
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maddalena Mereu
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rosa Mastrogiacomo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Gayatri Udayan
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Paola Valentini
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Cynthia S Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Pier Paolo Pompa
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Maria A De Luca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Vicari
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
24
|
Batra A, Cuesta S, Alves MB, Restrepo JM, Giroux M, Laureano DP, Mucellini Lovato AB, Miguel PM, Machado TD, Molle RD, Flores C, Silveira PP. Relationship between insulin and Netrin-1/DCC guidance cue pathway regulation in the prefrontal cortex of rodents exposed to prenatal dietary restriction. J Dev Orig Health Dis 2023; 14:501-507. [PMID: 37431265 PMCID: PMC10988268 DOI: 10.1017/s204017442300017x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Fetal restriction (FR) alters insulin sensitivity, but it is unknown how the metabolic profile associated with restriction affects development of the dopamine (DA) system and DA-related behaviors. The Netrin-1/DCC guidance cue system participates in maturation of the mesocorticolimbic DA circuitry. Therefore, our objective was to identify if FR modifies Netrin-1/DCC receptor protein expression in the prefrontal cortex (PFC) at birth and mRNA in adulthood in rodent males. We used cultured HEK293 cells to assess if levels of miR-218, microRNA regulator of DCC, are sensitive to insulin. To assess this, pregnant dams were subjected to a 50% FR diet from gestational day 10 until birth. Medial PFC (mPFC) DCC/Netrin-1 protein expression was measured at P0 at baseline and Dcc/Netrin-1 mRNA levels were quantified in adults 15 min after a saline/insulin injection. miR-218 levels in HEK-293 cells were measured in response to insulin exposure. At P0, Netrin-1 levels are downregulated in FR animals in comparison to controls. In adult rodents, insulin administration results in an increase in Dcc mRNA levels in control but not FR rats. In HEK293 cells, there is a positive correlation between insulin concentration and miR-218 levels. Since miR-218 is a Dcc gene expression regulator and our in vitro results show that insulin regulates miR-218 levels, we suggest that FR-induced changes in insulin sensitivity could be affecting Dcc expression via miR-218, impacting DA system maturation and organization. As fetal adversity is linked to nonadaptive behaviors later in life, this may contribute to early identification of vulnerability to chronic diseases associated with fetal adversity.
Collapse
Affiliation(s)
- Aashita Batra
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Santiago Cuesta
- Department of Cell Biology and Neuroscience, Rutgers University, New Brunswick, NJ, USA
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Marcio Bonesso Alves
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Jose Maria Restrepo
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Michel Giroux
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Daniela Pereira Laureano
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda Brondani Mucellini Lovato
- Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patrícia Maidana Miguel
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Tania Diniz Machado
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberta Dalle Molle
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
25
|
Reynolds LM, Hernandez G, MacGowan D, Popescu C, Nouel D, Cuesta S, Burke S, Savell KE, Zhao J, Restrepo-Lozano JM, Giroux M, Israel S, Orsini T, He S, Wodzinski M, Avramescu RG, Pokinko M, Epelbaum JG, Niu Z, Pantoja-Urbán AH, Trudeau LÉ, Kolb B, Day JJ, Flores C. Amphetamine disrupts dopamine axon growth in adolescence by a sex-specific mechanism in mice. Nat Commun 2023; 14:4035. [PMID: 37419977 PMCID: PMC10329029 DOI: 10.1038/s41467-023-39665-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Initiating drug use during adolescence increases the risk of developing addiction or other psychopathologies later in life, with long-term outcomes varying according to sex and exact timing of use. The cellular and molecular underpinnings explaining this differential sensitivity to detrimental drug effects remain unexplained. The Netrin-1/DCC guidance cue system segregates cortical and limbic dopamine pathways in adolescence. Here we show that amphetamine, by dysregulating Netrin-1/DCC signaling, triggers ectopic growth of mesolimbic dopamine axons to the prefrontal cortex, only in early-adolescent male mice, underlying a male-specific vulnerability to enduring cognitive deficits. In adolescent females, compensatory changes in Netrin-1 protect against the deleterious consequences of amphetamine on dopamine connectivity and cognitive outcomes. Netrin-1/DCC signaling functions as a molecular switch which can be differentially regulated by the same drug experience as function of an individual's sex and adolescent age, and lead to divergent long-term outcomes associated with vulnerable or resilient phenotypes.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Plasticité du Cerveau CNRS UMR8249, École supérieure de physique et de chimie industrielles de la Ville de Paris (ESPCI Paris), Paris, France
| | | | - Del MacGowan
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Christina Popescu
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Dominique Nouel
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Santiago Cuesta
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Samuel Burke
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Katherine E Savell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Janet Zhao
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Jose Maria Restrepo-Lozano
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Michel Giroux
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Sonia Israel
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Taylor Orsini
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Susan He
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | | | - Radu G Avramescu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Matthew Pokinko
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Julia G Epelbaum
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Zhipeng Niu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Andrea Harée Pantoja-Urbán
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Louis-Éric Trudeau
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Bryan Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
| |
Collapse
|
26
|
Mallard TT, Grotzinger AD, Smoller JW. Examining the shared etiology of psychopathology with genome-wide association studies. Physiol Rev 2023; 103:1645-1665. [PMID: 36634217 PMCID: PMC9988537 DOI: 10.1152/physrev.00016.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Genome-wide association studies (GWASs) have ushered in a new era of reproducible discovery in psychiatric genetics. The field has now identified hundreds of common genetic variants that are associated with mental disorders, and many of them influence more than one disorder. By advancing the understanding of causal biology underlying psychopathology, GWAS results are poised to inform the development of novel therapeutics, stratification of at-risk patients, and perhaps even the revision of top-down classification systems in psychiatry. Here, we provide a concise review of GWAS findings with an emphasis on findings that have elucidated the shared genetic etiology of psychopathology, summarizing insights at three levels of analysis: 1) genome-wide architecture; 2) networks, pathways, and gene sets; and 3) individual variants/genes. Three themes emerge from these efforts. First, all psychiatric phenotypes are heritable, highly polygenic, and influenced by many pleiotropic variants with incomplete penetrance. Second, GWAS results highlight the broad etiological roles of neuronal biology, system-wide effects over localized effects, and early neurodevelopment as a critical period. Third, many loci that are robustly associated with multiple forms of psychopathology harbor genes that are involved in synaptic structure and function. Finally, we conclude our review by discussing the implications that GWAS results hold for the field of psychiatry, as well as expected challenges and future directions in the next stage of psychiatric genetics.
Collapse
Affiliation(s)
- Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, United States
| | - Andrew D Grotzinger
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, United States
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, United States
| |
Collapse
|
27
|
Mahmud A, Avramescu RG, Niu Z, Flores C. Awakening the dormant: Role of axonal guidance cues in stress-induced reorganization of the adult prefrontal cortex leading to depression-like behavior. Front Neural Circuits 2023; 17:1113023. [PMID: 37035502 PMCID: PMC10079902 DOI: 10.3389/fncir.2023.1113023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic and disabling disorder affecting roughly 280 million people worldwide. While multiple brain areas have been implicated, dysfunction of prefrontal cortex (PFC) circuitry has been consistently documented in MDD, as well as in animal models for stress-induced depression-like behavioral states. During brain development, axonal guidance cues organize neuronal wiring by directing axonal pathfinding and arborization, dendritic growth, and synapse formation. Guidance cue systems continue to be expressed in the adult brain and are emerging as important mediators of synaptic plasticity and fine-tuning of mature neural networks. Dysregulation or interference of guidance cues has been linked to depression-like behavioral abnormalities in rodents and MDD in humans. In this review, we focus on the emerging role of guidance cues in stress-induced changes in adult prefrontal cortex circuitry and in precipitating depression-like behaviors. We discuss how modulating axonal guidance cue systems could be a novel approach for precision medicine and the treatment of depression.
Collapse
Affiliation(s)
- Ashraf Mahmud
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | | | - Zhipeng Niu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Department of Psychiatry, Neurology, and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
28
|
Cline MM, Juarez B, Hunker A, Regiarto EG, Hariadi B, Soden ME, Zweifel LS. Netrin-1 regulates the balance of synaptic glutamate signaling in the adult ventral tegmental area. eLife 2023; 12:e83760. [PMID: 36927614 PMCID: PMC10023152 DOI: 10.7554/elife.83760] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
The axonal guidance cue netrin-1 serves a critical role in neural circuit development by promoting growth cone motility, axonal branching, and synaptogenesis. Within the adult mouse brain, expression of the gene encoding (Ntn1) is highly enriched in the ventral midbrain where it is expressed in both GABAergic and dopaminergic neurons, but its function in these cell types in the adult system remains largely unknown. To address this, we performed viral-mediated, cell-type specific CRISPR-Cas9 mutagenesis of Ntn1 in the ventral tegmental area (VTA) of adult mice. Ntn1 loss-of-function in either cell type resulted in a significant reduction in excitatory postsynaptic connectivity. In dopamine neurons, the reduced excitatory tone had a minimal phenotypic behavioral outcome; however, reduced glutamatergic tone on VTA GABA neurons induced behaviors associated with a hyperdopaminergic phenotype. Simultaneous loss of Ntn1 function in both cell types largely rescued the phenotype observed in the GABA-only mutagenesis. These findings demonstrate an important role for Ntn1 in maintaining excitatory connectivity in the adult midbrain and that a balance in this connectivity within two of the major cell types of the VTA is critical for the proper functioning of the mesolimbic system.
Collapse
Affiliation(s)
- Marcella M Cline
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Barbara Juarez
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Avery Hunker
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Ernesto G Regiarto
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Bryan Hariadi
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Marta E Soden
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| |
Collapse
|
29
|
Xu R, Jin Y, Tang S, Wang W, Sun YE, Liu Y, Zhang W, Hou B, Huang Y, Ma Z. Association between single nucleotide variants and severe chronic pain in older adult patients after lower extremity arthroplasty. J Orthop Surg Res 2023; 18:184. [PMID: 36895017 PMCID: PMC9999576 DOI: 10.1186/s13018-023-03683-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Hip or knee osteoarthritis (OA) is one of the main causes of disability worldwide and occurs mostly in the older adults. Total hip or knee arthroplasty is the most effective method to treat OA. However, severe postsurgical pain leading to a poor prognosis. So, investigating the population genetics and genes related to severe chronic pain in older adult patients after lower extremity arthroplasty is helpful to improve the quality of treatment. METHODS We collected blood samples from elderly patients who underwent lower extremity arthroplasty from September 2020 to February 2021 at the Drum Tower Hospital Affiliated to Nanjing University Medical School. The enrolled patients provided measures of pain intensity using the numerical rating scale on the 90th day after surgery. Patients were divided into the case group (Group A) and the control group (Group B) including 10 patients respectively by the numerical rating scale. DNA was isolated from the blood samples of the two groups for whole-exome sequencing. RESULTS In total, 661 variants were identified in the 507 gene regions that were significantly different between both groups (P < 0.05), including CASP5, RASGEF1A, CYP4B1, etc. These genes are mainly involved in biological processes, including cell-cell adhesion, ECM-receptor interaction, metabolism, secretion of bioactive substances, ion binding and transport, regulation of DNA methylation, and chromatin assembly. CONCLUSIONS The current study shows some variants within genes are significantly associated with severe postsurgical chronic pain in older adult patients after lower extremity arthroplasty, indicating a genetic predisposition for chronic postsurgical pain. The study was registered according to ICMJE guidelines. The trial registration number is ChiCTR2000031655 and registration date is April 6th, 2020.
Collapse
Affiliation(s)
- Rui Xu
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Yinan Jin
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Suhong Tang
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Wenwen Wang
- Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Yu-E Sun
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Yue Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Wei Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Bailing Hou
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Yulin Huang
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
30
|
Darcq E, Nouel D, Hernandez G, Pokinko M, Ash P, Moquin L, Gratton A, Kieffer B, Flores C. Reduced dopamine release in Dcc haploinsufficiency male mice abolishes the rewarding effects of cocaine but not those of morphine and ethanol. Psychopharmacology (Berl) 2023; 240:637-646. [PMID: 36471064 PMCID: PMC10296775 DOI: 10.1007/s00213-022-06288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE The Netrin-1/DCC guidance cue pathway is critically involved in the adolescent organization of the mesocorticolimbic dopamine circuitry. Adult mice heterozygous for Dcc show reduced dopamine release in the nucleus accumbens in response to amphetamine and, in turn, blunted sensitivity to the rewarding effects of this drug. OBJECTIVE Here, we tested whether the protective effects of Dcc haploinsufficiency are specific to stimulant drugs of abuse or instead extrapolate to opioids and ethanol. METHODS We used the place preference paradigm to measure the rewarding effects of cocaine (20 mg/kg), morphine (5 or 10 mg/Kg), or ethanol (20%) in adult (PND 75) male Dcc haploinsufficient mice or their wild-type litter mates. In a second experiment, we compared in these two genotypes, in vivo dopamine release in the nucleus accumbens after a single i.p. injection of morphine (10 mg/kg). RESULTS We found reduced morphine-induced dopamine release in the nucleus accumbens of Dcc haploinsufficient male mice, but, contrary to the effects of stimulant drugs, there is no effect of genotype on morphine-induced conditioned preference. CONCLUSION These findings show that reduced drug-induced mesolimbic dopamine in Dcc haploinsufficient male mice protects specifically against the rewarding effects of stimulant drugs, but not against the rewarding properties of morphine and ethanol. These results suggest that these drugs exert their rewarding effect via different brain circuits.
Collapse
Affiliation(s)
- Emmanuel Darcq
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- INSERM U1114, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Dominique Nouel
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | | | - Matthew Pokinko
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Integrated Program in Neuroscience (IPN), McGill University, Montréal, Québec, Canada
| | - Polina Ash
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Integrated Program in Neuroscience (IPN), McGill University, Montréal, Québec, Canada
| | - Luc Moquin
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Alain Gratton
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - Brigitte Kieffer
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- INSERM U1114, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, Québec, Canada.
- Department of Psychiatry, McGill University, Montréal, Québec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada.
- Department of Psychiatry, Douglas Mental Health University Institute, Perry Pavilion, Room 2111, 6875 LaSalle Boulevard, Montréal (Verdun), Québec, H4H 1R3, Canada.
| |
Collapse
|
31
|
Ibrahim WW, Sayed RH, Kandil EA, Wadie W. Niacin mitigates blood-brain barrier tight junctional proteins dysregulation and cerebral inflammation in ketamine rat model of psychosis: Role of GPR109A receptor. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110583. [PMID: 35690118 DOI: 10.1016/j.pnpbp.2022.110583] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/05/2022] [Accepted: 06/05/2022] [Indexed: 01/25/2023]
Abstract
Dysregulated inflammatory responses and blood-brain barrier (BBB) dysfunction are recognized as central factors in the development of psychiatric disorders. The present study was designed to evaluate the effect of niacin on BBB integrity in ketamine-induced model of psychosis. Meanwhile, mepenzolate bromide (MPN), a GPR109A receptor blocker, was used to investigate the role of this receptor on the observed niacin's effect. Male Wistar rats received ketamine (30 mg/kg/day, i.p) for 5 consecutive days and then niacin (40 mg/kg/day, p.o), with or without MPN (5 mg/kg/day, i.p), was given for the subsequent 15 days. Three days before the end of experiment, rats were behaviorally tested using open field, novel object recognition, social interaction, and forced swimming tests. Niacin significantly ameliorated ketamine-induced behavioral deficits, amended gamma aminobutyric acid and glutamate concentration, decreased tumor necrosis factor-α and matrix metallopeptidase 9 levels, and increased netrin-1 contents in the hippocampus of rats. Niacin also augmented the hippocampal expression of ZO-1, occludin, and claudin-5 proteins, indicating the ability of niacin to restore the BBB integrity. Moreover, the histopathologic changes in hippocampal neurons were alleviated. Since all the beneficial effects of niacin in the present investigation were partially abolished by the co-administration of MPN; GPR109A receptor was proven to partially mediate the observed antipsychotic effects of niacin. These data revealed that GPR109A-mediated signaling pathways might represent potential targets for therapeutic interventions to prevent or slow the progression of psychosis.
Collapse
Affiliation(s)
- Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
32
|
Chen HM, Chung YCE, Chen HC, Liu YW, Chen IM, Lu ML, Hsiao FSH, Chen CH, Huang MC, Shih WL, Kuo PH. Exploration of the relationship between gut microbiota and fecal microRNAs in patients with major depressive disorder. Sci Rep 2022; 12:20977. [PMID: 36470908 PMCID: PMC9722658 DOI: 10.1038/s41598-022-24773-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Microbiota-gut-brain axis signaling plays a pivotal role in mood disorders. The communication between the host and the gut microbiota may involve complex regulatory networks. Previous evidence showed that host-fecal microRNAs (miRNAs) interactions partly shaped gut microbiota composition. We hypothesized that some miRNAs are correlated with specific bacteria in the fecal samples in patients with major depressive disorder (MDD), and these miRNAs would show enrichment in pathways associated with MDD. MDD patients and healthy controls were recruited to collect fecal samples. We performed 16S ribosome RNA sequence using the Illumina MiSeq sequencers and analysis of 798 fecal miRNAs using the nCounter Human-v2 miRNA Panel in 20 subjects. We calculated the Spearman correlation coefficient for bacteria abundance and miRNA expressions, and analyzed the predicted miRNA pathways by enrichment analysis with false-discovery correction (FDR). A total of 270 genera and 798 miRNAs were detected in the fecal samples. Seven genera (Anaerostipes, Bacteroides, Bifidobacterium, Clostridium, Collinsella, Dialister, and Roseburia) had fold changes greater than one and were present in over 90% of all fecal samples. In particular, Bacteroides and Dialister significantly differed between the MDD and control groups (p-value < 0.05). The correlation coefficients between the seven genera and miRNAs in patients with MDD showed 48 pairs of positive correlations and 36 negative correlations (p-value < 0.01). For miRNA predicted functions, there were 57 predicted pathways with a p-value < 0.001, including MDD-associated pathways, axon guidance, circadian rhythm, dopaminergic synapse, focal adhesion, long-term potentiation, and neurotrophin signaling pathway. In the current pilot study, our findings suggest specific genera highly correlated with the predicted miRNA functions, which might provide clues for the interaction between host factors and gut microbiota via the microbiota-gut-brain axis. Follow-up studies with larger sample sizes and refined experimental design are essential to dissect the roles between gut microbiota and miRNAs for depression.
Collapse
Affiliation(s)
- Hui-Mei Chen
- grid.19188.390000 0004 0546 0241Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 100 Taiwan
| | - Yu-Chu Ella Chung
- grid.19188.390000 0004 0546 0241Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 100 Taiwan ,grid.59784.370000000406229172Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 350 Taiwan
| | - Hsi-Chung Chen
- grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital, Taipei, 100 Taiwan ,grid.412094.a0000 0004 0572 7815Center of Sleep Disorders, National Taiwan University Hospital, Taipei, 100 Taiwan
| | - Yen-Wenn Liu
- grid.260539.b0000 0001 2059 7017Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan
| | - I-Ming Chen
- grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital, Taipei, 100 Taiwan ,grid.19188.390000 0004 0546 0241Institute of Health Policy and Management, College of Public Health, National Taiwan University, Taipei, 100 Taiwan
| | - Mong-Liang Lu
- grid.416930.90000 0004 0639 4389Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan ,grid.412896.00000 0000 9337 0481Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110 Taiwan
| | - Felix Shih-Hsiang Hsiao
- grid.412063.20000 0004 0639 3626Department of Biotechnology and Animal Science, National Ilan University, No. 1, Sec. 1, Shennong Rd., Yilan City, Yilan County, 260007 Taiwan
| | - Chun-Hsin Chen
- grid.416930.90000 0004 0639 4389Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan ,grid.412896.00000 0000 9337 0481Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110 Taiwan
| | - Ming-Chyi Huang
- grid.412896.00000 0000 9337 0481Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110 Taiwan ,Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, 110 Taiwan
| | - Wei-Liang Shih
- grid.19188.390000 0004 0546 0241Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 100 Taiwan ,grid.454740.6Infectious Diseases Research and Education Center, Ministry of Health and Welfare and National Taiwan University, Taipei, 100 Taiwan
| | - Po-Hsiu Kuo
- grid.19188.390000 0004 0546 0241Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 100 Taiwan ,grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital, Taipei, 100 Taiwan ,grid.416930.90000 0004 0639 4389Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
33
|
Wu Y, Wang L, Zhang CY, Li M, Li Y. Genetic similarities and differences among distinct definitions of depression. Psychiatry Res 2022; 317:114843. [PMID: 36115168 DOI: 10.1016/j.psychres.2022.114843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/22/2022] [Accepted: 09/09/2022] [Indexed: 01/04/2023]
Abstract
Depression is a common and complex psychiatric illness with considerable heritability. Genome-wide association studies (GWAS) have been conducted among different definitions of depression based on different diagnostic criteria. However, the heritability explained by different depression GWAS and the identified loci varied widely. To understand the genetic architectures of different definitions of depression, we conducted a series of genetic analyses including linkage disequilibrium score regression (LDSC), Mendelian randomization, and polygenic overlap quantification and identification. Different definitions of depression and other common psychiatric traits were included in this analysis. We found that although genetic correlations between different definitions of depression were relatively high, they showed substantially different genetic correlation and causality with other psychiatric traits. Using bivariate causal mixture mode (MiXeR) and conjunctional false discovery rate (conjFDR) approach, we observed both shared and unique risk loci across different definitions of depression. Further functional mapping with expression quantitative trait loci (eQTL) information from multiple brain tissues and single cell types indicated distinct genes underlying different definitions of depression, and pathways associated with synapses were significantly enriched in the illness. Our study showed that the genetic architectures of different definitions of depression were distinct and genetic studies of depression should be conducted more cautious.
Collapse
Affiliation(s)
- Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China.
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yi Li
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China; Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China; Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, 430012, Hubei, China.
| |
Collapse
|
34
|
Ding H, Ouyang M, Wang J, Xie M, Huang Y, Yuan F, Jia Y, Zhang X, Liu N, Zhang N. Shared genetics between classes of obesity and psychiatric disorders: A large-scale genome-wide cross-trait analysis. J Psychosom Res 2022; 162:111032. [PMID: 36137488 DOI: 10.1016/j.jpsychores.2022.111032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 10/31/2022]
Abstract
AIMS Epidemiological studies demonstrate an association between classes of obesity and psychiatric disorders, although little is known about shared genetics and causality of association. Thus, we aimed to investigate shared genetics and causal link between different classes of obesity and psychiatric disorders. METHODS We used genome-wide association study (GWAS) summary data range from 9725 to 500,199 sample sizes of European descent, conducted a large-scale genome-wide cross-trait association study to investigate genetic overlap between the classes of obesity and anorexia nervosa, attention-deficit/hyperactivity disorder, autism spectrum disorder, bipolar disorder, major depressive disorder, obsessive-compulsive disorder, schizophrenia, anxiety disorders and Tourette syndrome. We conducted transcriptome-wide association study analysis (TWAS) to identified variants regulated gene expression in those related disorders. Finally, pathway enrichment analysis to identified major pathways. RESULTS In the combined analysis, we replicated 211 previously reported loci and discovered 58 novel independent loci that were associated with all three classes of obesity and related psychiatric disorders. Functional analysis revealed that the identified variants regulated gene expression in major tissues belonging to exocrine/endocrine, digestive, circulatory, adipose, digestive, respiratory, and nervous systems, such as DCC, NEGR1, INO80E. Mendelian randomization analyses suggested that there may be a two-way or one-way causal relationship between obesity and psychiatric disorders. CONCLUSION This large-scale genome-wide cross-trait analysis identified shared genetics and potential causal links between classes of obesity and psychiatric disorders (attention deficit hyperactivity disorder, autism spectrum disorder, anorexia nervosa, major depressive disorder, schizophrenia, and obsessive-compulsive disorder). Such shared genetics suggests potential new biological functions in common among them.
Collapse
Affiliation(s)
- Hui Ding
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Mengyuan Ouyang
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Jinyi Wang
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Minyao Xie
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Yanyuan Huang
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Fangzheng Yuan
- School of Psychology, Nanjing Normal University, Nanjing 210023, China
| | - Yunhan Jia
- School of Psychology, Nanjing Normal University, Nanjing 210023, China
| | - Xuedi Zhang
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Na Liu
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| | - Ning Zhang
- The Affiliated Nanjing Brain Hospital of Nanjing Medical Univesity, 264 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
35
|
Dumrongprechachan V, Salisbury RB, Butler L, MacDonald ML, Kozorovitskiy Y. Dynamic proteomic and phosphoproteomic atlas of corticostriatal axons in neurodevelopment. eLife 2022; 11:e78847. [PMID: 36239373 PMCID: PMC9629834 DOI: 10.7554/elife.78847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Mammalian axonal development begins in embryonic stages and continues postnatally. After birth, axonal proteomic landscape changes rapidly, coordinated by transcription, protein turnover, and post-translational modifications. Comprehensive profiling of axonal proteomes across neurodevelopment is limited, with most studies lacking cell-type and neural circuit specificity, resulting in substantial information loss. We create a Cre-dependent APEX2 reporter mouse line and map cell-type-specific proteome of corticostriatal projections across postnatal development. We synthesize analysis frameworks to define temporal patterns of axonal proteome and phosphoproteome, identifying co-regulated proteins and phosphorylations associated with genetic risk for human brain disorders. We discover proline-directed kinases as major developmental regulators. APEX2 transgenic reporter proximity labeling offers flexible strategies for subcellular proteomics with cell type specificity in early neurodevelopment, a critical period for neuropsychiatric disease.
Collapse
Affiliation(s)
- Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- The Chemistry of Life Processes Institute, Northwestern UniversityEvanstonUnited States
| | - Ryan B Salisbury
- Department of Psychiatry, University of PittsburghPittsburghUnited States
| | - Lindsey Butler
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | | | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- The Chemistry of Life Processes Institute, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
36
|
Casas BS, Arancibia-Altamirano D, Acevedo-La Rosa F, Garrido-Jara D, Maksaev V, Pérez-Monje D, Palma V. It takes two to tango: Widening our understanding of the onset of schizophrenia from a neuro-angiogenic perspective. Front Cell Dev Biol 2022; 10:946706. [PMID: 36092733 PMCID: PMC9448889 DOI: 10.3389/fcell.2022.946706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic debilitating mental disorder characterized by perturbations in thinking, perception, and behavior, along with brain connectivity deficiencies, neurotransmitter dysfunctions, and loss of gray brain matter. To date, schizophrenia has no cure and pharmacological treatments are only partially efficacious, with about 30% of patients describing little to no improvement after treatment. As in most neurological disorders, the main descriptions of schizophrenia physiopathology have been focused on neural network deficiencies. However, to sustain proper neural activity in the brain, another, no less important network is operating: the vast, complex and fascinating vascular network. Increasing research has characterized schizophrenia as a systemic disease where vascular involvement is important. Several neuro-angiogenic pathway disturbances have been related to schizophrenia. Alterations, ranging from genetic polymorphisms, mRNA, and protein alterations to microRNA and abnormal metabolite processing, have been evaluated in plasma, post-mortem brain, animal models, and patient-derived induced pluripotent stem cell (hiPSC) models. During embryonic brain development, the coordinated formation of blood vessels parallels neuro/gliogenesis and results in the structuration of the neurovascular niche, which brings together physical and molecular signals from both systems conforming to the Blood-Brain barrier. In this review, we offer an upfront perspective on distinctive angiogenic and neurogenic signaling pathways that might be involved in the biological causality of schizophrenia. We analyze the role of pivotal angiogenic-related pathways such as Vascular Endothelial Growth Factor and HIF signaling related to hypoxia and oxidative stress events; classic developmental pathways such as the NOTCH pathway, metabolic pathways such as the mTOR/AKT cascade; emerging neuroinflammation, and neurodegenerative processes such as UPR, and also discuss non-canonic angiogenic/axonal guidance factor signaling. Considering that all of the mentioned above pathways converge at the Blood-Brain barrier, reported neurovascular alterations could have deleterious repercussions on overall brain functioning in schizophrenia.
Collapse
|
37
|
Schell G, Roy B, Prall K, Dwivedi Y. miR-218: A Stress-Responsive Epigenetic Modifier. Noncoding RNA 2022; 8:ncrna8040055. [PMID: 35893238 PMCID: PMC9326663 DOI: 10.3390/ncrna8040055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Understanding the epigenetic role of microRNAs (miRNAs) has been a critical development in the field of neuropsychiatry and in understanding their underlying pathophysiology. Abnormalities in miRNA expression are often seen as key to the pathogenesis of many stress-associated mental disorders, including major depressive disorder (MDD). Recent advances in omics biology have further contributed to this understanding and expanded the role of miRNAs in networking a diverse array of molecular pathways, which are essentially related to the stress adaptivity of a healthy brain. Studies have highlighted the role of many such miRNAs in causing maladaptive changes in the brain's stress axis. One such miRNA is miR-218, which is debated as a critical candidate for increased stress susceptibility. miR-218 is expressed throughout the brain, notably in the hippocampus and prefrontal cortex (PFC). It is expressed at various levels through life stages, as seen by adolescent and adult animal models. Until now, a minimal number of studies have been conducted on human subjects to understand its role in stress-related abnormalities in brain circuits. However, several studies, including animal and cell-culture models, have been used to understand the impact of miR-218 on stress response and hypothalamic-pituitary-adrenal (HPA) axis function. So far, expression changes in this miRNA have been found to regulate signaling pathways such as glucocorticoid signaling, serotonergic signaling, and glutamatergic signaling. Recently, the developmental role of miR-218 has generated interest, given its increasing expression from adolescence to adulthood and targeting the Netrin-1/DCC signaling pathway. Since miR-218 expression affects neuronal development and plasticity, it is expected that a change in miR-218 expression levels over the course of development may negatively impact the process and make individuals stress-susceptible in adulthood. In this review, we describe the role of miR-218 in stress-induced neuropsychiatric conditions with an emphasis on stress-related disorders.
Collapse
|
38
|
Differential expression of serum extracellular vesicle microRNAs and analysis of target-gene pathways in major depressive disorder. Biomark Neuropsychiatry 2022. [DOI: 10.1016/j.bionps.2022.100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
39
|
Transsynaptic cerebellin 4-neogenin 1 signaling mediates LTP in the mouse dentate gyrus. Proc Natl Acad Sci U S A 2022; 119:e2123421119. [PMID: 35544694 DOI: 10.1073/pnas.2123421119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SignificanceSynapses are controlled by transsynaptic adhesion complexes that mediate bidirectional signaling between pre- and postsynaptic compartments. Long-term potentiation (LTP) of synaptic transmission is thought to enable synaptic modifications during memory formation, but the signaling mechanisms involved remain poorly understood. We show that binding of cerebellin-4 (Cbln4), a secreted ligand of presynaptic neurexin adhesion molecules, to neogenin-1, a postsynaptic surface protein known as a developmental netrin receptor, is essential for normal LTP at entorhinal cortex→dentate gyrus synapses in mice. Cbln4 and neogenin-1 are dispensable for basal synaptic transmission and not involved in establishing synaptic connections as such. Our data identify a netrin receptor as a postsynaptic organizer of synaptic plasticity that collaborates specifically with the presynaptic neurexin-ligand Cbln4.
Collapse
|
40
|
Talarico F, Costa GO, Ota VK, Santoro ML, Noto C, Gadelha A, Bressan R, Azevedo H, Belangero SI. Systems-Level Analysis of Genetic Variants Reveals Functional and Spatiotemporal Context in Treatment-resistant Schizophrenia. Mol Neurobiol 2022; 59:3170-3182. [DOI: 10.1007/s12035-022-02794-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/06/2022] [Indexed: 10/18/2022]
|
41
|
Lo PS, Rymar VV, Kennedy TE, Sadikot AF. The Netrin-1 Receptor DCC Promotes the Survival of a Subpopulation of Midbrain Dopaminergic Neurons: Relevance for Ageing and Parkinson's Disease. J Neurochem 2022; 161:254-265. [PMID: 35118677 PMCID: PMC9305203 DOI: 10.1111/jnc.15579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022]
Abstract
Mechanisms that determine the survival of midbrain dopaminergic (mDA) neurons in the adult central nervous system (CNS) are not fully understood. Netrins are a family of secreted proteins that are essential for normal neural development. In the mature CNS, mDA neurons express particularly high levels of netrin‐1 and its receptor Deleted in Colorectal Cancer (DCC). Recent findings indicate that overexpressing netrin‐1 protects mDA neurons in animal models of Parkinson’s disease (PD), with a proposed pro‐apoptotic dependence function for DCC that triggers cell death in the absence of a ligand. Here, we sought to determine if DCC expression influences mDA neuron survival in young adult and ageing mice. To circumvent the perinatal lethality of DCC null mice, we selectively deleted DCC from mDA neurons utilizing DATcre/loxP gene‐targeting and examined neuronal survival in adult and aged animals. Reduced numbers of mDA neurons were detected in the substantia nigra pars compacta (SNc) of young adult DATcre/DCCfl/fl mice, with further reduction in aged DATcre/DCCfl/fl animals. In contrast to young adults, aged mice also exhibited a gene dosage effect, with fewer SNc mDA neurons in DCC heterozygotes (DATcre/DCCfl/wt). Notably, loss of mDA neurons in the SN was not uniform. Neuronal loss in the SN was limited to ventral tier mDA neurons, while mDA neurons in the dorsal tier of the SN, which resist degeneration in PD, were spared from the effect of DCC deletion in both young and aged mice. In the ventral tegmental area (VTA), young adult mice with conditional deletion of DCC had normal mDA neuronal numbers, while significant loss occurred in aged DATcre/DCCfl/fl and DATcre/DCCfl/wt mice compared to age‐matched wild‐type mice. Our results indicate that expression of DCC is required for the survival of subpopulations of mDA neurons and may be relevant to the degenerative processes in PD.![]()
Collapse
Affiliation(s)
- Pik-Shan Lo
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada
| | - Abbas F Sadikot
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Iqbal M, Cox SML, Jaworska N, Tippler M, Castellanos-Ryan N, Parent S, Dagher A, Vitaro F, Brendgen MR, Boivin M, Pihl RO, Côté SM, Tremblay RE, Séguin JR, Leyton M. A three-factor model of common early onset psychiatric disorders: temperament, adversity, and dopamine. Neuropsychopharmacology 2022; 47:752-758. [PMID: 34625707 PMCID: PMC8783001 DOI: 10.1038/s41386-021-01187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/03/2023]
Abstract
Commonly comorbid early onset psychiatric disorders might reflect the varying expression of overlapping risk factors. The mediating processes remain poorly understood, but three factors show some promise: adolescent externalizing traits, early life adversity, and midbrain dopamine autoreceptors. To investigate whether these features acquire greater predictive power when combined, a longitudinal study was conducted in youth who have been followed since birth. Cohort members were invited to participate based on externalizing scores between 11 to 16 years of age. At age 18 (age 18.5 ± 0.6 y.o.), 52 entry criteria meeting volunteers had a 90-min positron emission tomography scan with [18F]fallypride, completed the Childhood Trauma Questionnaire, and were assessed with the Structured Clinical Interview for DSM-5. The three-factor model identified those with a lifetime history of DSM-5 disorders with an overall accuracy of 90.4% (p = 2.4 × 10-5) and explained 91.5% of the area under the receiver operating characteristic curve [95% CI: .824, 1.000]. Targeting externalizing disorders specifically did not yield a more powerful model than targeting all disorders (p = 0.54). The model remained significant when including data from participants who developed their first disorders during a three-year follow-up period (p = 3.5 × 10-5). Together, these results raise the possibility that a combination of temperamental traits, childhood adversity, and poorly regulated dopamine transmission increases risk for diverse, commonly comorbid, early onset psychiatric problems, predicting this susceptibility prospectively.
Collapse
Affiliation(s)
- Maisha Iqbal
- grid.416102.00000 0004 0646 3639Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | | | - Natalia Jaworska
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON Canada ,grid.28046.380000 0001 2182 2255University of Ottawa Institute of Mental Health Research, Ottawa, ON Canada
| | - Maria Tippler
- grid.416102.00000 0004 0646 3639Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Natalie Castellanos-Ryan
- grid.14848.310000 0001 2292 3357School of Psychoeducation, Université de Montréal, Montreal, QC Canada
| | - Sophie Parent
- grid.14848.310000 0001 2292 3357School of Psychoeducation, Université de Montréal, Montreal, QC Canada
| | - Alain Dagher
- grid.416102.00000 0004 0646 3639Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Frank Vitaro
- grid.14848.310000 0001 2292 3357School of Psychoeducation, Université de Montréal, Montreal, QC Canada ,grid.411418.90000 0001 2173 6322CHU Ste-Justine Research Center, Montreal, QC Canada
| | - Mara R. Brendgen
- grid.411418.90000 0001 2173 6322CHU Ste-Justine Research Center, Montreal, QC Canada ,grid.38678.320000 0001 2181 0211Department of Psychology, Université de Québec à Montréal, Montreal, QC Canada
| | - Michel Boivin
- grid.23856.3a0000 0004 1936 8390Department of Psychology, Université Laval, Quebec, ON Canada ,grid.77602.340000 0001 1088 3909Institute of Genetic, Neurobiological and Social Foundations of Child Development, Tomsk State University, Siberia, Russia
| | - Robert O. Pihl
- grid.77602.340000 0001 1088 3909Institute of Genetic, Neurobiological and Social Foundations of Child Development, Tomsk State University, Siberia, Russia
| | - Sylvana M. Côté
- grid.411418.90000 0001 2173 6322CHU Ste-Justine Research Center, Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Department of Social & Preventative Medicine, Université de Montréal, Montreal, QC Canada
| | - Richard E. Tremblay
- grid.411418.90000 0001 2173 6322CHU Ste-Justine Research Center, Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Departments of Pediatrics & Psychology, Université de Montréal, Montreal, QC Canada ,grid.7886.10000 0001 0768 2743School of Public Health and Sports Science, University College Dublin, Dublin, Ireland ,grid.7429.80000000121866389INSERM, U669 Paris, France
| | - Jean R. Séguin
- grid.411418.90000 0001 2173 6322CHU Ste-Justine Research Center, Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada
| | - Marco Leyton
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada. .,Department of Psychiatry, McGill University, Montreal, QC, Canada. .,CHU Ste-Justine Research Center, Montreal, QC, Canada. .,Department of Psychology, McGill University, Montreal, QC, Canada. .,Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
43
|
Restrepo-Lozano JM, Pokhvisneva I, Wang Z, Patel S, Meaney MJ, Silveira PP, Flores C. Corticolimbic DCC gene co-expression networks as predictors of impulsivity in children. Mol Psychiatry 2022; 27:2742-2750. [PMID: 35388180 PMCID: PMC9156406 DOI: 10.1038/s41380-022-01533-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 12/16/2022]
Abstract
Inhibitory control deficits are prevalent in multiple neuropsychiatric conditions. The communication- as well as the connectivity- between corticolimbic regions of the brain are fundamental for eliciting inhibitory control behaviors, but early markers of vulnerability to this behavioral trait are yet to be discovered. The gradual maturation of the prefrontal cortex (PFC), in particular of the mesocortical dopamine innervation, mirrors the protracted development of inhibitory control; both are present early in life, but reach full maturation by early adulthood. Evidence suggests the involvement of the Netrin-1/DCC signaling pathway and its associated gene networks in corticolimbic development. Here we investigated whether an expression-based polygenic score (ePRS) based on corticolimbic-specific DCC gene co-expression networks associates with impulsivity-related phenotypes in community samples of children. We found that lower ePRS scores associate with higher measurements of impulsive choice in 6-year-old children tested in the Information Sampling Task and with impulsive action in 6- and 10-year-old children tested in the Stop Signal Task. We also found the ePRS to be a better overall predictor of impulsivity when compared to a conventional PRS score comparable in size to the ePRS (4515 SNPs in our discovery cohort) and derived from the latest GWAS for ADHD. We propose that the corticolimbic DCC-ePRS can serve as a novel type of marker for impulsivity-related phenotypes in children. By adopting a systems biology approach based on gene co-expression networks and genotype-gene expression (rather than genotype-disease) associations, these results further validate our methodology to construct polygenic scores linked to the overall biological function of tissue-specific gene networks.
Collapse
Affiliation(s)
- Jose M. Restrepo-Lozano
- grid.14709.3b0000 0004 1936 8649Integrated Program in Neuroscience, McGill University, Montreal, QC Canada ,grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada
| | - Irina Pokhvisneva
- grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, QC Canada
| | - Zihan Wang
- grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, QC Canada
| | - Sachin Patel
- grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, QC Canada
| | - Michael J. Meaney
- grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC Canada ,grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Patricia P. Silveira
- grid.412078.80000 0001 2353 5268Douglas Mental Health University Institute, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montreal, QC, Canada. .,Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada. .,Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
44
|
Luciana M, Collins PF. Is Adolescence a Sensitive Period for the Development of Incentive-Reward Motivation? Curr Top Behav Neurosci 2021; 53:79-99. [PMID: 34784026 DOI: 10.1007/7854_2021_275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Human adolescence is broadly construed as a time of heightened risk-taking and a vulnerability period for the emergence of psychopathology. These tendencies have been attributed to the age-related development of neural systems that mediate incentive motivation and other aspects of reward processing as well as individual difference factors that interact with ongoing development. Here, we describe the adolescent development of incentive motivation, which we view as an inherently positive developmental progression, and its associated neural mechanisms. We consider challenges in applying the sensitive period concept to these maturational events and discuss future directions that may help to clarify mechanisms of change.
Collapse
Affiliation(s)
- Monica Luciana
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA.
| | - Paul F Collins
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
45
|
Islam KUS, Meli N, Blaess S. The Development of the Mesoprefrontal Dopaminergic System in Health and Disease. Front Neural Circuits 2021; 15:746582. [PMID: 34712123 PMCID: PMC8546303 DOI: 10.3389/fncir.2021.746582] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Midbrain dopaminergic neurons located in the substantia nigra and the ventral tegmental area are the main source of dopamine in the brain. They send out projections to a variety of forebrain structures, including dorsal striatum, nucleus accumbens, and prefrontal cortex (PFC), establishing the nigrostriatal, mesolimbic, and mesoprefrontal pathways, respectively. The dopaminergic input to the PFC is essential for the performance of higher cognitive functions such as working memory, attention, planning, and decision making. The gradual maturation of these cognitive skills during postnatal development correlates with the maturation of PFC local circuits, which undergo a lengthy functional remodeling process during the neonatal and adolescence stage. During this period, the mesoprefrontal dopaminergic innervation also matures: the fibers are rather sparse at prenatal stages and slowly increase in density during postnatal development to finally reach a stable pattern in early adulthood. Despite the prominent role of dopamine in the regulation of PFC function, relatively little is known about how the dopaminergic innervation is established in the PFC, whether and how it influences the maturation of local circuits and how exactly it facilitates cognitive functions in the PFC. In this review, we provide an overview of the development of the mesoprefrontal dopaminergic system in rodents and primates and discuss the role of altered dopaminergic signaling in neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- K Ushna S Islam
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Norisa Meli
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
46
|
Reynolds LM, Flores C. Mesocorticolimbic Dopamine Pathways Across Adolescence: Diversity in Development. Front Neural Circuits 2021; 15:735625. [PMID: 34566584 PMCID: PMC8456011 DOI: 10.3389/fncir.2021.735625] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
Mesocorticolimbic dopamine circuity undergoes a protracted maturation during adolescent life. Stable adult levels of behavioral functioning in reward, motivational, and cognitive domains are established as these pathways are refined, however, their extended developmental window also leaves them vulnerable to perturbation by environmental factors. In this review, we highlight recent advances in understanding the mechanisms underlying dopamine pathway development in the adolescent brain, and how the environment influences these processes to establish or disrupt neurocircuit diversity. We further integrate these recent studies into the larger historical framework of anatomical and neurochemical changes occurring during adolescence in the mesocorticolimbic dopamine system. While dopamine neuron heterogeneity is increasingly appreciated at molecular, physiological, and anatomical levels, we suggest that a developmental facet may play a key role in establishing vulnerability or resilience to environmental stimuli and experience in distinct dopamine circuits, shifting the balance between healthy brain development and susceptibility to psychiatric disease.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Plasticité du Cerveau CNRS UMR8249, École supérieure de physique et de chimie industrielles de la Ville de Paris (ESPCI Paris), Paris, France.,Neuroscience Paris Seine CNRS UMR 8246 INSERM U1130, Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|
47
|
Peters KZ, Zlebnik NE, Cheer JF. Cannabis exposure during adolescence: A uniquely sensitive period for neurobiological effects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:95-120. [PMID: 34801175 DOI: 10.1016/bs.irn.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is a crucial developmental period where neural circuits are refined and the brain is especially vulnerable to external insults. The endocannabinoid (eCB) system undergoes changes during adolescence which affect the way in which it modulates the development of other systems, in particular dopamine circuits, which show protracted development into adolescence. Given the rise of cannabis use by adolescents and young people, as well as variants containing increasingly higher concentrations of THC, it is now crucial to understand the unique effects of adolescent exposure to cannabis on the developing brain and it might shape future adult vulnerabilities to conditions such as psychosis, schizophrenia, addiction and more. Here we discuss the development of the eCB system across the lifespan, how CB1 receptors modulate dopamine release and potential neurobiological and behavioral effects of adolescent THC exposure on the developing brain such as alterations in excitatory/inhibitory balance during this developmental period.
Collapse
Affiliation(s)
- K Z Peters
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom.
| | - N E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - J F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
48
|
Cheng A, Jia W, Kawahata I, Fukunaga K. Impact of Fatty Acid-Binding Proteins in α-Synuclein-Induced Mitochondrial Injury in Synucleinopathy. Biomedicines 2021; 9:biomedicines9050560. [PMID: 34067791 PMCID: PMC8156290 DOI: 10.3390/biomedicines9050560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Synucleinopathies are diverse diseases with motor and cognitive dysfunction due to progressive neuronal loss or demyelination, due to oligodendrocyte loss in the brain. While the etiology of neurodegenerative disorders (NDDs) is likely multifactorial, mitochondrial injury is one of the most vital factors in neuronal loss and oligodendrocyte dysfunction, especially in Parkinson’s disease, dementia with Lewy body, multiple system atrophy, and Krabbe disease. In recent years, the abnormal accumulation of highly neurotoxic α-synuclein in the mitochondrial membrane, which leads to mitochondrial dysfunction, was well studied. Furthermore, fatty acid-binding proteins (FABPs), which are members of a superfamily and are essential in fatty acid trafficking, were reported to trigger α-synuclein oligomerization in neurons and glial cells and to target the mitochondrial outer membrane, thereby causing mitochondrial loss. Here, we provide an updated overview of recent findings on FABP and α-synuclein interactions and mitochondrial injury in NDDs.
Collapse
Affiliation(s)
- An Cheng
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Wenbin Jia
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Ichiro Kawahata
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Kohji Fukunaga
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
- Correspondence: ; Tel.: +81-(22)-795-6837
| |
Collapse
|
49
|
Torres-Berrío A, Morgunova A, Giroux M, Cuesta S, Nestler EJ, Flores C. miR-218 in Adolescence Predicts and Mediates Vulnerability to Stress. Biol Psychiatry 2021; 89:911-919. [PMID: 33384174 PMCID: PMC8052258 DOI: 10.1016/j.biopsych.2020.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/30/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Adolescence is a period of increased vulnerability to psychiatric disorders, including depression. Discovering novel biomarkers to identify individuals who are at high risk is very much needed. Our previous work shows that the microRNA miR-218 mediates susceptibility to stress and depression in adulthood by targeting the netrin-1 guidance cue receptor gene Dcc in the medial prefrontal cortex (mPFC). METHODS Here, we investigated whether miR-218 regulates Dcc expression in adolescence and could serve as an early predictor of lifetime stress vulnerability in male mice. RESULTS miR-218 expression in the mPFC increases from early adolescence to adulthood and correlates negatively with Dcc levels. In blood, postnatal miR-218 expression parallels changes occurring in the mPFC. Notably, circulating miR-218 levels in adolescence associate with vulnerability to social defeat stress in adulthood, with high levels associated with social avoidance severity. Indeed, downregulation of miR-218 in the mPFC in adolescence promotes resilience to stress in adulthood. CONCLUSIONS miR-218 expression in adolescence may serve both as a marker of risk and as a target for early interventions.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice Morgunova
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada; Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Michel Giroux
- Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Santiago Cuesta
- Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Cecilia Flores
- Department of Psychiatry, McGill University, Montréal, Québec, Canada; Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
50
|
MicroRNA regulation of prefrontal cortex development and psychiatric risk in adolescence. Semin Cell Dev Biol 2021; 118:83-91. [PMID: 33933350 DOI: 10.1016/j.semcdb.2021.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/28/2022]
Abstract
In this review, we examine the role of microRNAs in the development of the prefrontal cortex (PFC) in adolescence and in individual differences in vulnerability to mental illness. We describe results from clinical and preclinical research indicating that adolescence coincides with drastic changes in local microRNA expression, including microRNAs that control gene networks involved in PFC and cognitive refinement. We highlight that altered levels of microRNAs in the PFC are associated with psychopathologies of adolescent onset, notably depression and schizophrenia. We show that microRNAs can be measured non-invasively in peripheral samples and could serve as longitudinal physiological readouts of brain expression and psychiatric risk in youth.
Collapse
|