1
|
Altas B, Tuffy LP, Patrizi A, Dimova K, Soykan T, Brandenburg C, Romanowski AJ, Whitten JR, Robertson CD, Khim SN, Crutcher GW, Ambrozkiewicz MC, Yagensky O, Krueger-Burg D, Hammer M, Hsiao HH, Laskowski PR, Dyck L, Puche AC, Sassoè-Pognetto M, Chua JJE, Urlaub H, Jahn O, Brose N, Poulopoulos A. Region-Specific Phosphorylation Determines Neuroligin-3 Localization to Excitatory Versus Inhibitory Synapses. Biol Psychiatry 2024; 96:815-828. [PMID: 38154503 PMCID: PMC11209832 DOI: 10.1016/j.biopsych.2023.12.020] [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: 09/29/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Neuroligin-3 is a postsynaptic adhesion molecule involved in synapse development and function. It is implicated in rare, monogenic forms of autism, and its shedding is critical to the tumor microenvironment of gliomas. While other members of the neuroligin family exhibit synapse-type specificity in localization and function through distinct interactions with postsynaptic scaffold proteins, the specificity of neuroligin-3 synaptic localization remains largely unknown. METHODS We investigated the synaptic localization of neuroligin-3 across regions in mouse and human brain samples after validating antibody specificity in knockout animals. We raised a phospho-specific neuroligin antibody and used phosphoproteomics, cell-based assays, and in utero CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9) knockout and gene replacement to identify mechanisms that regulate neuroligin-3 localization to distinct synapse types. RESULTS Neuroligin-3 exhibits region-dependent synapse specificity, largely localizing to excitatory synapses in cortical regions and inhibitory synapses in subcortical regions of the brain in both mice and humans. We identified specific phosphorylation of cortical neuroligin-3 at a key binding site for recruitment to inhibitory synapses, while subcortical neuroligin-3 remained unphosphorylated. In vitro, phosphomimetic mutation of that site disrupted neuroligin-3 association with the inhibitory postsynaptic scaffolding protein gephyrin. In vivo, phosphomimetic mutants of neuroligin-3 localized to excitatory postsynapses, while phospho-null mutants localized to inhibitory postsynapses. CONCLUSIONS These data reveal an unexpected region-specific pattern of neuroligin-3 synapse specificity, as well as a phosphorylation-dependent mechanism that regulates its recruitment to either excitatory or inhibitory synapses. These findings add to our understanding of how neuroligin-3 is involved in conditions that may affect the balance of excitation and inhibition.
Collapse
Affiliation(s)
- Bekir Altas
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Liam P Tuffy
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Annarita Patrizi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Kalina Dimova
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tolga Soykan
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Cheryl Brandenburg
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea J Romanowski
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Julia R Whitten
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Colin D Robertson
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Saovleak N Khim
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Garrett W Crutcher
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mateusz C Ambrozkiewicz
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Oleksandr Yagensky
- Research Group Protein Trafficking in Synaptic Development and Function, Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Matthieu Hammer
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - He-Hsuan Hsiao
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Pawel R Laskowski
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lydia Dyck
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Adam C Puche
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - John J E Chua
- Research Group Protein Trafficking in Synaptic Development and Function, Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Olaf Jahn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alexandros Poulopoulos
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
2
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [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: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
3
|
Cheng KH, Hung YC, Ling P, Hsu KS. Oxytocin treatment rescues irritability-like behavior in Cc2d1a conditional knockout mice. Neuropsychopharmacology 2024; 49:1792-1802. [PMID: 39014123 PMCID: PMC11399130 DOI: 10.1038/s41386-024-01920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/19/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Irritability, a state of excessive reactivity to negative emotional stimuli, is common in individuals with autism spectrum disorder (ASD). Although it has a significant negative impact of patients' disease severity and quality of life, the neural mechanisms underlying irritability in ASD remain largely unclear. We have previously demonstrated that male mice lacking the Coiled-coil and C2 domain containing 1a (Cc2d1a) in forebrain excitatory neurons recapitulate numerous ASD-like behavioral phenotypes, including impaired social behaviors and pronounced repetitive behaviors. Here, using the bottle-brush test (BBT) to trigger and evaluate aggressive and defensive responses, we show that Cc2d1a deletion increases irritability-like behavior in male but not female mice, which is correlated with reduced number of oxytocin (OXT)-expressing neurons in the paraventricular nucleus (PVN) of the hypothalamus. Intranasal OXT administration or chemogenetic activation of OXT neurons in the PVN rescues irritability-like behavior in Cc2d1a conditional knockout (cKO) mice. Administration of a selective melanocortin receptor 4 agonist, RO27-3225, which potentiates endogenous OXT release, also alleviates irritability-like behavior in Cc2d1a cKO mice, an effect blocked by a specific OXT receptor antagonist, L-368,899. We additionally identify a projection connecting the posterior ventral segment of the medial amygdala (MeApv) and ventromedial nucleus of the ventromedial hypothalamus (VMHvl) for governing irritability-like behavior during the BBT. Chemogenetic suppression of the MeApv-VMHvl pathway alleviates irritability-like behavior in Cc2d1a cKO mice. Together, our study uncovers dysregulation of OXT system in irritability-like behavior in Cc2d1a cKO mice during the BBT and provide translatable insights into the development of OXT-based therapeutics for clinical interventions.
Collapse
Affiliation(s)
- Kuan-Hsiang Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pin Ling
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
4
|
Wang T, Ye J, Zhang Y, Li J, Yang T, Wang Y, Jiang X, Yao Q. Role of oxytocin in bone. Front Endocrinol (Lausanne) 2024; 15:1450007. [PMID: 39290327 PMCID: PMC11405241 DOI: 10.3389/fendo.2024.1450007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Oxytocin (OT) is a posterior pituitary hormone that, in addition to its role in regulating childbirth and lactation, also exerts direct regulatory effects on the skeleton through peripheral OT and oxytocin receptor (OTR). Bone marrow mesenchymal stem cells (BMSCs), osteoblasts (OB), osteoclasts (OC), chondrocytes, and adipocytes all express OT and OTR. OT upregulates RUNX2, BMP2, ALP, and OCN, thereby enhancing the activity of BMSCs and promoting their differentiation towards OB rather than adipocytes. OT also directly regulates OPG/RANKL to inhibit adipocyte generation, increase the expression of SOX9 and COMP, and enhance chondrocyte differentiation. OB can secrete OT, exerting influence on the surrounding environment through autocrine and paracrine mechanisms. OT directly increases OC formation through the NκB/MAP kinase signaling pathway, inhibits osteoclast proliferation by triggering cytoplasmic Ca2+ release and nitric oxide synthesis, and has a dual regulatory effect on OCs. Under the stimulation of estrogen, OB synthesizes OT, amplifying the biological effects of estrogen and OT. Mediated by estrogen, the OT/OTR forms a feedforward loop with OB. Apart from estrogen, OT also interacts with arginine vasopressin (AVP), prostaglandins (PGE2), leptin, and adiponectin to regulate bone metabolism. This review summarizes recent research on the regulation of bone metabolism by OT and OTR, aiming to provide insights into their clinical applications and further research.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianya Ye
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Orthopedic Surgery, Huaian Hospital of Huaian City, Huaian, China
| | - Yongqiang Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiayi Li
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tianxiao Yang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yufeng Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Jiang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Blazie SM, Fortunati D, Zhao Y, Jin Y. C. elegans LIN-66 mediates EIF-3/eIF3-dependent protein translation via a cold-shock domain. Life Sci Alliance 2024; 7:e202402673. [PMID: 38886018 PMCID: PMC11184513 DOI: 10.26508/lsa.202402673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Protein translation initiation is a conserved process involving many proteins acting in concert. The 13 subunit eukaryotic initiation factor 3 (eIF3) complex is essential for assembly of the pre-initiation complex that scans mRNA and positions ribosome at the initiation codon. We previously reported that a gain-of-function (gf) mutation affecting the G subunit of the Caenorhabditis elegans eIF3 complex, eif-3.g(gf), selectively modulates protein translation in the ventral cord cholinergic motor neurons. Here, through unbiased genetic suppressor screening, we identified that the gene lin-66 mediates eif-3.g(gf)-dependent protein translation in motor neurons. LIN-66 is composed largely of low-complexity amino acid sequences with unknown functional domains. We combined bioinformatics analysis with in vivo functional dissection and identified a cold-shock domain in LIN-66 critical for its function. In cholinergic motor neurons, LIN-66 shows a close association with EIF-3.G in the cytoplasm. The low-complexity amino acid sequences of LIN-66 modulate its subcellular pattern. As cold-shock domains function broadly in RNA regulation, we propose that LIN-66 mediates stimulus-dependent protein translation by facilitating the interaction of mRNAs with EIF-3.G.
Collapse
Affiliation(s)
- Stephen M Blazie
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Daniel Fortunati
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yan Zhao
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Choi TY, Jeong S, Koo JW. Mesocorticolimbic circuit mechanisms of social dominance behavior. Exp Mol Med 2024; 56:1889-1899. [PMID: 39218974 PMCID: PMC11447232 DOI: 10.1038/s12276-024-01299-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/10/2024] [Accepted: 05/23/2024] [Indexed: 09/04/2024] Open
Abstract
Social animals, including rodents, primates, and humans, partake in competition for finite resources, thereby establishing social hierarchies wherein an individual's social standing influences diverse behaviors. Understanding the neurobiological underpinnings of social dominance is imperative, given its ramifications for health, survival, and reproduction. Social dominance behavior comprises several facets, including social recognition, social decision-making, and actions, indicating the concerted involvement of multiple brain regions in orchestrating this behavior. While extensive research has been dedicated to elucidating the neurobiology of social interaction, recent studies have increasingly delved into adverse social behaviors such as social competition and hierarchy. This review focuses on the latest advancements in comprehending the mechanisms of the mesocorticolimbic circuit governing social dominance, with a specific focus on rodent studies, elucidating the intricate dynamics of social hierarchies and their implications for individual well-being and adaptation.
Collapse
Affiliation(s)
- Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.
| | - Sejin Jeong
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Life Sciences, Yeungnam University, Gyeongsan, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
7
|
Wang Y, Xu H, Chen S, Chen J, Zheng Q, Ma Y, Zhao X, Shi Y, Xiao L. Oxytocin Protects Nigrostriatal Dopamine Signal via Activating GABAergic Circuit in the MPTP-Induced Parkinson's Disease Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310244. [PMID: 39099429 PMCID: PMC11423065 DOI: 10.1002/advs.202310244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/23/2024] [Indexed: 08/06/2024]
Abstract
The most pronounced neuropathological feature of Parkinson's disease (PD) is the loss of dopamine (DA) neurons in the substantia nigra compacta (SNc), which depletes striatal DA. Hypothalamic oxytocin is found to be reduced in PD patients and closely interacts with the DA system, but the role of oxytocin in PD remains unclear. Here, the disturbances of endogenous oxytocin level and the substantia nigra (SN) oxytocin receptor expression in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model is observed, correlated with the striatal tyrosine hydroxylase (TH) expression reduction. Killing/silencing hypothalamic oxytocin neurons aggravates the vulnerability of nigrostriatal DA signal to MPTP, whereas elevating oxytocin level by intranasal delivery or microinjecting into the SN promotes the resistance. In addition, knocking out SN oxytocin receptors induces the time-dependent reductions of SNc DA neurons, striatal TH expression, and striatal DA level by increasing neuronal excitotoxicity. These results further uncover that oxytocin dampens the excitatory synaptic inputs onto DA neurons via activating oxytocin receptor-expressed SN GABA neurons, which target GABA(B) receptors expressed in SNc DA neuron-projecting glutamatergic axons, to reduce excitotoxicity. Thus, besides the well-known prosocial effect, oxytocin acts as a key endogenous factor in protecting the nigrostriatal DA system.
Collapse
Affiliation(s)
- Yurong Wang
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hao Xu
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Saiyong Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Junhao Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qimeng Zheng
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xinru Zhao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying Shi
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
8
|
Brandner DD, Mashal MA, Grissom NM, Rothwell PE. Sex differences in morphine sensitivity of neuroligin-3 knockout mice. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06660-3. [PMID: 39083079 DOI: 10.1007/s00213-024-06660-3] [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: 06/02/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Sex has a strong influence on the prevalence and course of brain conditions, including autism spectrum disorders. The mechanistic basis for these sex differences remains poorly understood, due in part to historical bias in biomedical research favoring analysis of male subjects, and the exclusion of female subjects. For example, studies of male mice carrying autism-associated mutations in neuroligin-3 are over-represented in the literature, including our own prior work showing diminished responses to chronic morphine exposure in male neuroligin-3 knockout mice. We therefore studied how constitutive and conditional genetic knockout of neuroligin-3 affects morphine sensitivity of female mice, using locomotor activity as a proxy for differences in opioid sensitivity that may be related to the pathophysiology and treatment of autism spectrum disorders. In contrast to male mice, female neuroligin-3 knockout mice showed normal psychomotor sensitization after chronic morphine exposure. However, in the absence of neuroligin-3 expression, both female and male mice show a similar change in the topography of locomotor stimulation produced by morphine. Conditional genetic deletion of neuroligin-3 from dopamine neurons increased the locomotor response of female mice to high doses of morphine, contrasting with the decrease in psychomotor sensitization caused by the same manipulation in male mice. Together, our data reveal that knockout of neuroligin-3 has both common and distinct effects on morphine sensitivity in female and male mice. These results also support the notion that female sex can confer resilience against the impact of autism-associated gene variants.
Collapse
Affiliation(s)
- Dieter D Brandner
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
| | - Mohammed A Mashal
- Department of Neuroscience, University of Minnesota, 4-142 Wallin Medical Biosciences Building, 2101 6 Street SE, Minneapolis, MN, 55455, USA
| | - Nicola M Grissom
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Patrick E Rothwell
- Department of Neuroscience, University of Minnesota, 4-142 Wallin Medical Biosciences Building, 2101 6 Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
9
|
Kojima L, Seiriki K, Rokujo H, Nakazawa T, Kasai A, Hashimoto H. Optimization of AAV vectors for transactivator-regulated enhanced gene expression within targeted neuronal populations. iScience 2024; 27:109878. [PMID: 38799556 PMCID: PMC11126825 DOI: 10.1016/j.isci.2024.109878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/03/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Adeno-associated virus (AAV) vectors are potential tools for cell-type-selective gene delivery to the central nervous system. Although cell-type-specific enhancers and promoters have been identified for AAV systems, there is limited information regarding the effects of AAV genomic components on the selectivity and efficiency of gene expression. Here, we offer an alternative strategy to provide specific and efficient gene delivery to a targeted neuronal population by optimizing recombinant AAV genomic components, named TAREGET (TransActivator-Regulated Enhanced Gene Expression within Targeted neuronal populations). We established this strategy in oxytocinergic neurons and showed that the TAREGET enabled sufficient gene expression to label long-projecting axons in wild-type mice. Its application to other cell types, including serotonergic and dopaminergic neurons, was also demonstrated. These results demonstrate that optimization of AAV expression cassettes can improve the specificity and efficiency of cell-type-specific gene expression and that TAREGET can renew previously established cell-type-specific promoters with improved performance.
Collapse
Affiliation(s)
- Leo Kojima
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Rokujo
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takanobu Nakazawa
- Department of Bioscience, Tokyo University of Agriculture, Setagaya-ku, Tokyo 156-8502, Japan
| | - Atsushi Kasai
- Systems Neuropharmacology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
- Institute for Datability Science, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
10
|
Brandner DD, Mashal MA, Grissom NM, Rothwell PE. Sex Differences in Morphine Sensitivity of Neuroligin-3 Knockout Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596965. [PMID: 38854153 PMCID: PMC11160712 DOI: 10.1101/2024.06.01.596965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Sex has a strong influence on the prevalence and course of brain conditions, including autism spectrum disorders. The mechanistic basis for these sex differences remains poorly understood, due in part to historical bias in biomedical research favoring analysis of male subjects, and the exclusion of female subjects. For example, studies of male mice carrying autism-associated mutations in neuroligin-3 are over-represented in the literature, including our own prior work showing diminished responses to chronic morphine exposure in male neuroligin-3 knockout mice. We therefore studied how constitutive and conditional genetic knockout of neuroligin-3 affects morphine sensitivity of female mice. In contrast to male mice, female neuroligin-3 knockout mice showed normal psychomotor sensitization after chronic morphine exposure. However, in the absence of neuroligin-3 expression, both female and male mice show a similar change in the topography of locomotor stimulation produced by morphine. Conditional genetic deletion of neuroligin-3 from dopamine neurons increased the locomotor response of female mice to high doses of morphine, contrasting with the decrease in psychomotor sensitization caused by the same manipulation in male mice. Together, our data reveal that knockout of neuroligin-3 has both common and distinct effects on morphine sensitivity in female and male mice. These results also support the notion that female sex can confer resilience against the impact of autism-associated gene variants.
Collapse
|
11
|
Yin H, Jiang M, Han T, Xu X. Intranasal oxytocin as a treatment for anxiety and autism: From subclinical to clinical applications. Peptides 2024; 176:171211. [PMID: 38579916 DOI: 10.1016/j.peptides.2024.171211] [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: 02/02/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Animal and human studies have demonstrated that intranasal oxytocin (OT) can penetrate the brain and induce cognitive, emotional, and behavioral changes, particularly in social functioning. Consequently, numerous investigations have explored the potential of OT as a treatment for anxiety and autism, conditions characterized by social deficits. Although both subclinical and clinical studies provide converging evidence of the therapeutic effects of OT in reducing anxiety levels and improving social symptoms in autism, results are not always consistent. Additionally, the pharmacological mechanism of OT requires further elucidation for its effective clinical application. Therefore, this review aims to examine the contentious findings concerning the effects of OT on anxiety and autism, offer interpretations of the inconsistent results from the perspectives of individual differences and varying approaches to OT administration, and shed light on the underlying mechanisms of OT. Ultimately, standardization of dosage, frequency of administration, formulation characteristics, and nasal spray devices is proposed as essential for future human studies and clinical applications of OT treatment.
Collapse
Affiliation(s)
- Hailian Yin
- School of psychology, Shandong Normal University, Jinan 250014, China
| | - Meiyun Jiang
- School of psychology, Shandong Normal University, Jinan 250014, China
| | - Tao Han
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan 250000, China.
| | - Xiaolei Xu
- School of psychology, Shandong Normal University, Jinan 250014, China.
| |
Collapse
|
12
|
Wu S, Wang J, Zhang Z, Jin X, Xu Y, Si Y, Liang Y, Ge Y, Zhan H, Peng L, Bi W, Luo D, Li M, Meng B, Guan Q, Zhao J, Gao L, He Z. Shank3 deficiency elicits autistic-like behaviors by activating p38α in hypothalamic AgRP neurons. Mol Autism 2024; 15:14. [PMID: 38570876 PMCID: PMC10993499 DOI: 10.1186/s13229-024-00595-4] [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: 01/30/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND SH3 and multiple ankyrin repeat domains protein 3 (SHANK3) monogenic mutations or deficiency leads to excessive stereotypic behavior and impaired sociability, which frequently occur in autism cases. To date, the underlying mechanisms by which Shank3 mutation or deletion causes autism and the part of the brain in which Shank3 mutation leads to the autistic phenotypes are understudied. The hypothalamus is associated with stereotypic behavior and sociability. p38α, a mediator of inflammatory responses in the brain, has been postulated as a potential gene for certain cases of autism occurrence. However, it is unclear whether hypothalamus and p38α are involved in the development of autism caused by Shank3 mutations or deficiency. METHODS Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and immunoblotting were used to assess alternated signaling pathways in the hypothalamus of Shank3 knockout (Shank3-/-) mice. Home-Cage real-time monitoring test was performed to record stereotypic behavior and three-chamber test was used to monitor the sociability of mice. Adeno-associated viruses 9 (AAV9) were used to express p38α in the arcuate nucleus (ARC) or agouti-related peptide (AgRP) neurons. D176A and F327S mutations expressed constitutively active p38α. T180A and Y182F mutations expressed inactive p38α. RESULTS We found that Shank3 controls stereotypic behavior and sociability by regulating p38α activity in AgRP neurons. Phosphorylated p38 level in hypothalamus is significantly enhanced in Shank3-/- mice. Consistently, overexpression of p38α in ARC or AgRP neurons elicits excessive stereotypic behavior and impairs sociability in wild-type (WT) mice. Notably, activated p38α in AgRP neurons increases stereotypic behavior and impairs sociability. Conversely, inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. In contrast, activated p38α in pro-opiomelanocortin (POMC) neurons does not affect stereotypic behavior and sociability in mice. LIMITATIONS We demonstrated that SHANK3 regulates the phosphorylated p38 level in the hypothalamus and inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. However, we did not clarify the biochemical mechanism of SHANK3 inhibiting p38α in AgRP neurons. CONCLUSIONS These results demonstrate that the Shank3 deficiency caused autistic-like behaviors by activating p38α signaling in AgRP neurons, suggesting that p38α signaling in AgRP neurons is a potential therapeutic target for Shank3 mutant-related autism.
Collapse
Affiliation(s)
- Shanshan Wu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Jing Wang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Zicheng Zhang
- School of Modern Posts, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu, 210009, China
| | - Xinchen Jin
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yang Xu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Youwen Si
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences,East China Normal University, Shanghai, 200062, China
| | - Yixiao Liang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Yueping Ge
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Huidong Zhan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Li Peng
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Dandan Luo
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Mengzhu Li
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bo Meng
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences,East China Normal University, Shanghai, 200062, China
- Department of Pharmacology and Chemical Biology, Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, 544 Jingsi Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
13
|
Li LY, Imai A, Izumi H, Inoue R, Koshidaka Y, Takao K, Mori H, Yoshida T. Differential contribution of canonical and noncanonical NLGN3 pathways to early social development and memory performance. Mol Brain 2024; 17:16. [PMID: 38475840 PMCID: PMC10935922 DOI: 10.1186/s13041-024-01087-5] [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: 11/15/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Neuroligin (NLGN) 3 is a postsynaptic cell adhesion protein organizing synapse formation through two different types of transsynaptic interactions, canonical interaction with neurexins (NRXNs) and a recently identified noncanonical interaction with protein tyrosine phosphatase (PTP) δ. Although, NLGN3 gene is known as a risk gene for neurodevelopmental disorders such as autism spectrum disorder (ASD) and intellectual disability (ID), the pathogenic contribution of the canonical NLGN3-NRXN and noncanonical NLGN3-PTPδ pathways to these disorders remains elusive. In this study, we utilized Nlgn3 mutant mice selectively lacking the interaction with either NRXNs or PTPδ and investigated their social and memory performance. Neither Nlgn3 mutants showed any social cognitive deficiency in the social novelty recognition test. However, the Nlgn3 mutant mice lacking the PTPδ pathway exhibited significant decline in the social conditioned place preference (sCPP) at the juvenile stage, suggesting the involvement of the NLGN3-PTPδ pathway in the regulation of social motivation and reward. In terms of learning and memory, disrupting the canonical NRXN pathway attenuated contextual fear conditioning while disrupting the noncanonical NLGN3-PTPδ pathway enhanced it. Furthermore, disruption of the NLGN3-PTPδ pathway negatively affected the remote spatial reference memory in the Barnes maze test. These findings highlight the differential contributions of the canonical NLGN3-NRXN and noncanonical NLGN3-PTPδ synaptogenic pathways to the regulation of higher order brain functions associated with ASD and ID.
Collapse
Affiliation(s)
- Lin-Yu Li
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Ayako Imai
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Hironori Izumi
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Ran Inoue
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Yumie Koshidaka
- Division of Experimental Animal Resource and Development, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Keizo Takao
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
- Division of Experimental Animal Resource and Development, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan.
- Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
14
|
Matsushima T, Izumi T, Vallortigara G. The domestic chick as an animal model of autism spectrum disorder: building adaptive social perceptions through prenatally formed predispositions. Front Neurosci 2024; 18:1279947. [PMID: 38356650 PMCID: PMC10864568 DOI: 10.3389/fnins.2024.1279947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Equipped with an early social predisposition immediately post-birth, humans typically form associations with mothers and other family members through exposure learning, canalized by a prenatally formed predisposition of visual preference to biological motion, face configuration, and other cues of animacy. If impaired, reduced preferences can lead to social interaction impairments such as autism spectrum disorder (ASD) via misguided canalization. Despite being taxonomically distant, domestic chicks could also follow a homologous developmental trajectory toward adaptive socialization through imprinting, which is guided via predisposed preferences similar to those of humans, thereby suggesting that chicks are a valid animal model of ASD. In addition to the phenotypic similarities in predisposition with human newborns, accumulating evidence on the responsible molecular mechanisms suggests the construct validity of the chick model. Considering the recent progress in the evo-devo studies in vertebrates, we reviewed the advantages and limitations of the chick model of developmental mental diseases in humans.
Collapse
Affiliation(s)
- Toshiya Matsushima
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo, Japan
- Faculty of Pharmaceutical Science, Health Science University of Hokkaido, Tobetsu, Japan
- Centre for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Takeshi Izumi
- Faculty of Pharmaceutical Science, Health Science University of Hokkaido, Tobetsu, Japan
| | | |
Collapse
|
15
|
Pizzarelli R, Pimpinella D, Jacobs C, Tartacca A, Kullolli U, Monyer H, Alberini CM, Griguoli M. Insulin-like growth factor 2 (IGF-2) rescues social deficits in NLG3 -/y mouse model of ASDs. Front Cell Neurosci 2024; 17:1332179. [PMID: 38298376 PMCID: PMC10827848 DOI: 10.3389/fncel.2023.1332179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024] Open
Abstract
Autism spectrum disorders (ASDs) comprise developmental disabilities characterized by impairments of social interaction and repetitive behavior, often associated with cognitive deficits. There is no current treatment that can ameliorate most of the ASDs symptomatology; thus, identifying novel therapies is urgently needed. Here, we used the Neuroligin 3 knockout mouse (NLG3-/y), a model that recapitulates the social deficits reported in ASDs patients, to test the effects of systemic administration of IGF-2, a polypeptide that crosses the blood-brain barrier and acts as a cognitive enhancer. We show that systemic IGF-2 treatment reverses the typical defects in social interaction and social novelty discrimination reflective of ASDs-like phenotypes. This effect was not accompanied by any change in spontaneous glutamatergic synaptic transmission in CA2 hippocampal region, a mechanism found to be crucial for social novelty discrimination. However, in both NLG3+/y and NLG3-/y mice IGF-2 increased cell excitability. Although further investigation is needed to clarify the cellular and molecular mechanisms underpinning IGF-2 effect on social behavior, our findings highlight IGF-2 as a potential pharmacological tool for the treatment of social dysfunctions associated with ASDs.
Collapse
Affiliation(s)
| | | | | | | | | | - Hannah Monyer
- European Brain Research Institute (EBRI), Rome, Italy
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Marilena Griguoli
- European Brain Research Institute (EBRI), Rome, Italy
- Institute of Molecular Biology and Pathology of the National Council of Research (IBPM-CNR), Rome, Italy
| |
Collapse
|
16
|
El Yacoubi FA, Oukabli M, Ibrahimi A, Kisra H, Bensaid M. Unraveling the Role of Neuroligin3 in Autism Spectrum Disorders: Pathophysiological Insights and Targeted Therapies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:801-811. [PMID: 37497709 DOI: 10.2174/1871527323666230727102244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023]
Abstract
Autism Spectrum Disorder is a neurodevelopmental disorder characterized by impaired social and communication skills, repetitive behaviors, and/or restricted interests with a prevalence of as high as 1% of children. Autism spectrum has strongly associated with genetic factors and exhibits wide clinical and heterogeneous genetic architecture. Most genes associated with Autism are involved in neuronal and synaptic development. The neuroligin3, the sex-linked gene on the X chromosome, was the first gene to be associated with a monogenic form of Autism. Neuroligin3 is a postsynaptic cell adhesion protein involved in synapse transmission, brain formation, and neuronal development. In this review, we provide recent findings on different mutations in the Neuroligin3 gene linked to Autism spectrum disorder and their molecular pathway effect. We also give the behavioral, and synaptic alterations reported in the Neuroligin3 animal model of Autism and the potential therapeutic strategies targeting the biological processes and the main symptoms of autism spectrum disorder. In addition, we discuss the use of novel technologies like induced pluripotent stem cells from Autistic patients that have the potential to differentiate in human neurons and therefore have a variety of applications in therapy and biomedical studies to search specific biomarkers, and develop systems for screening chemical molecules in human cells to discover target therapies.
Collapse
Affiliation(s)
- Fatima Azzahrae El Yacoubi
- Laboratory of Immunology, Infectious Disease and Tropical Biotechnology, Faculty of Pharmacy, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
- Medical and Pharmacy School, University Mohammed V, Rabat, Morocco
| | - Mohamed Oukabli
- Laboratory of Pathological Anatomy, Military Hospital Mohamed V, Rabat, Morocco
| | - Azeddine Ibrahimi
- Biotechnology Lab (MedBiotech), Rabat Medical and Pharmacy School, University Mohammed V, Rabat, Morocco
- Centre Mohammed VI for Research & Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Hassan Kisra
- Medical and Pharmacy School, University Mohammed V, Rabat, Morocco
- Center of Child Psychiatry, Arrazi Hospital, Salé, Morocco
| | - Mounia Bensaid
- Laboratory of Pathological Anatomy, Military Hospital Mohamed V, Rabat, Morocco
- Royal School of Military Health Service. Rabat, Morocco
| |
Collapse
|
17
|
Li WK, Zhang SQ, Peng WL, Shi YH, Yuan B, Yuan YT, Xue ZY, Wang JC, Han WJ, Chen ZF, Shan SF, Xue BQ, Chen JL, Zhang C, Zhu SJ, Tai YL, Cheng TL, Qiu ZL. Whole-brain in vivo base editing reverses behavioral changes in Mef2c-mutant mice. Nat Neurosci 2024; 27:116-128. [PMID: 38012399 DOI: 10.1038/s41593-023-01499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
Whole-brain genome editing to correct single-base mutations and reduce or reverse behavioral changes in animal models of autism spectrum disorder (ASD) has not yet been achieved. We developed an apolipoprotein B messenger RNA-editing enzyme, catalytic polypeptide-embedded cytosine base editor (AeCBE) system for converting C·G to T·A base pairs. We demonstrate its effectiveness by targeting AeCBE to an ASD-associated mutation of the MEF2C gene (c.104T>C, p.L35P) in vivo in mice. We first constructed Mef2cL35P heterozygous mice. Male heterozygous mice exhibited hyperactivity, repetitive behavior and social abnormalities. We then programmed AeCBE to edit the mutated C·G base pairs of Mef2c in the mouse brain through the intravenous injection of blood-brain barrier-crossing adeno-associated virus. This treatment successfully restored Mef2c protein levels in several brain regions and reversed the behavioral abnormalities in Mef2c-mutant mice. Our work presents an in vivo base-editing paradigm that could potentially correct single-base genetic mutations in the brain.
Collapse
Affiliation(s)
- Wei-Ke Li
- Songjiang Research Institute, Songjiang Hospital & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shu-Qian Zhang
- Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Wan-Ling Peng
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Han Shi
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bo Yuan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Ting Yuan
- Songjiang Research Institute, Songjiang Hospital & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen-Yu Xue
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Cheng Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wen-Jian Han
- Songjiang Research Institute, Songjiang Hospital & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Fang Chen
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Shi-Fang Shan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bi-Qing Xue
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jin-Long Chen
- Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Cheng Zhang
- Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shu-Jia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Lin Tai
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Tian-Lin Cheng
- Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Zi-Long Qiu
- Songjiang Research Institute, Songjiang Hospital & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- Clinical Neuroscience Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Li Y, Du W, Liu R, Zan G, Ye B, Li Q, Sheng Z, Yuan Y, Song Y, Liu J, Liu Z. Paraventricular nucleus-central amygdala oxytocinergic projection modulates pain-related anxiety-like behaviors in mice. CNS Neurosci Ther 2023; 29:3493-3506. [PMID: 37248645 PMCID: PMC10580334 DOI: 10.1111/cns.14282] [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: 01/04/2023] [Revised: 05/04/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023] Open
Abstract
AIMS Anxiety disorders associated with pain are a common health problem. However, the underlying mechanisms remain poorly understood. We aimed to investigate the role of paraventricular nucleus (PVN)-central nucleus of the amygdala (CeA) oxytocinergic projections in anxiety-like behaviors induced by inflammatory pain. METHODS After inflammatory pain induction by complete Freund's adjuvant (CFA), mice underwent elevated plus maze, light-dark transition test, and marble burying test to examine the anxiety-like behaviors. Chemogenetic, optogenetic, and fiber photometry recordings were used to modulate and record the activity of the oxytocinergic projections of the PVN-CeA. RESULTS The key results are as follows: inflammatory pain-induced anxiety-like behaviors in mice accompanied by decreased activity of PVN oxytocin neurons. Chemogenetic activation of PVN oxytocin neurons prevented pain-related anxiety-like behaviors, whereas inhibition of PVN oxytocin neurons induced anxiety-like behaviors in naïve mice. PVN oxytocin neurons projected directly to the CeA, and microinjection of oxytocin into the CeA blocked anxiety-like behaviors. Inflammatory pain also decreased the activity of CeA neurons, and optogenetic activation of PVNoxytocin -CeA circuit prevented anxiety-like behavior in response to inflammatory pain. CONCLUSION The results of our study suggest that oxytocin has anti-anxiety effects and provide novel insights into the role of PVNoxytocin -CeA projections in the regulation of anxiety-like behaviors induced by inflammatory pain.
Collapse
Affiliation(s)
- Yu‐Jie Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Wei‐Jia Du
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Rui Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Gui‐Ying Zan
- Key Laboratory of Receptor Research, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Bing‐Lu Ye
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Qian Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Zhi‐Hao Sheng
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Ya‐Wei Yuan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yu‐Jie Song
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jing‐Gen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Zhi‐Qiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
19
|
Hooshmandi M, Sharma V, Thörn Perez C, Sood R, Krimbacher K, Wong C, Lister KC, Ureña Guzmán A, Bartley TD, Rocha C, Maussion G, Nadler E, Roque PM, Gantois I, Popic J, Lévesque M, Kaufman RJ, Avoli M, Sanz E, Nader K, Hagerman RJ, Durcan TM, Costa-Mattioli M, Prager-Khoutorsky M, Lacaille JC, Martinez-Cerdeno V, Gibson JR, Huber KM, Sonenberg N, Gkogkas CG, Khoutorsky A. Excitatory neuron-specific suppression of the integrated stress response contributes to autism-related phenotypes in fragile X syndrome. Neuron 2023; 111:3028-3040.e6. [PMID: 37473758 PMCID: PMC10592416 DOI: 10.1016/j.neuron.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/11/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023]
Abstract
Dysregulation of protein synthesis is one of the key mechanisms underlying autism spectrum disorder (ASD). However, the role of a major pathway controlling protein synthesis, the integrated stress response (ISR), in ASD remains poorly understood. Here, we demonstrate that the main arm of the ISR, eIF2α phosphorylation (p-eIF2α), is suppressed in excitatory, but not inhibitory, neurons in a mouse model of fragile X syndrome (FXS; Fmr1-/y). We further show that the decrease in p-eIF2α is mediated via activation of mTORC1. Genetic reduction of p-eIF2α only in excitatory neurons is sufficient to increase general protein synthesis and cause autism-like behavior. In Fmr1-/y mice, restoration of p-eIF2α solely in excitatory neurons reverses elevated protein synthesis and rescues autism-related phenotypes. Thus, we reveal a previously unknown causal relationship between excitatory neuron-specific translational control via the ISR pathway, general protein synthesis, and core phenotypes reminiscent of autism in a mouse model of FXS.
Collapse
Affiliation(s)
- Mehdi Hooshmandi
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Vijendra Sharma
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Carolina Thörn Perez
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Rapita Sood
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Konstanze Krimbacher
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Calvin Wong
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Kevin C Lister
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Alba Ureña Guzmán
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Trevor D Bartley
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern California, MIND Institute, UC Davis Medical Center, Sacramento, CA, USA
| | - Cecilia Rocha
- The Neuro's Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Gilles Maussion
- The Neuro's Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Emma Nadler
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Patricia Margarita Roque
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Ilse Gantois
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Jelena Popic
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Maxime Lévesque
- Montreal Neurological Institute-Hospital, Departments of Neurology & Neurosurgery and of Physiology, McGill University, Montréal, QC, Canada
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Massimo Avoli
- Montreal Neurological Institute-Hospital, Departments of Neurology & Neurosurgery and of Physiology, McGill University, Montréal, QC, Canada
| | - Elisenda Sanz
- Department of Cell Biology, Physiology and Immunology, and Neuroscience Institute, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Karim Nader
- Department of Psychology, Faculty of Science, McGill University, Montréal, QC, Canada
| | - Randi Jenssen Hagerman
- MIND Institute and Department of Pediatrics, University of California at Davis Medical Center, Sacramento, CA, USA
| | - Thomas M Durcan
- The Neuro's Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | | | | | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning, and Research Group on Neural Signaling and Circuitry, Université de Montréal, Montréal, QC, Canada
| | - Veronica Martinez-Cerdeno
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern California, MIND Institute, UC Davis Medical Center, Sacramento, CA, USA
| | - Jay R Gibson
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montréal, QC, Canada.
| | - Christos G Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece.
| | - Arkady Khoutorsky
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada.
| |
Collapse
|
20
|
Shen LP, Li W, Pei LZ, Yin J, Xie ST, Li HZ, Yan C, Wang JJ, Zhang Q, Zhang XY, Zhu JN. Oxytocin Receptor in Cerebellar Purkinje Cells Does Not Engage in Autism-Related Behaviors. CEREBELLUM (LONDON, ENGLAND) 2023; 22:888-904. [PMID: 36040660 DOI: 10.1007/s12311-022-01466-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
The classical motor center cerebellum is one of the most consistent structures of abnormality in autism spectrum disorders (ASD), and neuropeptide oxytocin is increasingly explored as a potential pharmacotherapy for ASD. However, whether oxytocin targets the cerebellum for therapeutic effects remains unclear. Here, we report a localization of oxytocin receptor (OXTR) in Purkinje cells (PCs) of cerebellar lobule Crus I, which is functionally connected with ASD-implicated circuits. OXTR activation neither affects firing activities, intrinsic excitability, and synaptic transmission of normal PCs nor improves abnormal intrinsic excitability and synaptic transmission of PCs in maternal immune activation (MIA) mouse model of autism. Furthermore, blockage of OXTR in Crus I in wild-type mice does not induce autistic-like social, stereotypic, cognitive, and anxiety-like behaviors. These results suggest that oxytocin signaling in Crus I PCs seems to be uninvolved in ASD pathophysiology, and contribute to understanding of targets and mechanisms of oxytocin in ASD treatment.
Collapse
Affiliation(s)
- Li-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wei Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ling-Zhu Pei
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jun Yin
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Qipeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
21
|
Kong XJ, Kang J, Liu K. Probiotic and intra-nasal oxytocin combination therapy on autonomic function and gut-brain axis signaling in young children and teens with autism spectrum disorder. J Psychiatr Res 2023; 166:1-9. [PMID: 37639877 DOI: 10.1016/j.jpsychires.2023.08.006] [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: 08/22/2022] [Revised: 07/05/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
Autonomic dysfunction has been widely studied in individuals with autism spectral disorder (ASD); however, the autonomic response to probiotic and oxytocin (OT) combination intervention has not yet been explored. We conducted the present study that includes 35 individuals with ASD aged 3-20 years to explore autonomic responses to daily Lactobacillus plantarum probiotic supplementation and OT nasal spray treatment both alone and in combination. We identified significant improvements in autonomic indices from subjects receiving combination treatment relative to those receiving placebo. Parameters that were observed to improve following combination treatment are time domain metrics of heart rate variability (HRV), including the root mean square of successive differences between normal heartbeats (RMSSD), standard deviation of normal-to-normal R-R intervals (SDNN), and proportion of the number of pairs of adjacent NN intervals that differ by more than 50ms (pNN50, p < 0.05). Furthermore, individuals that received either probiotics or OT alone demonstrated fewer changes in RMSSD, pNN50, and SDNN. Several parameters that demonstrated significant improvements in combination therapy were found to be correlated with baseline levels of OT (LF power: r = -0.86, p = 0.024; mean HR: r = 0.89, p = 0.012). Additionally, Social Responsiveness Scale (SRS) raw total scores (mean HR, r = 0.86, p = 0.024) and Aberrant Behavior Checklist (ABC) raw total scores (mean HR r = 0.94, p = 0.017) were correlated with mean heart rate (HR) and HRV-derived parameters. These results provide further evidence of synergy of probiotic and OT combination and help us gain a better understanding of the role of the gut-brain axis in ASD phenotypes and pathogenesis.
Collapse
Affiliation(s)
- Xue-Jun Kong
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jiayi Kang
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kevin Liu
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
22
|
Ling W, Ren Z, Wang W, Lu D, Zhou Q, Liu Q, Jiang G. Chronic Ambient Ozone Exposure Aggravates Autism-Like Symptoms in a Susceptible Mouse Model. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14248-14259. [PMID: 37676697 DOI: 10.1021/acs.est.3c00607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Although there is evidence that exposure to ground-level ozone (O3) may cause an increased risk of neurological disorders (e.g., autistic spectrum disorder), low-dose chronic ozone exposure and its adverse effects on the nervous system have not been fully understood. Here, we evaluated the potential neurotoxic effects of long-term exposure to environmentally relevant O3 concentration (200 μg/m3 via a whole-body inhalation system, 12 h/day for 5 days/week) using a susceptible mouse model of autism induced by valproic acid. Various indicators of oxidative stress, mitochondria, and synapse in the brain tissues were then measured to determine the overall damage of O3 to the mouse brain. The results showed an aggravated risk of autism in mice offspring, which was embodied in decreased antioxidant contents, disturbed energy generation in mitochondria, as well as reduced expressions of protein kinase Mζ (PKMζ) and synaptic proteins [e.g., Synapsin 1 (SYN 1), postsynaptic density protein-95 (PSD-95)]. Overall, our study indicates that prenatal exposure to environmentally relevant O3 may exacerbate the symptoms of autism, shedding light on possible molecular mechanisms and providing valuable insights into the pathogenesis of autism, especially concerning low-dose levels of those pollutants.
Collapse
Affiliation(s)
- Weibo Ling
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihua Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Weichao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Dawei Lu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Qian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Taishan Institute for Ecology and Environment (TIEE), Jinan 250100, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
23
|
László K, Vörös D, Correia P, Fazekas CL, Török B, Plangár I, Zelena D. Vasopressin as Possible Treatment Option in Autism Spectrum Disorder. Biomedicines 2023; 11:2603. [PMID: 37892977 PMCID: PMC10603886 DOI: 10.3390/biomedicines11102603] [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: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is rather common, presenting with prevalent early problems in social communication and accompanied by repetitive behavior. As vasopressin was implicated not only in salt-water homeostasis and stress-axis regulation, but also in social behavior, its role in the development of ASD might be suggested. In this review, we summarized a wide range of problems associated with ASD to which vasopressin might contribute, from social skills to communication, motor function problems, autonomous nervous system alterations as well as sleep disturbances, and altered sensory information processing. Beside functional connections between vasopressin and ASD, we draw attention to the anatomical background, highlighting several brain areas, including the paraventricular nucleus of the hypothalamus, medial preoptic area, lateral septum, bed nucleus of stria terminalis, amygdala, hippocampus, olfactory bulb and even the cerebellum, either producing vasopressin or containing vasopressinergic receptors (presumably V1a). Sex differences in the vasopressinergic system might underline the male prevalence of ASD. Moreover, vasopressin might contribute to the effectiveness of available off-label therapies as well as serve as a possible target for intervention. In this sense, vasopressin, but paradoxically also V1a receptor antagonist, were found to be effective in some clinical trials. We concluded that although vasopressin might be an effective candidate for ASD treatment, we might assume that only a subgroup (e.g., with stress-axis disturbances), a certain sex (most probably males) and a certain brain area (targeting by means of virus vectors) would benefit from this therapy.
Collapse
Affiliation(s)
- Kristóf László
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dávid Vörös
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Imola Plangár
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dóra Zelena
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| |
Collapse
|
24
|
Pavăl D. The dopamine hypothesis of autism spectrum disorder: A comprehensive analysis of the evidence. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:1-42. [PMID: 37993174 DOI: 10.1016/bs.irn.2023.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Despite intensive research into the etiopathogenesis of autism spectrum disorder (ASD), limited progress has been achieved so far. Among the plethora of models seeking to clarify how ASD arises, a coherent dopaminergic model was lacking until recently. In 2017, we provided a theoretical framework that we designated "the dopamine hypothesis of ASD". In the meantime, numerous studies yielded empirical evidence for this model. 4 years later, we provided a second version encompassing a refined and reconceptualized framework that accounted for these novel findings. In this chapter, we will review the evidence backing the previous versions of our model and add the most recent developments to the picture. Along these lines, we intend to lay out a comprehensive analysis of the supporting evidence for the dopamine hypothesis of ASD.
Collapse
Affiliation(s)
- Denis Pavăl
- The Romanian Association for Autoimmune Encephalitis, Cluj-Napoca, Romania; Department of Psychiatry, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
25
|
Li Y, Zhu M, Chen WX, Luo J, Li X, Cao Y, Zheng M, Ma S, Xiao Z, Zhang Y, Jiang L, Wang X, Tan T, Li X, Gong Q, Xiong X, Wang J, Tang M, Li M, Tang YP. A novel mutation in intron 1 of Wnt1 causes developmental loss of dopaminergic neurons in midbrain and ASD-like behaviors in rats. Mol Psychiatry 2023; 28:3795-3805. [PMID: 37658228 PMCID: PMC10730402 DOI: 10.1038/s41380-023-02223-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders with a strong genetic liability. Despite extensive studies, however, the underlying pathogenic mechanism still remains elusive. In the present study, we identified a homozygous mutation in the intron 1 of Wnt1 via large-scale screening of ASD risk/causative genes and verified that this mutation created a new splicing donor site in the intron 1, and consequently, a decrease of WNT1 expression. Interestingly, humanized rat models harboring this mutation exhibited robust ASD-like behaviors including impaired ultrasonic vocalization (USV), decreased social interactions, and restricted and repetitive behaviors. Moreover, in the substantia nigra compacta (SNpc) and the ventral tegmental area (VTA) of mutant rats, dopaminergic (DAergic) neurons were dramatically lost, together with a comparable decrease in striatal DAergic fibers. Furthermore, using single-cell RNA sequencing, we demonstrated that the decreased DAergic neurons in these midbrain areas might attribute to a shift of the boundary of the local pool of progenitor cells from the hypothalamic floor plate to the midbrain floor plate during the early embryonic stage. Moreover, treatments of mutant rats with levodopa could attenuate the impaired USV and social interactions almost completely, but not the restricted and repetitive behaviors. Our results for the first time documented that the developmental loss of DAergic neurons in the midbrain underlies the pathogenesis of ASD, and that the abnormal progenitor cell patterning is a cellular underpinning for this developmental DAergic neuronal loss. Importantly, the effective dopamine therapy suggests a translational significance in the treatment of ASD.
Collapse
Affiliation(s)
- Yongyi Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jing Luo
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yangyang Cao
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Meng Zheng
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhilan Xiao
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yani Zhang
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Linyan Jiang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiumin Wang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ting Tan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xia Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qian Gong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoli Xiong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jun Wang
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mingxi Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Ya-Ping Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
26
|
Portnova GV, Proskurnina EV, Skorokhodov IV, Sokolova SV, Semirechenko AN, Varlamov AA. Salivary Oxytocin and Antioxidative Response to Robotic Touch in Adults with Autism Spectrum Disorder. Int J Mol Sci 2023; 24:12322. [PMID: 37569698 PMCID: PMC10419114 DOI: 10.3390/ijms241512322] [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: 05/15/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 08/13/2023] Open
Abstract
Individuals with ASD are known to have a tendency to have tactile sensory processing issues that could be associated with their impairment as regards social communication. The alterations in tactile processing in autistic subjects are usually accompanied by hypersensitivity and other unpleasant emotions induced by tactile contact. In our study, we investigated the impact of the velocity and the force of a tactile stroke received impersonally by a custom-built robotic device. A total of 21 adults with ASD and 22 adults from a control group participated in our study. The participants' responses were assessed according to subjective scales, EEG changes, and the dynamics of saliva antioxidants and oxytocin. It was found that the oxytocin level was significantly lower in subjects with ASD but increased after tactile stimulation. However, contrary to expectations, the increase in the oxytocin level in the target group negatively correlated with the subjective pleasantness of tactile stimulation and was probably associated with a stress-induced effect. The basic levels of antioxidants did not differ between the TD and ASD groups; however, these had significantly increased in individuals with ASD by the end of the study. The EEG findings, which revealed enhanced antioxidant levels, contributed to the relief of the cognitive control during the study.
Collapse
Affiliation(s)
- Galina V. Portnova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, 5A Butlerova Str., 117485 Moscow, Russia
- Tactile Communication Research Laboratory, Pushkin State Russian Language Institute, 6 Volgina Str., 117485 Moscow, Russia
| | - Elena V. Proskurnina
- Laboratory of Molecular Biology, Research Centre for Medical Genetics, 1 Moskvorechye Str., 115522 Moscow, Russia;
| | - Ivan V. Skorokhodov
- Tactile Communication Research Laboratory, Pushkin State Russian Language Institute, 6 Volgina Str., 117485 Moscow, Russia
- Autonomous Non-Profit Organization “Our Sunny World”, 98 Nizhegorodskaya Str., 109052 Moscow, Russia
| | - Svetlana V. Sokolova
- Medical Scientific and Educational Center, Lomonosov Moscow State University, Lomonosovsky Prosp. 27-10, 119991 Moscow, Russia
| | - Alexey N. Semirechenko
- Tactile Communication Research Laboratory, Pushkin State Russian Language Institute, 6 Volgina Str., 117485 Moscow, Russia
| | - Anton A. Varlamov
- Autonomous Non-Profit Organization “Our Sunny World”, 98 Nizhegorodskaya Str., 109052 Moscow, Russia
| |
Collapse
|
27
|
Sato M, Nakai N, Fujima S, Choe KY, Takumi T. Social circuits and their dysfunction in autism spectrum disorder. Mol Psychiatry 2023; 28:3194-3206. [PMID: 37612363 PMCID: PMC10618103 DOI: 10.1038/s41380-023-02201-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
Social behaviors, how individuals act cooperatively and competitively with conspecifics, are widely seen across species. Rodents display various social behaviors, and many different behavioral paradigms have been used for investigating their neural circuit bases. Social behavior is highly vulnerable to brain network dysfunction caused by neurological and neuropsychiatric conditions such as autism spectrum disorders (ASDs). Studying mouse models of ASD provides a promising avenue toward elucidating mechanisms of abnormal social behavior and potential therapeutic targets for treatment. In this review, we outline recent progress and key findings on neural circuit mechanisms underlying social behavior, with particular emphasis on rodent studies that monitor and manipulate the activity of specific circuits using modern systems neuroscience approaches. Social behavior is mediated by a distributed brain-wide network among major cortical (e.g., medial prefrontal cortex (mPFC), anterior cingulate cortex, and insular cortex (IC)) and subcortical (e.g., nucleus accumbens, basolateral amygdala (BLA), and ventral tegmental area) structures, influenced by multiple neuromodulatory systems (e.g., oxytocin, dopamine, and serotonin). We particularly draw special attention to IC as a unique cortical area that mediates multisensory integration, encoding of ongoing social interaction, social decision-making, emotion, and empathy. Additionally, a synthesis of studies investigating ASD mouse models demonstrates that dysfunctions in mPFC-BLA circuitry and neuromodulation are prominent. Pharmacological rescues by local or systemic (e.g., oral) administration of various drugs have provided valuable clues for developing new therapeutic agents for ASD. Future efforts and technological advances will push forward the next frontiers in this field, such as the elucidation of brain-wide network activity and inter-brain neural dynamics during real and virtual social interactions, and the establishment of circuit-based therapy for disorders affecting social functions.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Kita, Sapporo, 060-8638, Japan
| | - Nobuhiro Nakai
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe, 650-0017, Japan
| | - Shuhei Fujima
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe, 650-0017, Japan
| | - Katrina Y Choe
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON, Canada
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe, 650-0017, Japan.
- RIKEN Center for Biosystems Dynamics Research, Chuo, Kobe, 650-0047, Japan.
| |
Collapse
|
28
|
Zhu J, Meng H, Li Y. Identification of target hub genes and construction of a novel miRNA regulatory network in autism spectrum disorder by integrated analysis. Medicine (Baltimore) 2023; 102:e34420. [PMID: 37478258 PMCID: PMC10662836 DOI: 10.1097/md.0000000000034420] [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: 04/06/2023] [Accepted: 06/29/2023] [Indexed: 07/23/2023] Open
Abstract
The incidence of autism spectrum disorder (ASD) is increasing year by year in children. The aim of the study was to find possible biomarkers for ASD diagnosis as well as examine MicroRNA (miRNA) signatures and crucial pathways. We conducted a two-stage study to explore potential target genes and functional miRNAs. Peripheral blood samples of children with ASD were enrolled and performed RNA sequencing analysis. The overlapped candidate genes were further screened in combination with differentially expressed genes (DEGs) of GSE77103 datasets. STRING established a protein-protein interaction network comprising DEGs. The hub genes were filtered out using the CytoHubba. Then, we set up a miRNA-mRNA regulatory network. Correlational analyses between hub genes and immune cells associated with ASD were carried out using the CIBERSORT software to assess the diversity of immune cell types in ASD. RNA-sequencing analysis was used to confirm the differential expression of 3 hub genes. Briefly, after blood samples were sequenced interrogating 867 differential genes in our internal screening dataset. After screening GEO databases, 551 DEGs obtained from GSE77103. Fourteen common genes were overlapped through DEGs of GEO datasets and internal screening dataset. Among protein-protein interaction network, 10 hub genes with high degree algorithm were screened out and 3 hub genes of them - ADIPOR1, LGALS3, and GZMB - that were thought to be most associated with the emergence of ASD. Then, we developed a network of miRNA-mRNA regulatory interactions by screening miRNAs (such as hsa-miR-20b-5p, hsa-miR-17-5p, and hsa-miR-216b-5p) that were closely associated to 3 hub genes. Additionally, we discovered 18 different immune cell types associated with ASD using the CIBERSORT algorithm, and we discovered that mononuclear macrophages differed considerably between the 2 groups. Overall, 3 hub genes (ADIPOR1, LGALS3, and GZMB) and 15 candidates miRNAs-target 3 genes regulatory pathways representing potentially novel biomarkers of ASD diseases were revealed. These findings could enhance our knowledge of ASD and offer possible therapeutic targets of ASD patients in the future.
Collapse
Affiliation(s)
- Jinyi Zhu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Haoran Meng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital affiliated to Qingdao University, Jinan, China
| |
Collapse
|
29
|
Oubraim S, Shen RY, Haj-Dahmane S. Oxytocin excites dorsal raphe serotonin neurons and bidirectionally gates their glutamate synapses. iScience 2023; 26:106707. [PMID: 37250336 PMCID: PMC10214716 DOI: 10.1016/j.isci.2023.106707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Oxytocin (OXT) modulates wide spectrum of social and emotional behaviors via modulation of numerous neurotransmitter systems, including serotonin (5-HT). However, how OXT controls the function of dorsal raphe nucleus (DRN) 5-HT neurons remains unknown. Here, we reveal that OXT excites and alters the firing pattern of 5-HT neurons via activation of postsynaptic OXT receptors (OXTRs). In addition, OXT induces cell-type-specific depression and potentiation of DRN glutamate synapses by two retrograde lipid messengers, 2-arachidonoylglycerol (2-AG) and arachidonic acid (AA), respectively. Neuronal mapping demonstrates that OXT preferentially potentiates glutamate synapses of 5-HT neurons projecting to medial prefrontal cortex (mPFC) and depresses glutamatergic inputs to 5-HT neurons projecting to lateral habenula (LHb) and central amygdala (CeA). Thus, by engaging distinct retrograde lipid messengers, OXT exerts a target-specific gating of glutamate synapses on the DRN. As such, our data uncovers the neuronal mechanisms by which OXT modulates the function of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
30
|
Morel C, Martinez Sanchez I, Cherifi Y, Chartrel N, Diaz Heijtz R. Perturbation of maternal gut microbiota in mice during a critical perinatal window influences early neurobehavioral outcomes in offspring. Neuropharmacology 2023; 229:109479. [PMID: 36870672 DOI: 10.1016/j.neuropharm.2023.109479] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut microbiota is increasingly recognized as a key environmental factor that shapes host development and physiology, including neural circuits formation and function. Concurrently, there has been growing concern that early-life antibiotic exposure may alter brain developmental trajectories, increasing the risk for neurodevelopmental disorders such as autism spectrum disorder (ASD). Here, we assessed whether perturbation of the maternal gut microbiota in mice during a narrow critical perinatal window (last week of pregnancy and first three postnatal days), induced by exposure to a commonly used broad-spectrum oral antibiotic (ampicillin), influences offspring neurobehavioral outcomes relevant to ASD. Our results demonstrate that neonatal offspring from antibiotic-treated dams display an altered pattern of ultrasonic communication, which was more pronounced in males. Moreover, juvenile male, but not female, offspring from antibiotic-treated dams showed reduced social motivation and social interaction, as well as context-dependent anxiety-like behavior. However, no changes were observed in locomotor or exploratory activity. This behavioral phenotype of exposed juvenile males was associated with reduced gene expression of the oxytocin receptor (OXTR) and several tight-junction proteins in the prefrontal cortex, a key region involved in the regulation of social and emotional behaviors, as well as a mild inflammatory response in the colon. Further, juvenile offspring from exposed dams also showed distinct alterations in several gut bacterial species, including, Lactobacillus murinus, and Parabacteroides goldsteinii. Overall, this study highlights the importance of the maternal microbiome in early-life, and how its perturbation by a widely used antibiotic could contribute to atypical social and emotional development of offspring in a sex-dependent manner.
Collapse
Affiliation(s)
- Cassandre Morel
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden; University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | | | - Yamina Cherifi
- University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | - Nicolas Chartrel
- University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | | |
Collapse
|
31
|
Chalkiadaki K, Hooshmandi M, Lach G, Statoulla E, Simbriger K, Amorim IS, Kouloulia S, Zafeiri M, Pothos P, Bonneil É, Gantois I, Popic J, Kim SH, Wong C, Cao R, Komiyama NH, Atlasi Y, Jafarnejad SM, Khoutorsky A, Gkogkas CG. Mnk1/2 kinases regulate memory and autism-related behaviours via Syngap1. Brain 2023; 146:2175-2190. [PMID: 36315645 PMCID: PMC10411928 DOI: 10.1093/brain/awac398] [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: 02/04/2022] [Revised: 09/03/2022] [Accepted: 10/01/2022] [Indexed: 11/14/2022] Open
Abstract
MAPK interacting protein kinases 1 and 2 (Mnk1/2) regulate a plethora of functions, presumably via phosphorylation of their best characterized substrate, eukaryotic translation initiation factor 4E (eIF4E) on Ser209. Here, we show that, whereas deletion of Mnk1/2 (Mnk double knockout) impairs synaptic plasticity and memory in mice, ablation of phospho-eIF4E (Ser209) does not affect these processes, suggesting that Mnk1/2 possess additional downstream effectors in the brain. Translational profiling revealed only a small overlap between the Mnk1/2- and phospho-eIF4E(Ser209)-regulated translatome. We identified the synaptic Ras GTPase activating protein 1 (Syngap1), encoded by a syndromic autism gene, as a downstream target of Mnk1 because Syngap1 immunoprecipitated with Mnk1 and showed reduced phosphorylation (S788) in Mnk double knockout mice. Knockdown of Syngap1 reversed memory deficits in Mnk double knockout mice and pharmacological inhibition of Mnks rescued autism-related phenotypes in Syngap1+/- mice. Thus, Syngap1 is a downstream effector of Mnk1, and the Mnks-Syngap1 axis regulates memory formation and autism-related behaviours.
Collapse
Affiliation(s)
- Kleanthi Chalkiadaki
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
- Centre for Discovery Brain Sciences and The Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Mehdi Hooshmandi
- Department of Anesthesia and Alan Edwards Centre for Research on Pain, McGill University, Montréal H3A 0G1, Canada
| | - Gilliard Lach
- Centre for Discovery Brain Sciences and The Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Elpida Statoulla
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Konstanze Simbriger
- Centre for Discovery Brain Sciences and The Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Ines S Amorim
- Centre for Discovery Brain Sciences and The Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Stella Kouloulia
- Centre for Discovery Brain Sciences and The Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Maria Zafeiri
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Panagiotis Pothos
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, Station Centreville, Montréal H3C 3J7, Canada
| | - Ilse Gantois
- Goodman Cancer Institute and Biochemistry Department, McGill University, Montréal H3A 1A3, Canada
| | - Jelena Popic
- Goodman Cancer Institute and Biochemistry Department, McGill University, Montréal H3A 1A3, Canada
| | - Sung-Hoon Kim
- Goodman Cancer Institute and Biochemistry Department, McGill University, Montréal H3A 1A3, Canada
| | - Calvin Wong
- Department of Anesthesia and Alan Edwards Centre for Research on Pain, McGill University, Montréal H3A 0G1, Canada
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Noboru H Komiyama
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast BT9 7AE, Northern Ireland, UK
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast BT9 7AE, Northern Ireland, UK
| | - Arkady Khoutorsky
- Department of Anesthesia and Alan Edwards Centre for Research on Pain, McGill University, Montréal H3A 0G1, Canada
| | - Christos G Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| |
Collapse
|
32
|
Evaluation of Individuals with Non-Syndromic Global Developmental Delay and Intellectual Disability. CHILDREN 2023; 10:children10030414. [PMID: 36979972 PMCID: PMC10047567 DOI: 10.3390/children10030414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Global Developmental Delay (GDD) and Intellectual Disability (ID) are two of the most common presentations encountered by physicians taking care of children. GDD/ID is classified into non-syndromic GDD/ID, where GDD/ID is the sole evident clinical feature, or syndromic GDD/ID, where there are additional clinical features or co-morbidities present. Careful evaluation of children with GDD and ID, starting with detailed history followed by a thorough examination, remain the cornerstone for etiologic diagnosis. However, when initial history and examination fail to identify a probable underlying etiology, further genetic testing is warranted. In recent years, genetic testing has been shown to be the single most important diagnostic modality for clinicians evaluating children with non-syndromic GDD/ID. In this review, we discuss different genetic testing currently available, review common underlying copy-number variants and molecular pathways, explore the recent evidence and recommendations for genetic evaluation and discuss an approach to the diagnosis and management of children with non-syndromic GDD and ID.
Collapse
|
33
|
Muscatelli F. [As early as birth, oxytocin plays a key role in both food and social behavior]. Biol Aujourdhui 2023; 216:131-143. [PMID: 36744979 DOI: 10.1051/jbio/2022017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Indexed: 02/07/2023]
Abstract
Oxytocin (OT) is a neurohormone that regulates the so-called "social brain" and is mainly studied in adulthood. During postnatal development, the mechanisms by which the OT system structures various behaviors are little studied. Here we present the dynamic process of postnatal development of the OT system as well as the OT functions in the perinatal period that are essential for shaping social behaviors. Specifically, we discuss the role of OT, in the newborn, in integrating and adapting responses to early sensory stimuli and in stimulating suckling activity. Sensory dialogue and suckling are involved in mother-infant bonds and structure future social interactions. In rodents and humans, neurodevelopmental diseases with autism spectrum disorders (ASD), such as Prader-Willi and Schaaf-Yang syndromes, are associated with sensory, feeding and behavioral deficits in infancy. We propose that in early postnatal life, OT plays a key role in stimulating the maturation of neural networks controlling feeding behavior and early social interactions from birth. Administration of OT at birth improves sensory integration of environmental factors and the relationship with the mother as well as sucking activity as we have shown in mouse models and in babies with Prader-Willi syndrome. Long-term effects have also been observed on social and cognitive behavior. Therefore, early feeding difficulties might be an early predictive marker of ASD, and OT treatment a promising option to improve feeding behavior and, in the longer term, social behavioral problems.
Collapse
Affiliation(s)
- Françoise Muscatelli
- INMED (Institut de Neurobiologie de la Méditerranée), INSERM, Aix Marseille Univ, Marseille, France
| |
Collapse
|
34
|
Zayan U, Caccialupi Da Prato L, Muscatelli F, Matarazzo V. Modulation of the thermosensory system by oxytocin. Front Mol Neurosci 2023; 15:1075305. [PMID: 36698777 PMCID: PMC9868264 DOI: 10.3389/fnmol.2022.1075305] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023] Open
Abstract
Oxytocin (OT) is a neurohormone involved early in neurodevelopment and is implicated in multiple functions, including sensory modulation. Evidence of such modulation has been observed for different sensory modalities in both healthy and pathological conditions. This review summarizes the pleiotropic modulation that OT can exercise on an often overlooked sensory system: thermosensation. This system allows us to sense temperature variations and compensate for the variation to maintain a stable core body temperature. Oxytocin modulates autonomic and behavioral mechanisms underlying thermoregulation at both central and peripheral levels. Hyposensitivity or hypersensitivity for different sensory modalities, including thermosensitivity, is a common feature in autism spectrum disorder (ASD), recapitulated in several ASD mouse models. These sensory dysregulations occur early in post-natal development and are correlated with dysregulation of the oxytocinergic system. In this study, we discussed the potential link between thermosensory atypia and the dysregulation of the oxytocinergic system in ASD.
Collapse
|
35
|
SCGN deficiency is a risk factor for autism spectrum disorder. Signal Transduct Target Ther 2023; 8:3. [PMID: 36588101 PMCID: PMC9806109 DOI: 10.1038/s41392-022-01225-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/15/2022] [Accepted: 09/30/2022] [Indexed: 01/03/2023] Open
Abstract
Autism spectrum disorder (ASD) affects 1-2% of all children and poses a great social and economic challenge for the globe. As a highly heterogeneous neurodevelopmental disorder, the development of its treatment is extremely challenging. Multiple pathways have been linked to the pathogenesis of ASD, including signaling involved in synaptic function, oxytocinergic activities, immune homeostasis, chromatin modifications, and mitochondrial functions. Here, we identify secretagogin (SCGN), a regulator of synaptic transmission, as a new risk gene for ASD. Two heterozygous loss-of-function mutations in SCGN are presented in ASD probands. Deletion of Scgn in zebrafish or mice leads to autism-like behaviors and impairs brain development. Mechanistically, Scgn deficiency disrupts the oxytocin signaling and abnormally activates inflammation in both animal models. Both ASD probands carrying Scgn mutations also show reduced oxytocin levels. Importantly, we demonstrate that the administration of oxytocin and anti-inflammatory drugs can attenuate ASD-associated defects caused by SCGN deficiency. Altogether, we identify a convergence between a potential autism genetic risk factor SCGN, and the pathological deregulation in oxytocinergic signaling and immune responses, providing potential treatment for ASD patients suffering from SCGN deficiency. Our study also indicates that it is critical to identify and stratify ASD patient populations based on their disease mechanisms, which could greatly enhance therapeutic success.
Collapse
|
36
|
Muscatelli F, Matarazzo V, Chini B. Neonatal oxytocin gives the tempo of social and feeding behaviors. Front Mol Neurosci 2022; 15:1071719. [PMID: 36583080 PMCID: PMC9792990 DOI: 10.3389/fnmol.2022.1071719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
The nonapeptide oxytocin (OT) is a master regulator of the social brain in early infancy, adolescence, and adult life. Here, we review the postnatal dynamic development of OT-system as well as early-life OT functions that are essential for shaping social behaviors. We specifically address the role of OT in neonates, focusing on its role in modulating/adapting sensory input and feeding behavior; both processes are involved in the establishing mother-infant bond, a crucial event for structuring all future social interactions. In patients and rodent models of Prader-Willi and Schaaf-Yang syndromes, two neurodevelopmental diseases characterized by autism-related features, sensory impairments, and feeding difficulties in early infancy are linked to an alteration of OT-system. Successful preclinical studies in mice and a phase I/II clinical trial in Prader-Willi babies constitute a proof of concept that OT-treatment in early life not only improves suckling deficit but has also a positive long-term effect on learning and social behavior. We propose that in early postnatal life, OT plays a pivotal role in stimulating and coordinating the maturation of neuronal networks controlling feeding behavior and the first social interactions. Consequently, OT therapy might be considered to improve feeding behavior and, all over the life, social cognition, and learning capabilities.
Collapse
Affiliation(s)
- Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France,*Correspondence: Françoise Muscatelli,
| | - Valery Matarazzo
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | - Bice Chini
- Institute of Neuroscience, National Research Council (CNR), Vedano al Lambro, Italy and NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
37
|
Can Translational Social Neuroscience Research Offer Insights to Mitigate Structural Racism in the United States? BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:1258-1267. [PMID: 35609781 DOI: 10.1016/j.bpsc.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 12/16/2022]
Abstract
Social isolation and conflict due to structural racism may result in human suffering and loneliness across the life span. Given the rising prevalence of these problems in the United States, combined with disruptions experienced during the COVID-19 pandemic, the neurobiology of affiliative behaviors may offer practical solutions to the pressing challenges associated with structural racism. Controlled experiments across species demonstrate that social connections are critical to survival, although strengthening individual resilience is insufficient to address the magnitude and impact of structural racism. In contrast, the multilevel construct of social resilience, defined by the power of groups to cultivate, engage in, and sustain positive relationships that endure and recuperate from social adversities, offers unique insights that may have greater impact, reach, and durability than individual-level interventions. Here, we review putative social resilience-enhancing interventions and, when available, their biological mediators, with the hope to stimulate discovery of novel approaches to mitigate structural racism. We first explore the social neuroscience principles underlying psychotherapy and other psychiatric interventions. Then, we explore translational efforts across species to tailor treatments that increase social resilience, with context and cultural sensitivity in mind. Finally, we conclude with some practical future directions for understudied areas that may be essential for progress in biological psychiatry, including ethical ways to increase representation in research and developing social paradigms that inform dynamics toward or away from socially resilient outcomes.
Collapse
|
38
|
Abstract
Recent advances in genomics have revealed a wide spectrum of genetic variants associated with neurodevelopmental disorders at an unprecedented scale. An increasing number of studies have consistently identified mutations-both inherited and de novo-impacting the function of specific brain circuits. This suggests that, during brain development, alterations in distinct neural circuits, cell types, or broad regulatory pathways ultimately shaping synapses might be a dysfunctional process underlying these disorders. Here, we review findings from human studies and animal model research to provide a comprehensive description of synaptic and circuit mechanisms implicated in neurodevelopmental disorders. We discuss how specific synaptic connections might be commonly disrupted in different disorders and the alterations in cognition and behaviors emerging from imbalances in neuronal circuits. Moreover, we review new approaches that have been shown to restore or mitigate dysfunctional processes during specific critical windows of brain development. Considering the heterogeneity of neurodevelopmental disorders, we also highlight the recent progress in developing improved clinical biomarkers and strategies that will help to identify novel therapeutic compounds and opportunities for early intervention.
Collapse
Affiliation(s)
- David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
- Current affiliation: Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
| |
Collapse
|
39
|
Triana-Del Rio R, Ranade S, Guardado J, LeDoux J, Klann E, Shrestha P. The modulation of emotional and social behaviors by oxytocin signaling in limbic network. Front Mol Neurosci 2022; 15:1002846. [PMID: 36466805 PMCID: PMC9714608 DOI: 10.3389/fnmol.2022.1002846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2024] Open
Abstract
Neuropeptides can exert volume modulation in neuronal networks, which account for a well-calibrated and fine-tuned regulation that depends on the sensory and behavioral contexts. For example, oxytocin (OT) and oxytocin receptor (OTR) trigger a signaling pattern encompassing intracellular cascades, synaptic plasticity, gene expression, and network regulation, that together function to increase the signal-to-noise ratio for sensory-dependent stress/threat and social responses. Activation of OTRs in emotional circuits within the limbic forebrain is necessary to acquire stress/threat responses. When emotional memories are retrieved, OTR-expressing cells act as gatekeepers of the threat response choice/discrimination. OT signaling has also been implicated in modulating social-exposure elicited responses in the neural circuits within the limbic forebrain. In this review, we describe the cellular and molecular mechanisms that underlie the neuromodulation by OT, and how OT signaling in specific neural circuits and cell populations mediate stress/threat and social behaviors. OT and downstream signaling cascades are heavily implicated in neuropsychiatric disorders characterized by emotional and social dysregulation. Thus, a mechanistic understanding of downstream cellular effects of OT in relevant cell types and neural circuits can help design effective intervention techniques for a variety of neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Sayali Ranade
- Department of Neurobiology and Behavior, School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Jahel Guardado
- Center for Neural Science, New York University, New York, NY, United States
| | - Joseph LeDoux
- Center for Neural Science, New York University, New York, NY, United States
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, United States
| | - Prerana Shrestha
- Department of Neurobiology and Behavior, School of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
40
|
Wang L, Suyama S, Lee SA, Ueta Y, Seino Y, Sharp GWG, Yada T. Fasting inhibits excitatory synaptic input on paraventricular oxytocin neurons via neuropeptide Y and Y1 receptor, inducing rebound hyperphagia, and weight gain. Front Nutr 2022; 9:994827. [PMID: 36337662 PMCID: PMC9627337 DOI: 10.3389/fnut.2022.994827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/28/2022] [Indexed: 10/15/2023] Open
Abstract
Fasting with varying intensities is used to treat obesity-related diseases. Re-feeding after fasting exhibits hyperphagia and often rebound weight gain. However, the mechanisms underlying the hyperphagia and rebound remain elusive. Here we show that 24 h food restriction (24 h FR) and milder 50% FR, both depress synaptic transmission in the hypothalamic paraventricular nucleus (PVN) and induce acute hyperphagia in rats. 24 h FR is followed by weight rebound but 50% FR is not. Orexigenic neuropeptide Y (NPY) via the Y1 receptor (Y1R) inhibited the miniature excitatory postsynaptic current (mEPSC) on anorexigenic oxytocin neurons in the PVN. 24 h FR and 50% FR activated this neuronal pathway to induce acute hyperphagia on Days 1-3 and Days 1-2 after FR, respectively. 24 h FR induced large mEPSC depression, recurrent hyperphagia on Days 9-12 and rebound weight gain on Days 12-17, whereas 50% FR induced moderate mEPSC depression and sustained weight reduction. Transverse data analysis on Day 1 after 24 h FR and 50% FR demonstrated saturation kinetics for the mEPSC depression-hyperphagiacurve, implying hysteresis. The results reveal FR-driven synaptic plasticity in the NPY-Y1R-oxytocin neurocircuit that drives acute hyperphagia. FR with the intensity that regulates the synapse-feeding relay without hysteresis is the key for successful dieting.
Collapse
Affiliation(s)
- Lei Wang
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shigetomo Suyama
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Samantha A. Lee
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yutaka Seino
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka, Japan
| | - Geoffrey W. G. Sharp
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
41
|
Ma X, Li L, Li Z, Huang Z, Yang Y, Liu P, Guo D, Li Y, Wu T, Luo R, Xu J, Ye W, Jiang B, Shi L. eEF2 in the prefrontal cortex promotes excitatory synaptic transmission and social novelty behavior. EMBO Rep 2022; 23:e54543. [PMID: 35993189 PMCID: PMC9535807 DOI: 10.15252/embr.202154543] [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: 12/29/2021] [Revised: 07/22/2022] [Accepted: 08/03/2022] [Indexed: 08/24/2023] Open
Abstract
Regulation of mRNA translation is essential for brain development and function. Translation elongation factor eEF2 acts as a molecular hub orchestrating various synaptic signals to protein synthesis control and participates in hippocampus-dependent cognitive functions. However, whether eEF2 regulates other behaviors in different brain regions has been unknown. Here, we construct a line of Eef2 heterozygous (HET) mice, which show a reduction in eEF2 and protein synthesis mainly in excitatory neurons of the prefrontal cortex. The mice also show lower spine density, reduced excitability, and AMPAR-mediated synaptic transmission in pyramidal neurons of the medial prefrontal cortex (mPFC). While HET mice exhibit normal learning and memory, they show defective social behavior and elevated anxiety. Knockdown of Eef2 in excitatory neurons of the mPFC specifically is sufficient to impair social novelty preference. Either chemogenetic activation of excitatory neurons in the mPFC or mPFC local infusion of the AMPAR potentiator PF-4778574 corrects the social novelty deficit of HET mice. Collectively, we identify a novel role for eEF2 in promoting prefrontal AMPAR-mediated synaptic transmission underlying social novelty behavior.
Collapse
Affiliation(s)
- Xuanyue Ma
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Liuren Li
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Ziming Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Zhengyi Huang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Yaorong Yang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Peng Liu
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Daji Guo
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Clinical Neuroscience InstituteThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yueyao Li
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Tianying Wu
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Ruixiang Luo
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Junyu Xu
- Department of Neurobiology and Department of Rehabilitation of the Children's HospitalZhejiang University School of MedicineHangzhouChina
| | - Wen‐Cai Ye
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Center for Bioactive Natural Molecules and Innovative Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Bin Jiang
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Lei Shi
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Center for Bioactive Natural Molecules and Innovative Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
| |
Collapse
|
42
|
Rigney N, de Vries GJ, Petrulis A, Young LJ. Oxytocin, Vasopressin, and Social Behavior: From Neural Circuits to Clinical Opportunities. Endocrinology 2022; 163:bqac111. [PMID: 35863332 PMCID: PMC9337272 DOI: 10.1210/endocr/bqac111] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Indexed: 11/19/2022]
Abstract
Oxytocin and vasopressin are peptide hormones secreted from the pituitary that are well known for their peripheral endocrine effects on childbirth/nursing and blood pressure/urine concentration, respectively. However, both peptides are also released in the brain, where they modulate several aspects of social behaviors. Oxytocin promotes maternal nurturing and bonding, enhances social reward, and increases the salience of social stimuli. Vasopressin modulates social communication, social investigation, territorial behavior, and aggression, predominantly in males. Both peptides facilitate social memory and pair bonding behaviors in monogamous species. Here we review the latest research delineating the neural circuitry of the brain oxytocin and vasopressin systems and summarize recent investigations into the circuit-based mechanisms modulating social behaviors. We highlight research using modern molecular genetic technologies to map, monitor activity of, or manipulate neuropeptide circuits. Species diversity in oxytocin and vasopressin effects on social behaviors are also discussed. We conclude with a discussion of the translational implications of oxytocin and vasopressin for improving social functioning in disorders with social impairments, such as autism spectrum disorder.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Geert J de Vries
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA
| | - Aras Petrulis
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Emory University, Atlanta, Georgia 30329, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
43
|
Advances in the identification and validation of autism biomarkers. Nat Rev Drug Discov 2022; 21:697-698. [PMID: 36008547 DOI: 10.1038/d41573-022-00141-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Takayama K, Tobori S, Andoh C, Kakae M, Hagiwara M, Nagayasu K, Shirakawa H, Ago Y, Kaneko S. Autism Spectrum Disorder Model Mice Induced by Prenatal Exposure to Valproic Acid Exhibit Enhanced Empathy-Like Behavior <i>via</i> Oxytocinergic Signaling. Biol Pharm Bull 2022; 45:1124-1132. [DOI: 10.1248/bpb.b22-00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kaito Takayama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shota Tobori
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Chihiro Andoh
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Masashi Kakae
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Masako Hagiwara
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
45
|
Zhao F, Zhang H, Wang P, Cui W, Xu K, Chen D, Hu M, Li Z, Geng X, Wei S. Oxytocin and serotonin in the modulation of neural function: Neurobiological underpinnings of autism-related behavior. Front Neurosci 2022; 16:919890. [PMID: 35937893 PMCID: PMC9354980 DOI: 10.3389/fnins.2022.919890] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorders (ASD) is a group of generalized neurodevelopmental disorders. Its main clinical features are social communication disorder and repetitive stereotyped behavioral interest. The abnormal structure and function of brain network is the basis of social dysfunction and stereotyped performance in patients with autism spectrum disorder. The number of patients diagnosed with ASD has increased year by year, but there is a lack of effective intervention and treatment. Oxytocin has been revealed to effectively improve social cognitive function and significantly improve the social information processing ability, empathy ability and social communication ability of ASD patients. The change of serotonin level also been reported affecting the development of brain and causes ASD-like behavioral abnormalities, such as anxiety, depression like behavior, stereotyped behavior. Present review will focus on the research progress of serotonin and oxytocin in the pathogenesis, brain circuit changes and treatment of autism. Revealing the regulatory effect and neural mechanism of serotonin and oxytocin on patients with ASD is not only conducive to a deeper comprehension of the pathogenesis of ASD, but also has vital clinical significance.
Collapse
Affiliation(s)
- Feng Zhao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjie Cui
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kaiyong Xu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghui Hu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zifa Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- Zifa Li,
| | - Xiwen Geng
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- Xiwen Geng,
| | - Sheng Wei
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Sheng Wei,
| |
Collapse
|
46
|
Ma Z, Eaton M, Liu Y, Zhang J, Chen X, Tu X, Shi Y, Que Z, Wettschurack K, Zhang Z, Shi R, Chen Y, Kimbrough A, Lanman NA, Schust L, Huang Z, Yang Y. Deficiency of autism-related Scn2a gene in mice disrupts sleep patterns and circadian rhythms. Neurobiol Dis 2022; 168:105690. [PMID: 35301122 PMCID: PMC9018617 DOI: 10.1016/j.nbd.2022.105690] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/21/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.
Collapse
Affiliation(s)
- Zhixiong Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Yushuang Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xiaoling Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yiqiang Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Kyle Wettschurack
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Zaiyang Zhang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Nadia A Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Leah Schust
- FamilieSCN2A Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
47
|
Pohl TT, Hörnberg H. Neuroligins in neurodevelopmental conditions: how mouse models of de novo mutations can help us link synaptic function to social behavior. Neuronal Signal 2022; 6:NS20210030. [PMID: 35601025 PMCID: PMC9093077 DOI: 10.1042/ns20210030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 11/19/2022] Open
Abstract
Neurodevelopmental conditions (or neurodevelopmental disorders, NDDs) are highly heterogeneous with overlapping characteristics and shared genetic etiology. The large symptom variability and etiological heterogeneity have made it challenging to understand the biological mechanisms underpinning NDDs. To accommodate this individual variability, one approach is to move away from diagnostic criteria and focus on distinct dimensions with relevance to multiple NDDs. This domain approach is well suited to preclinical research, where genetically modified animal models can be used to link genetic variability to neurobiological mechanisms and behavioral traits. Genetic factors associated with NDDs can be grouped functionally into common biological pathways, with one prominent functional group being genes associated with the synapse. These include the neuroligins (Nlgns), a family of postsynaptic transmembrane proteins that are key modulators of synaptic function. Here, we review how research using Nlgn mouse models has provided insight into how synaptic proteins contribute to behavioral traits associated with NDDs. We focus on how mutations in different Nlgns affect social behaviors, as differences in social interaction and communication are a common feature of most NDDs. Importantly, mice carrying distinct mutations in Nlgns share some neurobiological and behavioral phenotypes with other synaptic gene mutations. Comparing the functional implications of mutations in multiple synaptic proteins is a first step towards identifying convergent neurobiological pathways in multiple brain regions and circuits.
Collapse
Affiliation(s)
- Tobias T. Pohl
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Hanna Hörnberg
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| |
Collapse
|
48
|
Li H, Wang X, Hu C, Li H, Xu Z, Lei P, Luo X, Hao Y. JUN and PDGFRA as Crucial Candidate Genes for Childhood Autism Spectrum Disorder. Front Neuroinform 2022; 16:800079. [PMID: 35655651 PMCID: PMC9152672 DOI: 10.3389/fninf.2022.800079] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/19/2022] [Indexed: 01/11/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder, characterized by marked genetic heterogeneity. In this study, two independent microarray datasets of cerebellum of ASD were integrative analyzed by NetworkAnalyst to screen candidate crucial genes. NetworkAnalyst identified two up-regulated genes, Jun proto-oncogene (JUN) and platelet derived growth factor receptor alpha (PDGFRA), as the most crucial genes in cerebellum of ASD patients. Based on KEGG pathway database, genes associated with JUN in the cerebellum highlight the pathways of Th17 cell differentiation and Th1 and Th2 cell differentiation. Genes associated with PDGFRA in the cerebellum were found enriched in pathways in EGFR tyrosine kinase inhibitor resistance and Rap1 signaling pathway. Analyzing all differentially expressed genes (DEGs) from the two datasets, Gene Set Enrichment Analysis (GSEA) brought out IL17 signaling pathway, which is related to the expression of JUN and PDGFRA. The ImmuCellAI found the elevated expression of JUN and PDGFRA correlating with increased Th17 and monocytes suggests JUN and PDGFRA may regulate Th17 cell activation and monocytes infiltrating. Mice model of maternal immune activation demonstrated that JUN and PDGFRA are up-regulated and related to the ASD-like behaviors that provide insights into the molecular mechanisms underlying the altered IL17 signaling pathway in ASD and may enable novel therapeutic strategies.
Collapse
Affiliation(s)
- Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyuan Wang
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoshuo Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yan Hao
| |
Collapse
|
49
|
Gonçalves C, Kareklas K, Teles MC, Varela SAM, Costa J, Leite RB, Paixão T, Oliveira RF. Phenotypic architecture of sociality and its associated genetic polymorphisms in zebrafish. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12809. [PMID: 35524578 PMCID: PMC9744564 DOI: 10.1111/gbb.12809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 12/11/2022]
Abstract
Sociality relies on motivational and cognitive components that may have evolved independently, or may have been linked by phenotypic correlations driven by a shared selective pressure for increased social competence. Furthermore, these components may be domain-specific or of general-domain across social and non-social contexts. Here, we used zebrafish to test if the motivational and cognitive components of social behavior are phenotypically linked and if they are domain specific or of general domain. The behavioral phenotyping of zebrafish in social and equivalent non-social tests shows that the motivational (preference) and cognitive (memory) components of sociality: (1) are independent from each other, hence not supporting the occurrence of a sociality syndrome; and (2) are phenotypically linked to non-social traits, forming two general behavioral modules, suggesting that sociality traits have been co-opted from general-domain motivational and cognitive traits. Moreover, the study of the association between single nucleotide polymorphisms (SNPs) and each behavioral module further supports this view, since several SNPs from a list of candidate "social" genes, are statistically associated with the motivational, but not with the cognitive, behavioral module. Together, these results support the occurrence of general-domain motivational and cognitive behavioral modules in zebrafish, which have been co-opted for the social domain.
Collapse
Affiliation(s)
- Claúdia Gonçalves
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Kyriacos Kareklas
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Magda C. Teles
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Susana A. M. Varela
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - João Costa
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Ricardo B. Leite
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Tiago Paixão
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal
| | - Rui F. Oliveira
- Integrative Behavioural Biology LaboratoryGulbenkian Institute of ScienceOeirasPortugal,Department of BiosciencesISPA‐Instituto UniversitárioLisbonPortugal,Champalimaud Neuroscience Program, Champalimaud FoundationLisbonPortugal
| |
Collapse
|
50
|
Musardo S, Contestabile A, Knoop M, Baud O, Bellone C. Oxytocin neurons mediate the effect of social isolation via the VTA circuits. eLife 2022; 11:73421. [PMID: 35451958 PMCID: PMC9075949 DOI: 10.7554/elife.73421] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Social interaction during adolescence strongly influences brain function and behavior, and the recent pandemic has emphasized the devastating effect of social distancing on mental health. While accumulating evidence has shown the importance of the reward system in encoding specific aspects of social interaction, the consequences of social isolation on the reward system and the development of social skills later in adulthood are still largely unknown. Here, we found that one week of social isolation during adolescence in male mice increased social interaction at the expense of social habituation and social novelty preference. Behavioral changes were accompanied by the acute hyperexcitability of putative dopamine (pDA) neurons in the ventral tegmental area (VTA) and long-lasting expression of GluA2-lacking AMPARs at excitatory inputs onto pDA neurons that project to the prefrontal cortex (PFC). Social isolation-dependent behavioral deficits and changes in neural activity and synaptic plasticity were reversed by chemogenetic inhibition of oxytocin neurons in the paraventricular nucleus (PVN) of the hypothalamus. These results demonstrate that social isolation in male mice has acute and long-lasting effects on social interaction and suggest that homeostatic adaptations mediate these effects within the reward circuit.
Collapse
Affiliation(s)
- Stefano Musardo
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | | | - Marit Knoop
- Laboratory of Child Growth and Development, University of Geneva, Geneva, Switzerland
| | - Olivier Baud
- Laboratory of Child Growth and Development, University of Geneva, Geneva, Switzerland
| | - Camilla Bellone
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| |
Collapse
|