1
|
Chan SY, Low XZ, Ngoh ZM, Ong ZY, Kee MZL, Huang P, Kumar S, Rifkin-Graboi A, Chong YS, Chen H, Tan KH, Chan JKY, Fortier MV, Gluckman PD, Zhou JH, Meaney MJ, Tan AP. Neonatal Nucleus Accumbens Microstructure Modulates Individual Susceptibility to Preconception Maternal Stress in Relation to Externalizing Behaviors. J Am Acad Child Adolesc Psychiatry 2024; 63:1035-1046. [PMID: 38423282 DOI: 10.1016/j.jaac.2023.12.011] [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: 03/16/2023] [Revised: 11/20/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Maternal stress influences in utero brain development and is a modifiable risk factor for offspring psychopathologies. Reward circuitry dysfunction underlies various internalizing and externalizing psychopathologies. This study examined (1) the association between maternal stress and microstructural characteristics of the neonatal nucleus accumbens (NAcc), a major node of the reward circuitry, and (2) whether neonatal NAcc microstructure modulates individual susceptibility to maternal stress in relation to childhood behavioral problems. METHOD K-means longitudinal cluster analysis was performed to determine trajectories of maternal stress measures (Perceived Stress Scale [PSS], hair cortisol) from preconception to the third trimester. Neonatal NAcc microstructural measures (orientation density index [ODI] and intracellular volume fraction [ICVF]) were compared across trajectories. We then examined the interaction between maternal stress and neonatal NAcc microstructure on child internalizing and externalizing behaviors, assessed between ages 3 and 4 years. RESULTS Two trajectories of maternal stress magnitude ("low"/"high") were identified for both PSS (n = 287) and hair cortisol (n = 336). Right neonatal NAcc ODI (rNAcc-ODI) was significantly lower in "low" relative to "high" PSS trajectories (n = 77, p = .04). PSS at preconception had the strongest association with rNAcc-ODI (r = 0.293, p = .029). No differences in NAcc microstructure were found between hair cortisol trajectories. A significant interaction between preconception PSS and rNAcc-ODI on externalizing behavior was observed (n = 47, p = .047). CONCLUSION Our study showed that the preconception period contributes to in utero NAcc development, and that NAcc microstructure modulates individual susceptibility to preconception maternal stress in relation to externalizing problems. PLAIN LANGUAGE SUMMARY In the S-PRESTO population-based cohort study conducted in Singapore with 351 women and their children, higher levels of maternal perceived stress within the year before pregnancy were associated with increased dendritic complexity within offsprings' nucleus accumbens, indicative of a more advanced developmental profile. Variations in right nucleus accumbens microstructure significantly modulated the association between maternal perceived stress at preconception and externalizing behaviors in early childhood. Study findings suggest that maternal stress in the preconception period accelerates in-utero nucleus accumbens development, leading to differential risk to externalizing problems in later childhood.
Collapse
Affiliation(s)
- Shi Yu Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Xi Zhen Low
- National University Health System, Singapore, Singapore
| | - Zhen Ming Ngoh
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Zi Yan Ong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Michelle Z L Kee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Pei Huang
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | | | - Anne Rifkin-Graboi
- National Institute of Education, Nanyang Technological University, Singapore, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; National University Health System, Singapore, Singapore; National University of Singapore, Singapore, Singapore
| | - Helen Chen
- KK Women's and Children's Hospital, Duke-National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- KK Women's and Children's Hospital, Duke-National University of Singapore, Singapore, Singapore
| | - Jerry K Y Chan
- KK Women's and Children's Hospital, Duke-National University of Singapore, Singapore, Singapore
| | - Marielle V Fortier
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; KK Women's and Children's Hospital, Duke-National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Juan H Zhou
- National University of Singapore, Singapore, Singapore
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; National University of Singapore, Singapore, Singapore; Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Ai Peng Tan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; National University Health System, Singapore, Singapore; National University of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
Asad Z, Fakheir Y, Abukhaled Y, Khalil R. Implications of altered pyramidal cell morphology on clinical symptoms of neurodevelopmental disorders. Eur J Neurosci 2024; 60:4877-4892. [PMID: 39054743 DOI: 10.1111/ejn.16484] [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: 02/13/2024] [Revised: 05/26/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
The prevalence of pyramidal cells (PCs) in the mammalian cerebral cortex underscore their value as they play a crucial role in various brain functions, ranging from cognition, sensory processing, to motor output. PC morphology significantly influences brain connectivity and plays a critical role in maintaining normal brain function. Pathological alterations to PC morphology are thought to contribute to the aetiology of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. This review explores the relationship between abnormalities in PC morphology in key cortical areas and the clinical manifestations in schizophrenia and ASD. We focus largely on human postmortem studies and provide evidence that dendritic segment length, complexity and spine density are differentially affected in these disorders. These morphological alterations can lead to disruptions in cortical connectivity, potentially contributing to the cognitive and behavioural deficits observed in these disorders. Furthermore, we highlight the importance of investigating the functional and structural characteristics of PCs in these disorders to illuminate the underlying pathogenesis and stimulate further research in this area.
Collapse
Affiliation(s)
- Zummar Asad
- School of Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Yara Fakheir
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Yara Abukhaled
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Reem Khalil
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
3
|
Anitei M, Bruno F, Valkova C, Dau T, Cirri E, Mestres I, Calegari F, Kaether C. IER3IP1-mutations cause microcephaly by selective inhibition of ER-Golgi transport. Cell Mol Life Sci 2024; 81:334. [PMID: 39115595 PMCID: PMC11335259 DOI: 10.1007/s00018-024-05386-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/13/2024] [Accepted: 07/27/2024] [Indexed: 08/22/2024]
Abstract
Mutations in the IER3IP1 (Immediate Early Response-3 Interacting Protein 1) gene can give rise to MEDS1 (Microcephaly with Simplified Gyral Pattern, Epilepsy, and Permanent Neonatal Diabetes Syndrome-1), a severe condition leading to early childhood mortality. The small endoplasmic reticulum (ER)-membrane protein IER3IP1 plays a non-essential role in ER-Golgi transport. Here, we employed secretome and cell-surface proteomics to demonstrate that the absence of IER3IP1 results in the mistrafficking of proteins crucial for neuronal development and survival, including FGFR3, UNC5B and SEMA4D. This phenomenon correlates with the distension of ER membranes and increased lysosomal activity. Notably, the trafficking of cargo receptor ERGIC53 and KDEL-receptor 2 are compromised, with the latter leading to the anomalous secretion of ER-localized chaperones. Our investigation extended to in-utero knock-down of Ier3ip1 in mouse embryo brains, revealing a morphological phenotype in newborn neurons. In summary, our findings provide insights into how the loss or mutation of a 10 kDa small ER-membrane protein can cause a fatal syndrome.
Collapse
Affiliation(s)
- Mihaela Anitei
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Francesca Bruno
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Christina Valkova
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Therese Dau
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Emilio Cirri
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Iván Mestres
- Center for Regenerative Therapies, TU-Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Federico Calegari
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany
| | - Christoph Kaether
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Beutenbergstr 11, 07745, Jena, Germany.
- Center for Regenerative Therapies, TU-Dresden, Fetscherstraße 105, 01307, Dresden, Germany.
| |
Collapse
|
4
|
Speranza L, Molinari M, Volpicelli F, Lacivita E, Leopoldo M, Pulcrano S, Carlo Bellenchi G, Perrone Capano C, Crispino M. Modulation of neuronal morphology by antipsychotic drug: Involvement of serotonin receptor 7. Brain Res 2024; 1830:148815. [PMID: 38387714 DOI: 10.1016/j.brainres.2024.148815] [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: 12/07/2023] [Revised: 01/26/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Antipsychotic drugs (APDs) are the primary pharmacological treatment for schizophrenia, a complex disorder characterized by altered neuronal connectivity. Atypical or second-generation antipsychotics, such as Risperidone (RSP) and Clozapine (CZP) predominantly block dopaminergic D2 and serotonin receptor 2A (5-HT2A) neurotransmission. Both compounds also exhibit affinity for the 5-HT7R, with RSP acting as an antagonist and CZP as an inverse agonist. Our study aimed to determine whether RSP and CZP can influence neuronal morphology through a 5-HT7R-mediated mechanism. Here, we demonstrated that CZP promotes neurite outgrowth of early postnatal cortical neurons, and the 5-HT7R mediates its effect. Conversely, RSP leads to a reduction of neurite length of early postnatal cortical neurons, in a 5-HT7R-independent way. Furthermore, we found that the effects of CZP, mediated by 5-HT7R activation, require the participation of ERK and Cdk5 kinase pathways. At the same time, the modulation of neurite length by RSP does not involve these pathways. In conclusion, our findings provide valuable insights into the morphological changes induced by these two APDs in neurons and elucidate some of the associated molecular pathways. Investigating the 5-HT7R-dependent signaling pathways underlying the neuronal morphogenic effects of APDs may contribute to the identification of novel targets for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Marta Molinari
- Telethon Institute of Genetics & Medicine, Via Campi Flegrei, 34, 80078 Pozzuoli, Naples, Italy
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy.
| | - Enza Lacivita
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, 70125 Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, 70125 Bari, Italy
| | - Salvatore Pulcrano
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", CNR, 80131 Naples, Italy
| | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", CNR, 80131 Naples, Italy
| | - Carla Perrone Capano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
5
|
Shao Y, Cai Y, Chen T, Hao K, Luo B, Wang X, Guo W, Su X, Lv L, Yang Y, Li W. Impaired erythropoietin-producing hepatocellular B receptors signaling in the prefrontal cortex and hippocampus following maternal immune activation in male rats. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12863. [PMID: 37575018 PMCID: PMC10733575 DOI: 10.1111/gbb.12863] [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: 03/07/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
An environmental risk factor for schizophrenia (SZ) is maternal infection, which exerts longstanding effects on the neurodevelopment of offspring. Accumulating evidence suggests that synaptic disturbances may contribute to the pathology of the disease, but the underlying molecular mechanisms remain poorly understood. Erythropoietin-producing hepatocellular B (EphB) receptor signaling plays an important role in synaptic plasticity by regulating the formation and maturation of dendritic spines and regulating excitatory neurotransmission. We examined whether EphB receptors and downstream associated proteins are susceptible to environmental risk factors implicated in the etiology of synaptic disturbances in SZ. Using an established rodent model, which closely imitates the characteristics of SZ, we observed the behavioral performance and synaptic structure of male offspring in adolescence and early adulthood. We then analyzed the expression of EphB receptors and associated proteins in the prefrontal cortex and hippocampus. Maternal immune activation offspring showed significantly progressive cognitive impairment and pre-pulse inhibition deficits together with an increase in the expression of EphB2 receptors and NMDA receptor subunits. We also found changes in EphB receptor downstream signaling, in particular, a decrease in phospho-cofilin levels which may explain the reduced dendritic spine density. Besides, we found that the AMPA glutamate, another glutamate ionic receptor associated with cofilin, decreased significantly in maternal immune activation offspring. Thus, alterations in EphB signaling induced by immune activation during pregnancy may underlie disruptions in synaptic plasticity and function in the prefrontal cortex and hippocampus associated with behavioral and cognitive impairment. These findings may provide insight into the mechanisms underlying SZ.
Collapse
Affiliation(s)
- Yiqian Shao
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Yaqi Cai
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Tengfei Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Keke Hao
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Binbin Luo
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Xiujuan Wang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Weiyun Guo
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Stem Cell and Biological Treatment Engineering Research Center of Henan, College of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Xi Su
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Yongfeng Yang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
6
|
Wu X, Liu L, Xue X, Li X, Zhao K, Zhang J, Li W, Yao W, Ding S, Jia C, Zhu F. Captive ERVWE1 triggers impairment of 5-HT neuronal plasticity in the first-episode schizophrenia by post-transcriptional activation of HTR1B in ALKBH5-m6A dependent epigenetic mechanisms. Cell Biosci 2023; 13:213. [PMID: 37990254 PMCID: PMC10664518 DOI: 10.1186/s13578-023-01167-4] [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: 07/27/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Abnormalities in the 5-HT system and synaptic plasticity are hallmark features of schizophrenia. Previous studies suggest that the human endogenous retrovirus W family envelope (ERVWE1) is an influential risk factor for schizophrenia and inversely correlates with 5-HT4 receptor in schizophrenia. To our knowledge, no data describes the effect of ERVWE1 on 5-HT neuronal plasticity. N6-methyladenosine (m6A) regulates gene expression and impacts synaptic plasticity. Our research aims to systematically investigate the effects of ERVWE1 on 5-HT neuronal plasticity through m6A modification in schizophrenia. RESULTS HTR1B, ALKBH5, and Arc exhibited higher levels in individuals with first-episode schizophrenia compared to the controls and showed a strong positive correlation with ERVWE1. Interestingly, HTR1B was also correlated with ALKBH5 and Arc. Further analyses confirmed that ALKBH5 may be an independent risk factor for schizophrenia. In vitro studies, we discovered that ERVWE1 enhanced HTR1B expression, thereby activating the ERK-ELK1-Arc pathway and reducing the complexity and spine density of 5-HT neurons. Furthermore, ERVWE1 reduced m6A levels through ALKBH5 demethylation. ERVWE1 induced HTR1B upregulation by improving its mRNA stability in ALKBH5-m6A-dependent epigenetic mechanisms. Importantly, ALKBH5 mediated the observed alterations in 5-HT neuronal plasticity induced by ERVWE1. CONCLUSIONS Overall, HTR1B, Arc, and ALKBH5 levels were increased in schizophrenia and positively associated with ERVWE1. Moreover, ALKBH5 was a novel risk gene for schizophrenia. ERVWE1 impaired 5-HT neuronal plasticity in ALKBH5-m6A dependent mechanism by the HTR1B-ERK-ELK1-Arc pathway, which may be an important contributor to aberrant synaptic plasticity in schizophrenia.
Collapse
Affiliation(s)
- Xiulin Wu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | | | - Xing Xue
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuhang Li
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Kexin Zhao
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiahang Zhang
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenshi Li
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei Yao
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuang Ding
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Chen Jia
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Tat L, Cannizzaro N, Schaaf Z, Racherla S, Bottiglieri T, Green R, Zarbalis KS. Prenatal folic acid and vitamin B 12 imbalance alter neuronal morphology and synaptic density in the mouse neocortex. Commun Biol 2023; 6:1133. [PMID: 37938221 PMCID: PMC10632462 DOI: 10.1038/s42003-023-05492-9] [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: 01/30/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023] Open
Abstract
Previous reports have provided evidence that insufficient or excessive maternal folic acid (FA) intake during pregnancy can alter neurodevelopment of the offspring by modulating prenatal neurogenesis. Furthermore, our earlier work in a mouse model confirmed long-term structural changes at the cellular level of either deficient or excessive FA supply by comparably reducing dendritic arborization of cortical projection neurons. Here, we report that excessive amounts of FA decrease arborization of deep layer projection neurons, but not upper layer neurons and that reduced complexity of deep layer neurons is not observed when folic acid is replaced by folinic acid, a stable reduced form of folate. In addition, deficiency of B12, a vitamin that critically regulates folate metabolism, causes even more marked decreases in neuronal arborization in both deep and upper layer neurons and particularly in combination with FA excess. Furthermore, both FA excess and B12 deficiency affect synaptic density and morphology. Our findings point to neurodevelopmental risks associated with insufficient amounts of prenatal B12, particularly in association with high levels of FA intake, suggesting that the neurodevelopmental program is sensitive to an imbalance in the status of these interacting micronutrients.
Collapse
Affiliation(s)
- Lyvin Tat
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Noemi Cannizzaro
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Zachary Schaaf
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Shailaja Racherla
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Teodoro Bottiglieri
- Baylor Scott & White Research Institute, Center of Metabolomics, 3434 Live Oak, Dallas, TX, 75204, USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| | - Konstantinos S Zarbalis
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
- MIND Institute, University of California, Davis, CA, 95817, USA.
| |
Collapse
|
8
|
Velásquez MM, Lattig MC, Chitiva LC, Costa GM, Sutachan JJ, Albarracin SL. Dendritogenic Potential of the Ethanol Extract from Lippia alba Leaves in Rat Cortical Neurons. Molecules 2023; 28:6666. [PMID: 37764442 PMCID: PMC10537230 DOI: 10.3390/molecules28186666] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
A reduced dendritic complexity, especially in regions such as the hippocampus and the prefrontal cortex, has been linked to the pathophysiology of some neuropsychiatric disorders, in which synaptic plasticity and functions such as emotional and cognitive processing are compromised. For this reason, the identification of new therapeutic strategies would be enriched by the search for metabolites that promote structural plasticity. The present study evaluated the dendritogenic potential of the ethanol extract of Lippia alba, an aromatic plant rich in flavonoids and terpenes, which has been widely used in traditional medicine for its presumed analgesic, anxiolytic, and antidepressant potential. An in vitro model of rat cortical neurons was used to determine the kinetics of the plant's effect at different time intervals. Changes in morphological parameters of the neurons were determined, as well as the dendritic complexity, by Sholl analysis. The extract promotes the outgrowth of dendritic branching in a rapid and sustained fashion, without being cytotoxic to the cells. We found that this effect could be mediated by the phosphatidylinositol 3-kinase pathway, which is involved in mechanisms of neuronal plasticity, differentiation, and survival. The evidence presented in this study provides a basis for further research that, through in vivo models, can delve into the plant's therapeutic potential.
Collapse
Affiliation(s)
- María Marcela Velásquez
- Instituto de Genética Humana, Facultad de Medicina, Pontifica Universidad Javeriana, Bogotá 110911, Colombia
| | | | - Luis Carlos Chitiva
- Departamento de Química, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Geison M. Costa
- Departamento de Química, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Jhon Jairo Sutachan
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Sonia Luz Albarracin
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| |
Collapse
|
9
|
Li BZ, Sumera A, Booker SA, McCullagh EA. Current Best Practices for Analysis of Dendritic Spine Morphology and Number in Neurodevelopmental Disorder Research. ACS Chem Neurosci 2023; 14:1561-1572. [PMID: 37070364 PMCID: PMC10161226 DOI: 10.1021/acschemneuro.3c00062] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
Quantitative methods for assessing neural anatomy have rapidly evolved in neuroscience and provide important insights into brain health and function. However, as new techniques develop, it is not always clear when and how each may be used to answer specific scientific questions posed. Dendritic spines, which are often indicative of synapse formation and neural plasticity, have been implicated across many brain regions in neurodevelopmental disorders as a marker for neural changes reflecting neural dysfunction or alterations. In this Perspective we highlight several techniques for staining, imaging, and quantifying dendritic spines as well as provide a framework for avoiding potential issues related to pseudoreplication. This framework illustrates how others may apply the most rigorous approaches. We consider the cost-benefit analysis of the varied techniques, recognizing that the most sophisticated equipment may not always be necessary for answering some research questions. Together, we hope this piece will help researchers determine the best strategy toward using the ever-growing number of techniques available to determine neural changes underlying dendritic spine morphology in health and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ben-Zheng Li
- Department
of Physiology and Biophysics, University
of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Anna Sumera
- Simons
Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Sam A Booker
- Simons
Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Elizabeth A. McCullagh
- Department
of Integrative Biology, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
10
|
Casellas-Vidal D, Mademont-Soler I, Sánchez J, Plaja A, Castells N, Camós M, Nieto-Moragas J, Del Mar García M, Rodriguez-Solera C, Rivera H, Brunet J, Álvarez S, Perapoch J, Queralt X, Obón M. ZDHHC15 as a candidate gene for autism spectrum disorder. Am J Med Genet A 2023; 191:941-947. [PMID: 36565021 DOI: 10.1002/ajmg.a.63099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
The phenotypic repercussion of ZDHHC15 haploinsufficiency is not well-known. This gene was initially suggested as a candidate for X-linked mental retardation, but such an association was later questioned. We studied a multiplex family with three members with autism spectrum disorder (ASD) by array CGH, karyotype, exome sequencing and X-chromosome inactivation patterns. Medical history interviews, cognitive and physical examinations, and sensory profiling were also assessed. The three family members with ASD (with normal cognitive abilities and an abnormal sensory profile) were the only carriers of a 1.7 Mb deletion in the long arm of chromosome X, involving: ZDHHC15, MAGEE2, PBDC1, MAGEE1, MIR384 and MIR325. The normal chromosome X was preferentially inactivated in female carriers, and the whole exome sequencing of an affected family member did not reveal any additional genetic variant that could explain the phenotype. Thus, in the present family, ASD segregates with a deletion on chromosome X that includes ZDHHC15. Considering our results together with gene data (regarding function, expression, conservation and animal/cellular models), ZDHHC15 is a candidate gene for ASD. Emerging evidence also suggests that this gene could be associated with other neurodevelopmental disorders, with incomplete penetrance and variable expressivity.
Collapse
Affiliation(s)
| | - Irene Mademont-Soler
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | - Joana Sánchez
- Centre de Salut Mental Infantil i Juvenil, Institut d'Assistència Sanitària, Girona, Spain
| | - Alberto Plaja
- Unitat d'Arrays, Departament de Genètica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Neus Castells
- Unitat d'Arrays, Departament de Genètica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Maria Camós
- Servei de Pediatria, Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | - Javier Nieto-Moragas
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | | | | | - Helena Rivera
- Centre de Salut Mental Infantil i Juvenil, Institut d'Assistència Sanitària, Girona, Spain
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology, Hospital Universitari Doctor Josep Trueta, IDIBGI, Girona, Spain
| | - Sara Álvarez
- Servicio de Diagnóstico Genético, NIMGenetics, Madrid, Spain
| | - Josep Perapoch
- Servei de Pediatria, Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | - Xavier Queralt
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | - María Obón
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| |
Collapse
|
11
|
Wang Z, Niu M, Zheng N, Meng J, Jiang Y, Yang D, Yao P, Yao T, Luo H, Xu H, Ge Y, Zhang YW, Zhang X. Increased level of RAB39B leads to neuronal dysfunction and behavioural changes in mice. J Cell Mol Med 2023; 27:1214-1226. [PMID: 36977207 PMCID: PMC10148058 DOI: 10.1111/jcmm.17704] [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: 03/31/2022] [Revised: 01/22/2023] [Accepted: 02/22/2023] [Indexed: 03/30/2023] Open
Abstract
Duplications of the Xq28 region are a common cause of X-linked intellectual disability (XLID). The RAB39B gene locates in Xq28 and has been implicated in disease pathogenesis. However, whether increased dosage of RAB39B leads to cognitive impairment and synaptic dysfunction remains elusive. Herein, we overexpressed RAB39B in mouse brain by injecting AAVs into bilateral ventricles of neonatal animals. We found that at 2 months of age, neuronal overexpression of RAB39B impaired the recognition memory and the short-term working memory in mice and resulted in certain autism-like behaviours, including social novelty defect and repetitive grooming behaviour in female mice. Moreover, overexpression of RAB39B decreased dendritic arborization of primary neurons in vitro and reduced synaptic transmission in female mice. Neuronal overexpression of RAB39B also altered autophagy without affecting levels and PSD distribution of synaptic proteins. Our results demonstrate that overexpression of RAB39B compromises normal neuronal development, thereby resulting in dysfunctional synaptic transmission and certain intellectual disability and behavioural abnormalities in mice. These findings identify a molecular mechanism underlying XLID with increased copy numbers of Xq28 and provide potential strategies for disease intervention.
Collapse
Affiliation(s)
- Zijie Wang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
- Department of Neurosurgery, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Mengxi Niu
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Naizhen Zheng
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Jian Meng
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Yiru Jiang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Dingting Yang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Peijie Yao
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Tingting Yao
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Hong Luo
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Huaxi Xu
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Yunlong Ge
- Department of Neurosurgery, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yun-Wu Zhang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| | - Xian Zhang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
da Costa Souza F, Grodzki ACG, Morgan RK, Zhang Z, Taha AY, Lein PJ. Oxidized linoleic acid metabolites regulate neuronal morphogenesis in vitro. Neurochem Int 2023; 164:105506. [PMID: 36758902 PMCID: PMC10495953 DOI: 10.1016/j.neuint.2023.105506] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Linoleic acid (LA, 18:2n-6) is an essential nutrient for optimal infant growth and brain development. The effects of LA in the brain are thought to be mediated by oxygenated metabolites of LA known as oxidized LA metabolites (OXLAMs), but evidence is lacking to directly support this hypothesis. This study investigated whether OXLAMs modulate key neurodevelopmental processes including axon outgrowth, dendritic arborization, cell viability and synaptic connectivity. Primary cortical neuron-glia co-cultures from postnatal day 0-1 male and female rats were exposed for 48h to the following OXLAMs: 1) 13-hydroxyoctadecadienoic acid (13-HODE); 2) 9-hydroxyoctadecadienoic acid (9-HODE); 3) 9,10-dihydroxyoctadecenoic acid (9,10-DiHOME); 4) 12(13)-epoxyoctadecenoic acid (12(13)-EpOME); 5) 9,10,13-trihydroxyoctadecenoic acid (9,10,13-TriHOME); 6) 9-oxo-octadecadienoic acid (9-OxoODE); and 7) 12,13-dihydroxyoctadecenoic acid (12,13-DiHOME). Axonal outgrowth, evaluated by Tau-1 immunostaining, was increased by 9-HODE, but decreased by 12,13-DiHOME in male but not female neurons. Dendrite arborization, evaluated by MAP2B-eGFP expression, was affected by 9-HODE, 9-OxoODE, and 12(13)-EpOME in male neurons and, by 12(13)-EpOME in female neurons. Neither cell viability nor synaptic connectivity were significantly altered by OXLAMs. Overall, this study shows select OXLAMs modulate neuron morphology in a sex-dependent manner, with male neurons being more susceptible.
Collapse
Affiliation(s)
- Felipe da Costa Souza
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA; Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Ana Cristina G Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Rhianna K Morgan
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Zhichao Zhang
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA.
| | - Ameer Y Taha
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA.
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
13
|
Li Q, Xu X, Qian Y, Cai H, Zhao W, Zhu J, Yu Y. Resting-state brain functional alterations and their genetic mechanisms in drug-naive first-episode psychosis. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:13. [PMID: 36841861 PMCID: PMC9968350 DOI: 10.1038/s41537-023-00338-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/27/2023]
Abstract
Extensive research has established the presence of resting-state brain functional damage in psychosis. However, the genetic mechanisms of such disease phenotype are yet to be unveiled. We investigated resting-state brain functional alterations in patients with drug-naive first-episode psychosis (DFP) by performing a neuroimaging meta-analysis of 8 original studies comprising 500 patients and 469 controls. Combined with the Allen Human Brain Atlas, we further conducted transcriptome-neuroimaging spatial correlations to identify genes whose expression levels were linked to brain functional alterations in DFP, followed by a range of gene functional characteristic analyses. Meta-analysis revealed a mixture of increased and decreased brain function in widespread areas including the default-mode, visual, motor, striatal, and cerebellar systems in DFP. Moreover, these brain functional alterations were spatially associated with the expression of 1662 genes, which were enriched for molecular functions, cellular components, and biological processes of the cerebral cortex, as well as psychiatric disorders including schizophrenia. Specific expression analyses demonstrated that these genes were specifically expressed in the brain tissue, in cortical neurons and immune cells, and during nearly all developmental periods. Concurrently, the genes could construct a protein-protein interaction network supported by hub genes and were linked to multiple behavioral domains including emotion, attention, perception, and motor. Our findings provide empirical evidence for the notion that brain functional damage in DFP involves a complex interaction of polygenes with various functional characteristics.
Collapse
Affiliation(s)
- Qian Li
- grid.459419.4Department of Radiology, Chaohu Hospital of Anhui Medical University, 238000 Hefei, China ,grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Xiaotao Xu
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Yinfeng Qian
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Huanhuan Cai
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Wenming Zhao
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China. .,Research Center of Clinical Medical Imaging, Anhui Province, 230032, Hefei, China. .,Anhui Provincial Institute of Translational Medicine, 230032, Hefei, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China. .,Research Center of Clinical Medical Imaging, Anhui Province, 230032, Hefei, China. .,Anhui Provincial Institute of Translational Medicine, 230032, Hefei, China.
| |
Collapse
|
14
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [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] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Maternal preconception circulating blood biomarker mixtures, child behavioural symptom scores and the potential mediating role of neonatal brain microstructure: the S-PRESTO cohort. Transl Psychiatry 2023; 13:38. [PMID: 36737601 PMCID: PMC9898508 DOI: 10.1038/s41398-023-02332-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Human brain development starts in the embryonic period. Maternal preconception nutrition and nutrient availability to the embryo may influence brain development at this critical period following conception and early cellular differentiation, thereby affecting offspring neurodevelopmental and behavioural disorder risk. However, studying this is challenging due to difficulties in characterizing preconception nutritional status and few studies have objective neurodevelopmental imaging measures in children. We investigated the associations of maternal preconception circulating blood nutrient-related biomarker mixtures (~15 weeks before conception) with child behavioural symptoms (Child Behaviour Checklist (CBCL), aged 3 years) within the Singapore Preconception Study of Long-Term Maternal and Child Outcomes (S-PRESTO) study. The CBCL preschool form evaluates child behaviours based on syndrome scales and Diagnostic and Statistical Manual of Mental Disorders (DSM) oriented scales. These scales consist of internalizing problems, externalizing problems, anxiety problems, pervasive developmental problems, oppositional defiant, etc. We applied data-driven clustering and a method for modelling mixtures (Bayesian kernel machine regression, BKMR) to account for complex, non-linear dependencies between 67 biomarkers. We used effect decomposition analyses to explore the potential mediating role of neonatal (week 1) brain microstructure, specifically orientation dispersion indices (ODI) of 49 cortical and subcortical grey matter regions. We found that higher levels of a nutrient cluster including thiamine, thiamine monophosphate (TMP), pyridoxal phosphate, pyridoxic acid, and pyridoxal were associated with a higher CBCL score for internalizing problems (posterior inclusion probability (PIP) = 0.768). Specifically, thiamine independently influenced CBCL (Conditional PIP = 0.775). Higher maternal preconception thiamine level was also associated with a lower right subthalamic nucleus ODI (P-value = 0.01) while a lower right subthalamic nucleus ODI was associated with higher CBCL scores for multiple domains (P-value < 0.05). One potential mechanism is the suboptimal metabolism of free thiamine to active vitamin B1, but additional follow-up and replication studies in other cohorts are needed.
Collapse
|
16
|
Relationship of neurite architecture to brain activity during task-based fMRI. Neuroimage 2022; 262:119575. [PMID: 35987489 DOI: 10.1016/j.neuroimage.2022.119575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
Functional MRI (fMRI) has been widely used to examine changes in neuronal activity during cognitive tasks. Commonly used measures of gray matter macrostructure (e.g., cortical thickness, surface area, volume) do not consistently appear to serve as structural correlates of brain function. In contrast, gray matter microstructure, measured using neurite orientation dispersion and density imaging (NODDI), enables the estimation of indices of neurite density (neurite density index; NDI) and organization (orientation dispersion index; ODI) in gray matter. Our study explored the relationship among neurite architecture, BOLD (blood-oxygen-level-dependent) fMRI, and cognition, using a large sample (n = 750) of young adults of the human connectome project (HCP) and two tasks that index more cortical (working memory) and more subcortical (emotion processing) targeting of brain functions. Using NODDI, fMRI, structural MRI and task performance data, hierarchical regression analyses revealed that higher working memory- and emotion processing-evoked BOLD activity was related to lower ODI in the right DLPFC, and lower ODI and NDI values in the right and left amygdala, respectively. Common measures of brain macrostructure (i.e., DLPFC thickness/surface area and amygdala volume) did not explain any additional variance (beyond neurite architecture) in BOLD activity. A moderating effect of neurite architecture on the relationship between emotion processing task-evoked BOLD response and performance was observed. Our findings provide evidence that neuro-/social-affective cognition-related BOLD activity is partially driven by the local neurite organization and density with direct impact on emotion processing. In vivo gray matter microstructure represents a new target of investigation providing strong potential for clinical translation.
Collapse
|
17
|
Chaudry S, Vasudevan N. mTOR-Dependent Spine Dynamics in Autism. Front Mol Neurosci 2022; 15:877609. [PMID: 35782388 PMCID: PMC9241970 DOI: 10.3389/fnmol.2022.877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
Collapse
|
18
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
19
|
Guzmán Salas S, Weber A, Malci A, Lin X, Herrera-Molina R, Cerpa W, Dorador C, Signorelli J, Zamorano P. The metabolite p-cresol impairs dendritic development, synaptogenesis and synapse function in hippocampal neurons: Implications for autism spectrum disorder. J Neurochem 2022; 161:335-349. [PMID: 35257373 DOI: 10.1111/jnc.15604] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogenous neurodevelopment disorder resulting from different etiological factors, both genetic and/or environmental. These factors can lead to abnormal neuronal development on dendrite and synaptic function at the central nervous system. Recent studies have shown that a subset of ASD patients display increased circulation levels of the tyrosine metabolite, p-cresol, related to chronic intestinal disorders due to dysbiosis of the intestinal microbiota. In particular, abnormal presence of intestinal Clostridium sp. has been linked to high levels of p-cresol in ASD children younger than 8 years. However, the role of p-cresol during development of the central nervous system is unknown. Here, we evaluated in vitro the effect of p-cresol on neurite outgrowth in N2a and PC12 cell lines and dendritic morphology, synaptic density, neuronal activity, and calcium responses in primary rat hippocampal neurons. p-cresol inhibits neural differentiation and neurites outgrowth in N2a and PC12 neuronal cell lines. In hippocampal neuronal cultures, Sholl´s analysis shows a decrease in the dendritic arborization of neurons treated with p-cresol. Synaptic density analyzed with the synaptic markers Piccolo and Shank2 is diminished in hippocampal neurons treated with p-cresol. Electrically-evoked intracellular calcium rise was drastically, but reversely, blocked by p-cresol, whereas that spontaneous neuronal activity was severely affected by early addition of the metabolite. These findings show that p-cresol alters dendrite development, synaptogenesis and synapse function of neurons in culture, therefore, neuronal alterations occurring in ASD children may be related to this metabolite and dysbiosis of the intestinal microbiota.
Collapse
Affiliation(s)
- Sheyla Guzmán Salas
- Departamento Biomédico, Universidad de Antofagasta, Antofagasta, Chile.,Centre for Biotechnology and Bioengineering CeBiB, Antofagasta
| | - André Weber
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ayse Malci
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Xiao Lin
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Rodrigo Herrera-Molina
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile.,Center for Behavioral Brain Sciences and Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA); Universidad de Magallanes, Punta Arenas, Chile.,Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Dorador
- Centre for Biotechnology and Bioengineering CeBiB, Antofagasta.,Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | | | - Pedro Zamorano
- Departamento Biomédico, Universidad de Antofagasta, Antofagasta, Chile.,Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
20
|
Liu L, Liu TT, Xie GG, Zhu XQ, Wang Y. Ubiquitin ligase TRIM32 promotes dendrite arborization by mediating degradation of the epigenetic factor CDYL. FASEB J 2021; 36:e22087. [PMID: 34888944 DOI: 10.1096/fj.202100031rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 11/11/2022]
Abstract
Proper dendritic morphology is fundamental to nerve signal transmission; thus, revealing the mechanism by which dendrite arborization is regulated is of great significance. Our previous studies have found that the epigenetic molecule chromodomain Y-like (CDYL) negatively regulates dendritic branching. Current research mostly focuses on the processes downstream of CDYL, whereas the upstream regulatory process has not been investigated to date. In this study, we identified an upstream regulator of CDYL, the E3 ubiquitin ligase tripartite motif-containing protein 32 (TRIM32), which promotes dendrite arborization by mediating the ubiquitylation and degradation of CDYL. By using mass spectrometry and biochemistry strategies, we proved that TRIM32 interacted with CDYL and mediated CDYL ubiquitylation modification in vivo and in vitro. Overexpressing TRIM32 decreased the protein level of CDYL, leading to an increase in the dendritic complexity of primary cultured rat neurons. In contrast, knocking down TRIM32 increased the protein level of CDYL and decreased the dendritic complexity. The truncated form of TRIM32 without E3 ligase activity (ΔRING) lost its ability to regulate dendritic complexity. Most importantly, knockdown of CDYL abolished the reduced complexity of dendrites caused by TRIM32 knockdown, indicating that the TRIM32-mediated regulation of dendritic development depends on its regulation of downstream CDYL. Hence, our findings reveal that TRIM32 could promote dendrite arborization by mediating CDYL degradation. This work initially defines a novel biological role of TRIM32 in regulating mechanisms upstream of CDYL and further presents a potential therapeutic target for the treatment of CDYL-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lei Liu
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Ting-Ting Liu
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Guo-Guang Xie
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Xiao-Qi Zhu
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yun Wang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
21
|
Bahry JA, Fedder-Semmes KN, Sceniak MP, Sabo SL. An Autism-Associated de novo Mutation in GluN2B Destabilizes Growing Dendrites by Promoting Retraction and Pruning. Front Cell Neurosci 2021; 15:692232. [PMID: 34393725 PMCID: PMC8363002 DOI: 10.3389/fncel.2021.692232] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in GRIN2B, which encodes the GluN2B subunit of NMDA receptors, lead to autism spectrum disorders (ASD), but the pathophysiological mechanisms remain unclear. Recently, we showed that a GluN2B variant that is associated with severe ASD (GluN2B724t) impairs dendrite morphogenesis. To determine which aspects of dendrite growth are affected by GluN2B724t, we investigated the dynamics of dendrite growth and branching in rat neocortical neurons using time-lapse imaging. GluN2B724t expression shifted branch motility toward retraction and away from extension. GluN2B724t and wild-type neurons formed new branches at similar rates, but mutant neurons exhibited increased pruning of dendritic branches. The observed changes in dynamics resulted in nearly complete elimination of the net expansion of arbor size and complexity that is normally observed during this developmental period. These data demonstrate that ASD-associated mutant GluN2B interferes with dendrite morphogenesis by reducing rates of outgrowth while promoting retraction and subsequent pruning. Because mutant dendrites remain motile and capable of growth, it is possible that reducing pruning or promoting dendrite stabilization could overcome dendrite arbor defects associated with GRIN2B mutations.
Collapse
Affiliation(s)
- Jacob A Bahry
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States.,Graduate Program in Biochemistry, Cell and Molecular Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Karlie N Fedder-Semmes
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
| | - Michael P Sceniak
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Shasta L Sabo
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States.,Graduate Program in Biochemistry, Cell and Molecular Biology, Central Michigan University, Mount Pleasant, MI, United States.,Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States
| |
Collapse
|
22
|
Histone Deacetylase Inhibitors Ameliorate Morphological Defects and Hypoexcitability of iPSC-Neurons from Rubinstein-Taybi Patients. Int J Mol Sci 2021; 22:ijms22115777. [PMID: 34071322 PMCID: PMC8197986 DOI: 10.3390/ijms22115777] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022] Open
Abstract
Rubinstein-Taybi syndrome (RSTS) is a rare neurodevelopmental disorder caused by mutations in CREBBP or EP300 genes encoding CBP/p300 lysine acetyltransferases. We investigated the efficacy of the histone deacetylase inhibitor (HDACi) Trichostatin A (TSA) in ameliorating morphological abnormalities of iPSC-derived young neurons from P149 and P34 CREBBP-mutated patients and hypoexcitability of mature neurons from P149. Neural progenitors from both patients’ iPSC lines were cultured one week with TSA 20 nM and, only P149, for 6 weeks with TSA 0.2 nM, in parallel to neural progenitors from controls. Immunofluorescence of MAP2/TUJ1 positive cells using the Skeletonize Image J plugin evidenced that TSA partially rescued reduced nuclear area, and decreased branch length and abnormal end points number of both 45 days patients’ neurons, but did not influence the diminished percentage of their neurons with respect to controls. Patch clamp recordings of TSA-treated post-mitotic P149 neurons showed complete/partial rescue of sodium/potassium currents and significant enhancement of neuron excitability compared to untreated replicas. Correction of abnormalities of P149 young neurons was also affected by valproic acid 1 mM for 72 h, with some variation, with respect to TSA, on the morphological parameter. These findings hold promise for development of an epigenetic therapy to attenuate RSTS patients cognitive impairment.
Collapse
|
23
|
Latchney SE, Majewska AK. Persistent organic pollutants at the synapse: Shared phenotypes and converging mechanisms of developmental neurotoxicity. Dev Neurobiol 2021; 81:623-652. [PMID: 33851516 DOI: 10.1002/dneu.22825] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022]
Abstract
The developing nervous system is sensitive to environmental and physiological perturbations in part due to its protracted period of prenatal and postnatal development. Epidemiological and experimental studies link developmental exposures to persistent organic pollutants (POPs) including polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, polybrominated diphenyl ethers, and benzo(a)pyrene to increased risk for neurodevelopmental disorders in children. Mechanistic studies reveal that many of the complex cellular processes that occur during sensitive periods of rapid brain development are cellular targets for developmental neurotoxicants. One area of research interest has focused on synapse formation and plasticity, processes that involve the growth and retraction of dendrites and dendritic spines. For each chemical discussed in this review, we summarize the morphological and electrophysiological data that provide evidence that developmental POP exposure produces long-lasting effects on dendritic morphology, spine formation, glutamatergic and GABAergic signaling systems, and synaptic transmission. We also discuss shared intracellular mechanisms, with a focus on calcium and thyroid hormone homeostasis, by which these chemicals act to modify synapses. We conclude our review highlighting research gaps that merit consideration when characterizing synaptic pathology elicited by chemical exposure. These gaps include low-dose and nonmonotonic dose-response effects, the temporal relationship between dendritic growth, spine formation, and synaptic activity, excitation-inhibition balance, hormonal effects, and the need for more studies in females to identify sex differences. By identifying converging pathological mechanisms elicited by POP exposure at the synapse, we can define future research directions that will advance our understanding of these chemicals on synapse structure and function.
Collapse
Affiliation(s)
- Sarah E Latchney
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA.,Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA.,Center for Visual Science, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Disrupted Excitatory Synaptic Contacts and Altered Neuronal Network Activity Underpins the Neurological Phenotype in PCDH19-Clustering Epilepsy (PCDH19-CE). Mol Neurobiol 2021; 58:2005-2018. [PMID: 33411240 DOI: 10.1007/s12035-020-02242-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
PCDH19-Clustering Epilepsy (PCDH19-CE) is an infantile onset disorder caused by mutation of the X-linked PCDH19 gene. Intriguingly, heterozygous females are affected while hemizygous males are not. While there is compelling evidence that this disorder stems from the coexistence of WT and PCDH19-null cells, the cellular mechanism underpinning the neurological phenotype remains unclear. Here, we investigate the impact of Pcdh19 WT and KO neuron mosaicism on synaptogenesis and network activity. Using our previously established knock-in and knock-out mouse models, together with CRISPR-Cas9 genome editing technology, we demonstrate a reduction in excitatory synaptic contacts between PCDH19-expressing and PCDH19-null neurons. Significantly altered neuronal morphology and neuronal network activities were also identified in the mixed populations. In addition, we show that in Pcdh19 heterozygous mice, where the coexistence of WT and KO neurons naturally occurs, aberrant contralateral axonal branching is present. Overall, our data show that mosaic expression of PCDH19 disrupts physiological neurite communication leading to abnormal neuronal activity, a hallmark of PCDH19-CE.
Collapse
|
25
|
Ferreira Castro A, Baltruschat L, Stürner T, Bahrami A, Jedlicka P, Tavosanis G, Cuntz H. Achieving functional neuronal dendrite structure through sequential stochastic growth and retraction. eLife 2020; 9:e60920. [PMID: 33241995 PMCID: PMC7837678 DOI: 10.7554/elife.60920] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
Class I ventral posterior dendritic arborisation (c1vpda) proprioceptive sensory neurons respond to contractions in the Drosophila larval body wall during crawling. Their dendritic branches run along the direction of contraction, possibly a functional requirement to maximise membrane curvature during crawling contractions. Although the molecular machinery of dendritic patterning in c1vpda has been extensively studied, the process leading to the precise elaboration of their comb-like shapes remains elusive. Here, to link dendrite shape with its proprioceptive role, we performed long-term, non-invasive, in vivo time-lapse imaging of c1vpda embryonic and larval morphogenesis to reveal a sequence of differentiation stages. We combined computer models and dendritic branch dynamics tracking to propose that distinct sequential phases of stochastic growth and retraction achieve efficient dendritic trees both in terms of wire and function. Our study shows how dendrite growth balances structure-function requirements, shedding new light on general principles of self-organisation in functionally specialised dendrites.
Collapse
Affiliation(s)
- André Ferreira Castro
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Ernst Strüngmann Institute (ESI) for Neuroscience in cooperation with Max Planck SocietyFrankfurt am MainGermany
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | | | - Tomke Stürner
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | | | - Peter Jedlicka
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Faculty of Medicine, ICAR3R – Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University GiessenGiessenGermany
- Neuroscience Center, Institute of Clinical Neuroanatomy, Goethe UniversityFrankfurt am MainGermany
| | - Gaia Tavosanis
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
- LIMES Institute, University of BonnBonnGermany
| | - Hermann Cuntz
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Ernst Strüngmann Institute (ESI) for Neuroscience in cooperation with Max Planck SocietyFrankfurt am MainGermany
| |
Collapse
|
26
|
Polychlorinated Biphenyls (PCBs): Risk Factors for Autism Spectrum Disorder? TOXICS 2020; 8:toxics8030070. [PMID: 32957475 PMCID: PMC7560399 DOI: 10.3390/toxics8030070] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders defined clinically by core deficits in social reciprocity and communication, restrictive interests and repetitive behaviors. ASD affects one in 54 children in the United States, one in 89 children in Europe, and one in 277 children in Asia, with an estimated worldwide prevalence of 1-2%. While there is increasing consensus that ASD results from complex gene x environment interactions, the identity of specific environmental risk factors and the mechanisms by which environmental and genetic factors interact to determine individual risk remain critical gaps in our understanding of ASD etiology. Polychlorinated biphenyls (PCBs) are ubiquitous environmental contaminants that have been linked to altered neurodevelopment in humans. Preclinical studies demonstrate that PCBs modulate signaling pathways implicated in ASD and phenocopy the effects of ASD risk genes on critical morphometric determinants of neuronal connectivity, such as dendritic arborization. Here, we review human and experimental evidence identifying PCBs as potential risk factors for ASD and discuss the potential for PCBs to influence not only core symptoms of ASD, but also comorbidities commonly associated with ASD, via effects on the central and peripheral nervous systems, and/or peripheral target tissues, using bladder dysfunction as an example. We also discuss critical data gaps in the literature implicating PCBs as ASD risk factors. Unlike genetic factors, which are currently irreversible, environmental factors are modifiable risks. Therefore, data confirming PCBs as risk factors for ASD may suggest rational approaches for the primary prevention of ASD in genetically susceptible individuals.
Collapse
|
27
|
Jin H, Kim B. Neurite Branching Regulated by Neuronal Cell Surface Molecules in Caenorhabditis elegans. Front Neuroanat 2020; 14:59. [PMID: 32973467 PMCID: PMC7471659 DOI: 10.3389/fnana.2020.00059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/04/2020] [Indexed: 01/02/2023] Open
Abstract
The high synaptic density in the nervous system results from the ability of neurites to branch. Neuronal cell surface molecules play central roles during neurite branch formation. The underlying mechanisms of surface molecule activity have often been elucidated using invertebrates with simple nervous systems. Here, we review recent advances in understanding the molecular mechanisms of neurite branching in the nematode Caenorhabditis elegans. We discuss how cell surface receptor complexes link to and modulate actin dynamics to regulate dendritic and axonal branch formation. The mechanisms of neurite branching are often coupled with other neural circuit developmental processes, such as synapse formation and axon guidance, via the same cell-cell surface molecular interactions. We also cover ectopic and sex-specific neurite branching in C. elegans in an attempt to illustrate the importance of these studies in contributing to our understanding of conserved cell surface molecule regulation of neurite branch formation.
Collapse
Affiliation(s)
- HoYong Jin
- Department of Life Science, Dongguk University-Seoul, Goyang, South Korea
| | - Byunghyuk Kim
- Department of Life Science, Dongguk University-Seoul, Goyang, South Korea
| |
Collapse
|
28
|
Bird AD, Cuntz H. Dissecting Sholl Analysis into Its Functional Components. Cell Rep 2020; 27:3081-3096.e5. [PMID: 31167149 DOI: 10.1016/j.celrep.2019.04.097] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/20/2018] [Accepted: 04/19/2019] [Indexed: 12/31/2022] Open
Abstract
Sholl analysis has been an important technique in dendritic anatomy for more than 60 years. The Sholl intersection profile is obtained by counting the number of dendritic branches at a given distance from the soma and is a key measure of dendritic complexity; it has applications from evaluating the changes in structure induced by pathologies to estimating the expected number of anatomical synaptic contacts. We find that the Sholl intersection profiles of most neurons can be reproduced from three basic, functional measures: the domain spanned by the dendritic arbor, the total length of the dendrite, and the angular distribution of how far dendritic segments deviate from a direct path to the soma (i.e., the root angle distribution). The first two measures are determined by axon location and hence microcircuit structure; the third arises from optimal wiring and represents a branching statistic estimating the need for conduction speed in a neuron.
Collapse
Affiliation(s)
- Alex D Bird
- Frankfurt Institute for Advanced Studies, Frankfurt-am-Main 60438, Germany; Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt-am-Main 60528, Germany.
| | - Hermann Cuntz
- Frankfurt Institute for Advanced Studies, Frankfurt-am-Main 60438, Germany; Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt-am-Main 60528, Germany
| |
Collapse
|
29
|
Saxena R, Babadi M, Namvarhaghighi H, Roullet FI. Role of environmental factors and epigenetics in autism spectrum disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:35-60. [PMID: 32711816 DOI: 10.1016/bs.pmbts.2020.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder thought to be caused by predisposing high-risk genes that may be altered during the early development by environmental factors. The impact of maternal challenges during pregnancy on the prevalence of ASD has been widely studied in clinical and animal studies. Here, we review some clinical and pre-clinical evidence that links environmental factors (i.e., infection, air pollution, pesticides, valproic acid and folic acid) and the risk of ASD. Additionally, certain prenatal environmental challenges such as the valproate and folate prenatal exposures allow us to study mechanisms possibly linked to the etiology of ASD, for instance the epigenetic processes. These mechanistic pathways are also presented and discussed in this chapter.
Collapse
Affiliation(s)
- Roheeni Saxena
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Melika Babadi
- School of Interdisciplinary Science, McMaster University, Hamilton, ON, Canada
| | | | - Florence I Roullet
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
30
|
A Caenorhabditis elegans Model for Integrating the Functions of Neuropsychiatric Risk Genes Identifies Components Required for Normal Dendritic Morphology. G3-GENES GENOMES GENETICS 2020; 10:1617-1628. [PMID: 32132169 PMCID: PMC7202017 DOI: 10.1534/g3.119.400925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Analysis of patient-derived DNA samples has identified hundreds of variants that are likely involved in neuropsychiatric diseases such as autism spectrum disorder (ASD) and schizophrenia (SCZ). While these studies couple behavioral phenotypes to individual genotypes, the number and diversity of candidate genes implicated in these disorders highlights the fact that the mechanistic underpinnings of these disorders are largely unknown. Here, we describe a RNAi-based screening platform that uses C. elegans to screen candidate neuropsychiatric risk genes (NRGs) for roles in controlling dendritic arborization. To benchmark this approach, we queried published lists of NRGs whose variants in ASD and SCZ are predicted to result in complete or partial loss of gene function. We found that a significant fraction (>16%) of these candidate NRGs are essential for dendritic development. Furthermore, these gene sets are enriched for dendritic arbor phenotypes (>14 fold) when compared to control RNAi datasets of over 500 human orthologs. The diversity of PVD structural abnormalities observed in these assays suggests that the functions of diverse NRGs (encoding transcription factors, chromatin remodelers, molecular chaperones and cytoskeleton-related proteins) converge to regulate neuronal morphology and that individual NRGs may play distinct roles in dendritic branching. We also demonstrate that the experimental value of this platform by providing additional insights into the molecular frameworks of candidate NRGs. Specifically, we show that ANK2/UNC-44 function is directly integrated with known regulators of dendritic arborization and suggest that altering the dosage of ARID1B/LET-526 expression during development affects neuronal morphology without diminishing aspects of cell fate specification.
Collapse
|
31
|
Nazeri A, Schifani C, Anderson JAE, Ameis SH, Voineskos AN. In Vivo Imaging of Gray Matter Microstructure in Major Psychiatric Disorders: Opportunities for Clinical Translation. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 5:855-864. [PMID: 32381477 DOI: 10.1016/j.bpsc.2020.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
Postmortem studies reveal that individuals with major neuropsychiatric disorders such as schizophrenia and autism spectrum disorder have gray matter microstructural abnormalities. These include abnormalities in neuropil organization, expression of proteins supporting neuritic and synaptic integrity, and myelination. Genetic and postmortem studies suggest that these changes may be causally linked to the pathogenesis of these disorders. Advances in diffusion-weighted magnetic resonance image (dMRI) acquisition techniques and biophysical modeling allow for the quantification of gray matter microstructure in vivo. While several biophysical models for imaging microstructural properties are available, one in particular, neurite orientation dispersion and density imaging (NODDI), holds great promise for clinical applications. NODDI can be applied to both gray and white matter and requires only a single extra shell beyond a standard dMRI acquisition. Since its development only a few years ago, the NODDI algorithm has been used to characterize gray matter microstructure in schizophrenia, Alzheimer's disease, healthy aging, and development. These investigations have shown that microstructural findings in vivo, using NODDI, align with postmortem findings. Not only do NODDI and other advanced dMRI-based modeling methods provide a window into the brain previously only available postmortem, but they may be more sensitive to certain brain changes than conventional magnetic resonance imaging approaches. This opens up exciting new possibilities for clinicians to more rapidly detect disease signatures and allows earlier intervention in the course of the disease. Given that neurites and gray matter microstructure have the capacity to rapidly remodel, these novel dMRI-based methods represent an opportunity to noninvasively monitor neuroplastic changes posttherapy within much shorter time scales.
Collapse
Affiliation(s)
- Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Christin Schifani
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - John A E Anderson
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Stephanie H Ameis
- Margaret and Wallace McCain Centre for Child, Youth and Family Mental Health, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Centre for Brain and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Klocke C, Sethi S, Lein PJ. The developmental neurotoxicity of legacy vs. contemporary polychlorinated biphenyls (PCBs): similarities and differences. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:8885-8896. [PMID: 31713823 PMCID: PMC7220795 DOI: 10.1007/s11356-019-06723-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 10/07/2019] [Indexed: 05/11/2023]
Abstract
Although banned from production for decades, PCBs remain a significant risk to human health. A primary target of concern is the developing brain. Epidemiological studies link PCB exposures in utero or during infancy to increased risk of neuropsychiatric deficits in children. Nonclinical studies of legacy congeners found in PCB mixtures synthesized prior to the ban on PCB production suggest that non-dioxin-like (NDL) congeners are predominantly responsible for the developmental neurotoxicity associated with PCB exposures. Mechanistic studies suggest that NDL PCBs alter neurodevelopment via ryanodine receptor-dependent effects on dendritic arborization. Lightly chlorinated congeners, which were not present in the industrial mixtures synthesized prior to the ban on PCB production, have emerged as contemporary environmental contaminants, but there is a paucity of data regarding their potential developmental neurotoxicity. PCB 11, a prevalent contemporary congener, is found in the serum of children and their mothers, as well as in the serum of pregnant women at increased risk for having a child diagnosed with a neurodevelopmental disorder (NDD). Recent data demonstrates that PCB 11 modulates neuronal morphogenesis via mechanisms that are convergent with and divergent from those implicated in the developmental neurotoxicity of legacy NDL PCBs. This review summarizes these data and discusses their relevance to adverse neurodevelopmental outcomes in humans.
Collapse
Affiliation(s)
- Carolyn Klocke
- Department of Molecular Biosciences, University of California, Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, University of California, Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA.
| |
Collapse
|
33
|
In vitro modeling of dendritic atrophy in Rett syndrome: determinants for phenotypic drug screening in neurodevelopmental disorders. Sci Rep 2020; 10:2491. [PMID: 32051524 PMCID: PMC7016139 DOI: 10.1038/s41598-020-59268-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/21/2020] [Indexed: 01/16/2023] Open
Abstract
Dendritic atrophy, defined as the reduction in complexity of the neuronal arborization, is a hallmark of several neurodevelopmental disorders, including Rett Syndrome (RTT). RTT, affecting 1:10,000 girls worldwide, is mainly caused by mutations in the MECP2 gene and has no cure. We describe here an in vitro model of dendritic atrophy in Mecp2−/y mouse hippocampal primary cultures, suitable for phenotypic drug-screening. Using High-Content Imaging techniques, we systematically investigated the impact of culturing determinants on several parameters such as neuronal survival, total dendritic length, dendritic endpoints, soma size, cell clusterization, spontaneous activity. Determinants included cell-seeding density, glass or polystyrene substrates, coating with poly-Ornithine with/without Matrigel and miniaturization from 24 to 96-half surface multiwell plates. We show that in all plate-sizes at densities below 320 cells/mm2, morphological parameters remained constant while spontaneous network activity decreased according to the cell-density. Mecp2−/y neurons cultured at 160 cells/mm2 density in 96 multiwell plates, displayed significant dendritic atrophy and showed a marked increase in dendritic length following treatment with Brain-derived neurotrophic factor (BDNF) or Mirtazapine. In conclusion, we have established a phenotypic assay suitable for fast screening of hundreds of compounds, which may be extended to other neurodevelopmental diseases with dendritic atrophy.
Collapse
|
34
|
Klocke C, Lein PJ. Evidence Implicating Non-Dioxin-Like Congeners as the Key Mediators of Polychlorinated Biphenyl (PCB) Developmental Neurotoxicity. Int J Mol Sci 2020; 21:E1013. [PMID: 32033061 PMCID: PMC7037228 DOI: 10.3390/ijms21031013] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/15/2022] Open
Abstract
Despite being banned from production for decades, polychlorinated biphenyls (PCBs) continue to pose a significant risk to human health. This is due to not only the continued release of legacy PCBs from PCB-containing equipment and materials manufactured prior to the ban on PCB production, but also the inadvertent production of PCBs as byproducts of contemporary pigment and dye production. Evidence from human and animal studies clearly identifies developmental neurotoxicity as a primary endpoint of concern associated with PCB exposures. However, the relative role(s) of specific PCB congeners in mediating the adverse effects of PCBs on the developing nervous system, and the mechanism(s) by which PCBs disrupt typical neurodevelopment remain outstanding questions. New questions are also emerging regarding the potential developmental neurotoxicity of lower chlorinated PCBs that were not present in the legacy commercial PCB mixtures, but constitute a significant proportion of contemporary human PCB exposures. Here, we review behavioral and mechanistic data obtained from experimental models as well as recent epidemiological studies that suggest the non-dioxin-like (NDL) PCBs are primarily responsible for the developmental neurotoxicity associated with PCBs. We also discuss emerging data demonstrating the potential for non-legacy, lower chlorinated PCBs to cause adverse neurodevelopmental outcomes. Molecular targets, the relevance of PCB interactions with these targets to neurodevelopmental disorders, and critical data gaps are addressed as well.
Collapse
Affiliation(s)
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA;
| |
Collapse
|
35
|
Pravoverov K, Whiting K, Thapa S, Bushong T, Trang K, Lein PJ, Chandrasekaran V. MicroRNAs are Necessary for BMP-7-induced Dendritic Growth in Cultured Rat Sympathetic Neurons. Cell Mol Neurobiol 2019; 39:917-934. [PMID: 31104181 PMCID: PMC6713596 DOI: 10.1007/s10571-019-00688-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/14/2019] [Indexed: 01/28/2023]
Abstract
Neuronal connectivity is dependent on size and shape of the dendritic arbor. However, mechanisms controlling dendritic arborization, especially in the peripheral nervous system, are not completely understood. Previous studies have shown that bone morphogenetic proteins (BMPs) are important initiators of dendritic growth in peripheral neurons. In this study, we examined the hypothesis that post-transcriptional regulation mediated by microRNAs (miRNAs) is necessary for BMP-7-induced dendritic growth in these neurons. To examine the role of miRNAs in BMP-7-induced dendritic growth, microarray analyses was used to profile miRNA expression in cultured sympathetic neurons from the superior cervical ganglia of embryonic day 21 rat pups at 6 and 24 h after treatment with BMP-7 (50 ng/mL). Our data showed that BMP-7 significantly regulated the expression of 43 of the 762 miRNAs. Of the 43 miRNAs, 22 showed robust gene expression; 14 were upregulated by BMP-7 and 8 were downregulated by BMP-7. The expression profile for miR-335, miR-664-1*, miR-21, and miR-23b was confirmed using qPCR analyses. Functional studies using morphometric analyses of dendritic growth in cultured sympathetic neurons transfected with miRNA mimics and inhibitors indicated that miR-664-1*, miR-23b, and miR-21 regulated early stages of BMP-7-induced dendritic growth. In summary, our data provide evidence for miRNA-mediated post-transcriptional regulation as important downstream component of BMP-7 signaling during early stages of dendritic growth in sympathetic neurons.
Collapse
Affiliation(s)
- Kristina Pravoverov
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Katherine Whiting
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Slesha Thapa
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Trevor Bushong
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Karen Trang
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, 1089 Veterinary Medicine Drive, Davis, Davis, CA 95616
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556.,Corresponding author: Vidya Chandrasekaran, Department of Biology, Saint Mary’s College of California, Moraga, CA 94556.
| |
Collapse
|
36
|
Duman JG, Mulherkar S, Tu YK, Erikson KC, Tzeng CP, Mavratsas VC, Ho TSY, Tolias KF. The adhesion-GPCR BAI1 shapes dendritic arbors via Bcr-mediated RhoA activation causing late growth arrest. eLife 2019; 8:47566. [PMID: 31461398 PMCID: PMC6713510 DOI: 10.7554/elife.47566] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/03/2019] [Indexed: 12/17/2022] Open
Abstract
Dendritic arbor architecture profoundly impacts neuronal connectivity and function, and aberrant dendritic morphology characterizes neuropsychiatric disorders. Here, we identify the adhesion-GPCR BAI1 as an important regulator of dendritic arborization. BAI1 loss from mouse or rat hippocampal neurons causes dendritic hypertrophy, whereas BAI1 overexpression precipitates dendrite retraction. These defects specifically manifest as dendrites transition from growth to stability. BAI1-mediated growth arrest is independent of its Rac1-dependent synaptogenic function. Instead, BAI1 couples to the small GTPase RhoA, driving late RhoA activation in dendrites coincident with growth arrest. BAI1 loss lowers RhoA activation and uncouples it from dendrite dynamics, causing overgrowth. None of BAI1's known downstream effectors mediates BAI1-dependent growth arrest. Rather, BAI1 associates with the Rho-GTPase regulatory protein Bcr late in development and stimulates its cryptic RhoA-GEF activity, which functions together with its Rac1-GAP activity to terminate arborization. Our results reveal a late-acting signaling pathway mediating a key transition in dendrite development.
Collapse
Affiliation(s)
- Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Yen-Kuei Tu
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, United States
| | - Kelly C Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Christopher P Tzeng
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Vasilis C Mavratsas
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Rice University, Houston, United States
| | - Tammy Szu-Yu Ho
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| |
Collapse
|
37
|
Szczurkowska J, Pischedda F, Pinto B, Managò F, Haas CA, Summa M, Bertorelli R, Papaleo F, Schäfer MK, Piccoli G, Cancedda L. NEGR1 and FGFR2 cooperatively regulate cortical development and core behaviours related to autism disorders in mice. Brain 2019; 141:2772-2794. [PMID: 30059965 PMCID: PMC6113639 DOI: 10.1093/brain/awy190] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorders are neurodevelopmental conditions with diverse aetiologies, all characterized by common core symptoms such as impaired social skills and communication, as well as repetitive behaviour. Cell adhesion molecules, receptor tyrosine kinases and associated downstream signalling have been strongly implicated in both neurodevelopment and autism spectrum disorders. We found that downregulation of the cell adhesion molecule NEGR1 or the receptor tyrosine kinase fibroblast growth factor receptor 2 (FGFR2) similarly affects neuronal migration and spine density during mouse cortical development in vivo and results in impaired core behaviours related to autism spectrum disorders. Mechanistically, NEGR1 physically interacts with FGFR2 and modulates FGFR2-dependent extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) signalling by decreasing FGFR2 degradation from the plasma membrane. Accordingly, FGFR2 overexpression rescues all defects due to Negr1 knockdown in vivo. Negr1 knockout mice present phenotypes similar to Negr1-downregulated animals. These data indicate that NEGR1 and FGFR2 cooperatively regulate cortical development and suggest a role for defective NEGR1-FGFR2 complex and convergent downstream ERK and AKT signalling in autism spectrum disorders.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Università degli Studi di Genova, Via Balbi, 5, Genoa, Italy
| | - Francesca Pischedda
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Bio@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Francesca Managò
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Summa
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Rosalia Bertorelli
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Michael K Schäfer
- Department of Anesthesiology and Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Giovanni Piccoli
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| |
Collapse
|
38
|
Liang C, Carrel D, Omelchenko A, Kim H, Patel A, Fanget I, Firestein BL. Cortical Neuron Migration and Dendrite Morphology are Regulated by Carboxypeptidase E. Cereb Cortex 2019; 29:2890-2903. [PMID: 29982499 PMCID: PMC6611459 DOI: 10.1093/cercor/bhy155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 12/28/2022] Open
Abstract
Higher brain function relies on proper development of the cerebral cortex, including correct positioning of neurons and dendrite morphology. Disruptions in these processes may result in various neurocognitive disorders. Mutations in the CPE gene, which encodes carboxypeptidase E (CPE), have been linked to depression and intellectual disability. However, it remains unclear whether CPE is involved in early brain development and in turn contributes to the pathophysiology of neurocognitive disorders. Here, we investigate the effects of CPE knockdown on early brain development and explore the functional significance of the interaction between CPE and its binding partner p150Glued. We demonstrate that CPE is required for cortical neuron migration and dendrite arborization. Furthermore, we show that expression of CPE-C10 redistributes p150Glued from the centrosome and that disruption of CPE interaction with p150Glued leads to abnormal neuronal migration and dendrite morphology, suggesting that a complex between CPE and p150Glued is necessary for proper neurodevelopment.
Collapse
Affiliation(s)
- Chen Liang
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
- Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
| | - Damien Carrel
- Neurophotonics Laboratory, Université Paris Descartes, Sorbonne Paris Cité, Centre National de la Recherche Scientifique UMR 8250, Paris, France
| | - Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 683 Hoes Lane West, USA
| | - Hyuck Kim
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
| | - Aashini Patel
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
| | - Isabelle Fanget
- Neurophotonics Laboratory, Université Paris Descartes, Sorbonne Paris Cité, Centre National de la Recherche Scientifique UMR 8250, Paris, France
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA
| |
Collapse
|
39
|
Abstract
Proper neuronal wiring is central to all bodily functions, sensory perception, cognition, memory, and learning. Establishment of a functional neuronal circuit is a highly regulated and dynamic process involving axonal and dendritic branching and navigation toward appropriate targets and connection partners. This intricate circuitry includes axo-dendritic synapse formation, synaptic connections formed with effector cells, and extensive dendritic arborization that function to receive and transmit mechanical and chemical sensory inputs. Such complexity is primarily achieved by extensive axonal and dendritic branch formation and pruning. Fundamental to neuronal branching are cytoskeletal dynamics and plasma membrane expansion, both of which are regulated via numerous extracellular and intracellular signaling mechanisms and molecules. This review focuses on recent advances in understanding the biology of neuronal branching.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Stephanie Gupton
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, 27599, USA.,Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
40
|
Sethi S, Keil KP, Lein PJ. 3,3'-Dichlorobiphenyl (PCB 11) promotes dendritic arborization in primary rat cortical neurons via a CREB-dependent mechanism. Arch Toxicol 2018; 92:3337-3345. [PMID: 30225637 PMCID: PMC6196112 DOI: 10.1007/s00204-018-2307-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023]
Abstract
PCB 11 (3,3'-dichlorobiphenyl), a contemporary congener produced as a byproduct of current pigment production processes, has recently emerged as a prevalent worldwide pollutant. We recently demonstrated that exposure to PCB 11 increases dendritic arborization in vitro, but the mechanism(s) mediating this effect remain unknown. To address this data gap, primary cortical neuron-glia co-cultures derived from neonatal Sprague-Dawley rats were exposed for 48 h to either vehicle (0.1% DMSO) or PCB 11 at concentrations ranging from 1 fM to 1 nM in the absence or presence of pharmacologic antagonists of established molecular targets of higher chlorinated PCBs. Reporter cell lines were used to test activity of PCB 11 at the aryl hydrocarbon receptor (AhR) and thyroid hormone receptor (THR). PCB 11 lacked activity at the AhR and THR, and antagonism of these receptors had no effect on the dendrite-promoting activity of PCB 11. Pharmacologic antagonism of various calcium channels or treatment with antioxidants also did not alter PCB 11-induced dendritic arborization. In contrast, pharmacologic blockade or shRNA knockdown of cAMP response element-binding protein (CREB) significantly decreased dendritic growth in PCB 11-exposed cultures, suggesting PCB 11 promotes dendritic growth via CREB-mediated mechanisms. Since CREB signaling is crucial for normal neurodevelopment, and perturbations of CREB signaling have been associated with neurodevelopmental disorders, our findings suggest that this contemporary pollutant poses a threat to the developing brain, particularly in individuals with heritable mutations that promote CREB signaling.
Collapse
Affiliation(s)
- Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA.
| |
Collapse
|
41
|
PCB 95 promotes dendritic growth in primary rat hippocampal neurons via mTOR-dependent mechanisms. Arch Toxicol 2018; 92:3163-3173. [PMID: 30132043 PMCID: PMC6162988 DOI: 10.1007/s00204-018-2285-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/06/2018] [Indexed: 01/28/2023]
Abstract
Polychlorinated biphenyls (PCBs), and in particular non-dioxin-like (NDL) congeners, continue to pose a significant risk to the developing nervous system. PCB 95, a prevalent NDL congener in the human chemosphere, promotes dendritic growth in rodent primary neurons by activating calcium-dependent transcriptional mechanisms that normally function to link activity to dendritic growth. Activity-dependent dendritic growth is also mediated by calcium-dependent translational mechanisms involving mechanistic target of rapamycin (mTOR), suggesting that the dendrite-promoting activity of PCB 95 may also involve mTOR signaling. Here, we test this hypothesis using primary neuron-glia co-cultures derived from the hippocampi of postnatal day 0 Sprague Dawley rats. PCB 95 (1 nM) activated mTOR in hippocampal cultures as evidenced by increased phosphorylation of mTOR at ser2448. Pharmacologic inhibition of mTOR signaling using rapamycin (20 nM), FK506 (5 nM), or 4EGI-1 (1 µM), and siRNA knockdown of mTOR, or the mTOR complex binding proteins, raptor or rictor, blocked PCB 95-induced dendritic growth. These data identify mTOR activation as a novel molecular mechanism contributing to the effects of PCB 95 on dendritic arborization. In light of clinical data linking gain-of-function mutations in mTOR signaling to neurodevelopmental disorders, our findings suggest that mTOR signaling may represent a convergence point for gene by environment interactions that confer risk for adverse neurodevelopmental outcomes.
Collapse
|
42
|
Nithianandam V, Chien CT. Actin blobs prefigure dendrite branching sites. J Cell Biol 2018; 217:3731-3746. [PMID: 30042190 PMCID: PMC6168249 DOI: 10.1083/jcb.201711136] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 07/09/2018] [Indexed: 02/08/2023] Open
Abstract
Nithianandam and Chien show via in vivo imaging that a dynamic population of F-actin termed actin blobs propagates bidirectionally in dendrites and stalls at future branching sites. The F-actin–severing protein Tsr/cofilin is a regulator of actin blob dynamics and dendrite branching. The actin cytoskeleton provides structural stability and adaptability to the cell. Neuronal dendrites frequently undergo morphological changes by emanating, elongating, and withdrawing branches. However, the knowledge about actin dynamics in dendrites during these processes is limited. By performing in vivo imaging of F-actin markers, we found that F-actin was highly dynamic and heterogeneously distributed in dendritic shafts with enrichment at terminal dendrites. A dynamic F-actin population that we named actin blobs propagated bidirectionally at an average velocity of 1 µm/min. Interestingly, these actin blobs stalled at sites where new dendrites would branch out in minutes. Overstabilization of F-actin by the G15S mutant abolished actin blobs and dendrite branching. We identified the F-actin–severing protein Tsr/cofilin as a regulator of dynamic actin blobs and branching activity. Hence, actin blob localization at future branching sites represents a dendrite-branching mechanism to account for highly diversified dendritic morphology.
Collapse
Affiliation(s)
- Vanitha Nithianandam
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Cheng-Ting Chien
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan .,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
43
|
Miller GW, Chandrasekaran V, Yaghoobi B, Lein PJ. Opportunities and challenges for using the zebrafish to study neuronal connectivity as an endpoint of developmental neurotoxicity. Neurotoxicology 2018; 67:102-111. [PMID: 29704525 PMCID: PMC6177215 DOI: 10.1016/j.neuro.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 01/28/2023]
Abstract
Chemical exposures have been implicated as environmental risk factors that interact with genetic susceptibilities to influence individual risk for complex neurodevelopmental disorders, including autism spectrum disorder, schizophrenia, attention deficit hyperactivity disorder and intellectual disabilities. Altered patterns of neuronal connectivity represent a convergent mechanism of pathogenesis for these and other neurodevelopmental disorders, and growing evidence suggests that chemicals can interfere with specific signaling pathways that regulate the development of neuronal connections. There is, therefore, a growing interest in developing screening platforms to identify chemicals that alter neuronal connectivity. Cell-cell, cell-matrix interactions and systemic influences are known to be important in defining neuronal connectivity in the developing brain, thus, a systems-based model offers significant advantages over cell-based models for screening chemicals for effects on neuronal connectivity. The embryonic zebrafish represents a vertebrate model amenable to higher throughput chemical screening that has proven useful in characterizing conserved mechanisms of neurodevelopment. Moreover, the zebrafish is readily amenable to gene editing to integrate genetic susceptibilities. Although use of the zebrafish model in toxicity testing has increased in recent years, the diverse tools available for imaging structural differences in the developing zebrafish brain have not been widely applied to studies of the influence of gene by environment interactions on neuronal connectivity in the developing zebrafish brain. Here, we discuss tools available for imaging of neuronal connectivity in the developing zebrafish, review what has been published in this regard, and suggest a path forward for applying this information to developmental neurotoxicity testing.
Collapse
Affiliation(s)
- Galen W. Miller
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary’s College of California, Moraga, CA 94575, USA
| | - Bianca Yaghoobi
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
44
|
Hansen SN, Schou-Pedersen AMV, Lykkesfeldt J, Tveden-Nyborg P. Spatial Memory Dysfunction Induced by Vitamin C Deficiency Is Associated with Changes in Monoaminergic Neurotransmitters and Aberrant Synapse Formation. Antioxidants (Basel) 2018; 7:antiox7070082. [PMID: 29966224 PMCID: PMC6070945 DOI: 10.3390/antiox7070082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/21/2018] [Accepted: 06/27/2018] [Indexed: 01/11/2023] Open
Abstract
Vitamin C (vitC) is important in the developing brain, acting both as an essential antioxidant and as co-factor in the synthesis and metabolism of monoaminergic neurotransmitters. In guinea pigs, vitC deficiency results in increased oxidative stress, reduced hippocampal volume and neuronal numbers, and deficits in spatial memory. This study investigated the effects of 8 weeks of either sufficient (923 mg vitC/kg feed) or deficient (100 mg vitC/kg feed) levels of dietary vitC on hippocampal monoaminergic neurotransmitters and markers of synapse formation in young guinea pigs with spatial memory deficits. Western blotting and high performance liquid chromatography (HPLC) were used to quantify the selected markers. VitC deficiency resulted in significantly reduced protein levels of synaptophysin (p = 0.016) and a decrease in 5-hydroxyindoleacetic acid/5-hydroxytryptamine ratio (p = 0.0093). Protein expression of the N-methyl-d-aspartate receptor subunit 1 and monoamine oxidase A were reduced, albeit not reaching statistical significance (p = 0.0898 and p = 0.067, respectively). Our findings suggest that vitC deficiency induced spatial memory deficits might be mediated by impairments in neurotransmission and synaptic development.
Collapse
Affiliation(s)
- Stine Normann Hansen
- Section for Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Thorvaldensvej 57, Ground Floor, 1870 Frederiksberg C, Denmark.
| | - Anne Marie V Schou-Pedersen
- Section for Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Thorvaldensvej 57, Ground Floor, 1870 Frederiksberg C, Denmark.
| | - Jens Lykkesfeldt
- Section for Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Thorvaldensvej 57, Ground Floor, 1870 Frederiksberg C, Denmark.
| | - Pernille Tveden-Nyborg
- Section for Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Thorvaldensvej 57, Ground Floor, 1870 Frederiksberg C, Denmark.
| |
Collapse
|
45
|
Hansen SN, Jørgensen JMB, Nyengaard JR, Lykkesfeldt J, Tveden-Nyborg P. Early Life Vitamin C Deficiency Does Not Alter Morphology of Hippocampal CA1 Pyramidal Neurons or Markers of Synaptic Plasticity in a Guinea Pig Model. Nutrients 2018; 10:nu10060749. [PMID: 29890692 PMCID: PMC6024653 DOI: 10.3390/nu10060749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 05/28/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
Approximately 15% of the Western world population, including pregnant women and their children, is characterized as vitamin C (vitC) deficient. In guinea pigs, early life vitC deficiency causes spatial memory deficits, decreased hippocampal volume and neuron numbers, in otherwise clinically healthy animals. We hypothesized that vitC deficiency leads to decreased brain-derived neurotrophic factor and synaptic plasticity markers in selected brain areas (frontal cortex, hippocampus and striatum) and cause morphological changes in cornu ammonis 1 pyramidal neurons of the hippocampus either through a direct effect or indirectly by increased oxidative stress. Fifty-seven female guinea pigs were allocated to three groups receiving either 1390, 100 or 0–50 mg vitC/kg feed for 11 weeks. Dietary vitC levels were reflected in the plasma, cortical and adrenal gland levels, however, redox imbalance was only present in the adrenal glands allowing for the investigation of a direct influence of vitC deficiency on the chosen parameters in the brain. Synaptic plasticity markers were not affected in the investigated brain areas and no differences in isolated pyramidal neuron morphology was recorded. Based on our findings, it appears that vitC deficiency may primarily elicit impaired neuronal function through increased levels of oxidative stress.
Collapse
Affiliation(s)
- Stine N Hansen
- Section for Experimental Animals, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| | - Jane M Bjørn Jørgensen
- Section for Experimental Animals, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| | - Jens R Nyengaard
- Section for Experimental Animals, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
- Core Center of Molecular Morphology, Section for Stereology and Microscopy, Centre for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, Noerrebrogade 44, Building 10G, 3rd Floor, 8000 Aarhus, Denmark.
| | - Jens Lykkesfeldt
- Section for Experimental Animals, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| | - Pernille Tveden-Nyborg
- Section for Experimental Animals, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| |
Collapse
|
46
|
Chen H, Streifel KM, Singh V, Yang D, Mangini L, Wulff H, Lein PJ. From the Cover: BDE-47 and BDE-49 Inhibit Axonal Growth in Primary Rat Hippocampal Neuron-Glia Co-Cultures via Ryanodine Receptor-Dependent Mechanisms. Toxicol Sci 2018; 156:375-386. [PMID: 28003438 DOI: 10.1093/toxsci/kfw259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are widespread environmental contaminants associated with adverse neurodevelopmental outcomes in children and preclinical models; however, the mechanisms by which PBDEs cause developmental neurotoxicity remain speculative. The structural similarity between PBDEs and nondioxin-like (NDL) polychlorinated biphenyls (PCBs) suggests shared toxicological properties. Consistent with this, both NDL PCBs and PBDEs have been shown to stabilize ryanodine receptors (RyRs) in the open configuration. NDL PCB effects on RyR activity are causally linked to increased dendritic arborization, but whether PBDEs similarly enhance dendritic growth is not known. In this study, we quantified the effects of individual PBDE congeners on not only dendritic but also axonal growth since both are regulated by RyR-dependent mechanisms, and both are critical determinants of neuronal connectivity. Neuronal-glial co-cultures dissociated from the neonatal rat hippocampus were exposed to BDE-47 or BDE-49 in the culture medium. At concentrations ranging from 20 pM to 2 µM, neither PBDE congener altered dendritic arborization. In contrast, at concentrations ≥ 200 pM, both congeners delayed neuronal polarization resulting in significant inhibition of axonal outgrowth during the first few days in vitro. The axon inhibitory effects of these PBDE congeners occurred independent of cytotoxicity, and were blocked by pharmacological antagonism of RyR or siRNA knockdown of RyR2. These results demonstrate that the molecular and cellular mechanisms by which PBDEs interfere with neurodevelopment overlap with but are distinct from those of NDL PCBs, and suggest that altered patterns of neuronal connectivity may contribute to the developmental neurotoxicity of PBDEs.
Collapse
Affiliation(s)
- Hao Chen
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Karin M Streifel
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Vikrant Singh
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, California 95616
| | - Dongren Yang
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Linley Mangini
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, California 95616
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine
| |
Collapse
|
47
|
Sethi S, Keil KP, Lein PJ. Species and Sex Differences in the Morphogenic Response of Primary Rodent Neurons to 3,3'-Dichlorobiphenyl (PCB 11). TOXICS 2017; 6:toxics6010004. [PMID: 29295518 PMCID: PMC5874777 DOI: 10.3390/toxics6010004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022]
Abstract
PCB 11 is an emerging global pollutant that we recently showed promotes axonal and dendritic growth in primary rat neuronal cell cultures. Here, we address the influence of sex and species on neuronal responses to PCB 11. Neuronal morphology was quantified in sex-specific primary hippocampal and cortical neuron-glia co-cultures derived from neonatal C57BL/6J mice and Sprague Dawley rats exposed for 48 h to vehicle (0.1% DMSO) or PCB 11 at concentrations ranging from 1 fM to 1 nM. Total axonal length was quantified in tau-1 immunoreactive neurons at day in vitro (DIV) 2; dendritic arborization was assessed by Sholl analysis at DIV 9 in neurons transfected with MAP2B-FusRed. In mouse cultures, PCB 11 enhanced dendritic arborization in female, but not male, hippocampal neurons and male, but not female, cortical neurons. In rat cultures, PCB 11 promoted dendritic arborization in male and female hippocampal and cortical neurons. PCB 11 also increased axonal growth in mouse and rat neurons of both sexes and neuronal cell types. These data demonstrate that PCB 11 exerts sex-specific effects on neuronal morphogenesis that vary depending on species, neurite type, and neuronal cell type. These findings have significant implications for risk assessment of this emerging developmental neurotoxicant.
Collapse
Affiliation(s)
- Sunjay Sethi
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| | - Kimberly P Keil
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| |
Collapse
|
48
|
Rai-Bhogal R, Ahmad E, Li H, Crawford DA. Microarray analysis of gene expression in the cyclooxygenase knockout mice - a connection to autism spectrum disorder. Eur J Neurosci 2017; 47:750-766. [PMID: 29161772 DOI: 10.1111/ejn.13781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
The cellular and molecular events that take place during brain development play an important role in governing function of the mature brain. Lipid-signalling molecules such as prostaglandin E2 (PGE2 ) play an important role in healthy brain development. Abnormalities along the COX-PGE2 signalling pathway due to genetic or environmental causes have been linked to autism spectrum disorder (ASD). This study aims to evaluate the effect of altered COX-PGE2 signalling on development and function of the prenatal brain using male mice lacking cyclooxygenase-1 and cyclooxygenase-2 (COX-1-/- and COX-2-/- ) as potential model systems of ASD. Microarray analysis was used to determine global changes in gene expression during embryonic days 16 (E16) and 19 (E19). Gene Ontology: Biological Process (GO:BP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were implemented to identify affected developmental genes and cellular processes. We found that in both knockouts the brain at E16 had nearly twice as many differentially expressed genes, and affected biological pathways containing various ASD-associated genes important in neuronal function. Interestingly, using GeneMANIA and Cytoscape we also show that the ASD-risk genes identified in both COX-1-/- and COX-2-/- models belong to protein-interaction networks important for brain development despite of different cellular localization of these enzymes. Lastly, we identified eight genes that belong to the Wnt signalling pathways exclusively in the COX-2-/- mice at E16. The level of PKA-phosphorylated β-catenin (S552), a major activator of the Wnt pathway, was increased in this model, suggesting crosstalk between the COX-2-PGE2 and Wnt pathways during early brain development. Overall, these results provide further molecular insight into the contribution of the COX-PGE2 pathways to ASD and demonstrate that COX-1-/- and COX-2-/- animals might be suitable new model systems for studying the disorders.
Collapse
Affiliation(s)
- Ravneet Rai-Bhogal
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada
| | - Eizaaz Ahmad
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada
| | - Hongyan Li
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Dorota A Crawford
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
49
|
Ledda F, Paratcha G. Mechanisms regulating dendritic arbor patterning. Cell Mol Life Sci 2017; 74:4511-4537. [PMID: 28735442 PMCID: PMC11107629 DOI: 10.1007/s00018-017-2588-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 06/14/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022]
Abstract
The nervous system is populated by diverse types of neurons, each of which has dendritic trees with strikingly different morphologies. These neuron-specific morphologies determine how dendritic trees integrate thousands of synaptic inputs to generate different firing properties. To ensure proper neuronal function and connectivity, it is necessary that dendrite patterns are precisely controlled and coordinated with synaptic activity. Here, we summarize the molecular and cellular mechanisms that regulate the formation of cell type-specific dendrite patterns during development. We focus on different aspects of vertebrate dendrite patterning that are particularly important in determining the neuronal function; such as the shape, branching, orientation and size of the arbors as well as the development of dendritic spine protrusions that receive excitatory inputs and compartmentalize postsynaptic responses. Additionally, we briefly comment on the implications of aberrant dendritic morphology for nervous system disease.
Collapse
Affiliation(s)
- Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina.
| |
Collapse
|
50
|
Peek SL, Mah KM, Weiner JA. Regulation of neural circuit formation by protocadherins. Cell Mol Life Sci 2017; 74:4133-4157. [PMID: 28631008 PMCID: PMC5643215 DOI: 10.1007/s00018-017-2572-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/20/2022]
Abstract
The protocadherins (Pcdhs), which make up the most diverse group within the cadherin superfamily, were first discovered in the early 1990s. Data implicating the Pcdhs, including ~60 proteins encoded by the tandem Pcdha, Pcdhb, and Pcdhg gene clusters and another ~10 non-clustered Pcdhs, in the regulation of neural development have continually accumulated, with a significant expansion of the field over the past decade. Here, we review the many roles played by clustered and non-clustered Pcdhs in multiple steps important for the formation and function of neural circuits, including dendrite arborization, axon outgrowth and targeting, synaptogenesis, and synapse elimination. We further discuss studies implicating mutation or epigenetic dysregulation of Pcdh genes in a variety of human neurodevelopmental and neurological disorders. With recent structural modeling of Pcdh proteins, the prospects for uncovering molecular mechanisms of Pcdh extracellular and intracellular interactions, and their role in normal and disrupted neural circuit formation, are bright.
Collapse
Affiliation(s)
- Stacey L Peek
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
- Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Kar Men Mah
- Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Joshua A Weiner
- Department of Biology, The University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry, The University of Iowa, 143 Biology Building, Iowa City, IA, 52242, USA.
| |
Collapse
|