1
|
Putman JN, Watson SD, Zhang Z, Khandelwal N, Kulkarni A, Gibson JR, Huber KM. Pre- and Postsynaptic MEF2C Promotes Experience-Dependent, Input-Specific Development of Cortical Layer 4 to Layer 2/3 Excitatory Synapses and Regulates Activity-Dependent Expression of Synaptic Cell Adhesion Molecules. J Neurosci 2024; 44:e0098242024. [PMID: 39317473 PMCID: PMC11551898 DOI: 10.1523/jneurosci.0098-24.2024] [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: 01/12/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Experience- and activity-dependent transcription is a candidate mechanism to mediate development and refinement of specific cortical circuits. Here, we demonstrate that the activity-dependent transcription factor myocyte enhancer factor 2C (MEF2C) is required in both presynaptic layer (L) 4 and postsynaptic L2/3 mouse (male and female) somatosensory (S1) cortical neurons for development of this specific synaptic connection. While postsynaptic deletion of Mef2c weakens L4 synaptic inputs, it has no effect on inputs from local L2/3, contralateral S1, or the ipsilateral frontal/motor cortex. Similarly, homozygous or heterozygous deletion of Mef2c in presynaptic L4 neurons weakens L4 to L2/3 excitatory synaptic inputs by decreasing presynaptic release probability. Postsynaptic MEF2C is specifically required during an early postnatal, experience-dependent, period for L4 to L2/3 synapse function, and expression of transcriptionally active MEF2C (MEF2C-VP16) rescues weak L4 to L2/3 synaptic strength in sensory-deprived mice. Together, these results suggest that experience- and/or activity-dependent transcriptional activation of MEF2C promotes development of L4 to L2/3 synapses. Additionally, MEF2C regulates the expression of many pre- and postsynaptic genes in postnatal cortical neurons. Interestingly, MEF2C was necessary for activity-dependent expression of many presynaptic genes, including those that function in transsynaptic adhesion and neurotransmitter release. This work provides mechanistic insight into the experience-dependent development of specific cortical circuits.
Collapse
Affiliation(s)
- Jennifer N Putman
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Sean D Watson
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Zhe Zhang
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Nitin Khandelwal
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Jay R Gibson
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Kimberly M Huber
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
2
|
Liu Z, Cheng L, Cao W, Shen C, Qiu Y, Li C, Xiong Y, Yang SB, Chen Z, Yin X, Zhang X. Present and future use of exosomes containing proteins and RNAs in neurodegenerative diseases for synaptic function regulation: A comprehensive review. Int J Biol Macromol 2024; 280:135826. [PMID: 39322147 DOI: 10.1016/j.ijbiomac.2024.135826] [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: 07/23/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Neurodegenerative diseases (NDDs) are increasingly prevalent with global aging, demanding effective treatments. Exosomes, which contain biological macromolecules such as RNA (including miRNAs) and proteins like α-synuclein, tau, and amyloid-beta, are gaining attention as innovative therapeutics. This comprehensive review systematically explores the potential roles of exosomes in NDDs, with a particular focus on their role in synaptic dysfunction. We present the synaptic pathophysiology of NDDs and discuss the mechanisms of exosome formation, secretion, and action. Subsequently, we review the roles of exosomes in different types of NDDs, such as Alzheimer's disease and Parkinson's disease, with a special focus on their regulation of synaptic function. In addition, we explore the potential use of exosomes as biomarkers, as well as the challenges and opportunities in their clinical application. We provide perspectives on future research directions and development trends to provide a more comprehensive understanding of and guidance for the application of exosomes in the treatment of NDDs. In conclusion, exosomes rich in biological macromolecules, as a novel therapeutic strategy, have opened up new possibilities for the treatment of NDDs and brought new hope to patients.
Collapse
Affiliation(s)
- Ziying Liu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Lin Cheng
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Wa Cao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Respiratory Medicine, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Chunxiao Shen
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yuemin Qiu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Chuan Li
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Rehabilitation, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Seung Bum Yang
- Department of Medical Non-commissioned Officer, Wonkwang Health Science University Iksan-si, Jeollabuk-do 54538, South Korea
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaorong Zhang
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
3
|
Rodríguez-Prieto Á, Mateos-White I, Aníbal-Martínez M, Navarro-González C, Gil-Sanz C, Domínguez-Canterla Y, González-Manteiga A, Del Buey Furió V, López-Bendito G, Fazzari P. Nrg1 intracellular signaling regulates the development of interhemispheric callosal axons in mice. Life Sci Alliance 2024; 7:e202302250. [PMID: 38918041 PMCID: PMC11200272 DOI: 10.26508/lsa.202302250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Schizophrenia is associated with altered cortical circuitry. Although the schizophrenia risk gene NRG1 is known to affect the wiring of inhibitory interneurons, its role in excitatory neurons and axonal development is unclear. Here, we investigated the role of Nrg1 in the development of the corpus callosum, the major interhemispheric connection formed by cortical excitatory neurons. We found that deletion of Nrg1 impaired callosal axon development in vivo. Experiments in vitro and in vivo demonstrated that Nrg1 is cell-autonomously required for axonal outgrowth and that intracellular signaling of Nrg1 is sufficient to promote axonal development in cortical neurons and specifically in callosal axons. Furthermore, our data suggest that Nrg1 signaling regulates the expression of Growth Associated Protein 43, a key regulator of axonal growth. In conclusion, our study demonstrates that NRG1 is involved in the formation of interhemispheric callosal connections and provides a novel perspective on the relevance of NRG1 in excitatory neurons and in the etiology of schizophrenia.
Collapse
Affiliation(s)
- Ángela Rodríguez-Prieto
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Isabel Mateos-White
- Lab of Neural Development, BIOTECMED Institute, Universidad de Valencia, Valencia, Spain
| | - Mar Aníbal-Martínez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Carmen Navarro-González
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
- Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Cristina Gil-Sanz
- Lab of Neural Development, BIOTECMED Institute, Universidad de Valencia, Valencia, Spain
| | - Yaiza Domínguez-Canterla
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Ana González-Manteiga
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Verónica Del Buey Furió
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Pietro Fazzari
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| |
Collapse
|
4
|
Martins D, Abbasi M, Egas C, Arrais JP. Enhancing schizophrenia phenotype prediction from genotype data through knowledge-driven deep neural network models. Genomics 2024; 116:110910. [PMID: 39111546 DOI: 10.1016/j.ygeno.2024.110910] [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: 04/15/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Abstract
This article explores deep learning model design, drawing inspiration from the omnigenic model and genetic heterogeneity concepts, to improve schizophrenia prediction using genotype data. It introduces an innovative three-step approach leveraging neural networks' capabilities to efficiently handle genetic interactions. A locally connected network initially routes input data from variants to their corresponding genes. The second step employs an Encoder-Decoder to capture relationships among identified genes. The final model integrates knowledge from the first two and incorporates a parallel component to consider the effects of additional genes. This expansion enhances prediction scores by considering a larger number of genes. Trained models achieved an average AUC of 0.83, surpassing other genotype-trained models and matching gene expression dataset-based approaches. Additionally, tests on held-out sets reported an average sensitivity of 0.72 and an accuracy of 0.76, aligning with schizophrenia heritability predictions. Moreover, the study addresses genetic heterogeneity challenges by considering diverse population subsets.
Collapse
Affiliation(s)
- Daniel Martins
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Maryam Abbasi
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal; Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, Portugal; Research Centre for Natural Resources Environment and Society (CERNAS), Polytechnic Institute of Coimbra, Coimbra, Portugal.
| | - Conceição Egas
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Biocant - Transfer Technology Association, Cantanhede, Portugal; CNC - CNC Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Joel P Arrais
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Jiang A, You L, Handley RR, Hawkins V, Reid SJ, Jacobsen JC, Patassini S, Rudiger SR, Mclaughlan CJ, Kelly JM, Verma PJ, Bawden CS, Gusella JF, MacDonald ME, Waldvogel HJ, Faull RLM, Lehnert K, Snell RG. Single nuclei RNA-seq reveals a medium spiny neuron glutamate excitotoxicity signature prior to the onset of neuronal death in an ovine Huntington's disease model. Hum Mol Genet 2024; 33:1524-1539. [PMID: 38776957 PMCID: PMC11336116 DOI: 10.1093/hmg/ddae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Huntington's disease (HD) is a neurodegenerative genetic disorder caused by an expansion in the CAG repeat tract of the huntingtin (HTT) gene resulting in behavioural, cognitive, and motor defects. Current knowledge of disease pathogenesis remains incomplete, and no disease course-modifying interventions are in clinical use. We have previously reported the development and characterisation of the OVT73 transgenic sheep model of HD. The 73 polyglutamine repeat is somatically stable and therefore likely captures a prodromal phase of the disease with an absence of motor symptomatology even at 5-years of age and no detectable striatal cell loss. To better understand the disease-initiating events we have undertaken a single nuclei transcriptome study of the striatum of an extensively studied cohort of 5-year-old OVT73 HD sheep and age matched wild-type controls. We have identified transcriptional upregulation of genes encoding N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors in medium spiny neurons, the cell type preferentially lost early in HD. Further, we observed an upregulation of astrocytic glutamate uptake transporters and medium spiny neuron GABAA receptors, which may maintain glutamate homeostasis. Taken together, these observations support the glutamate excitotoxicity hypothesis as an early neurodegeneration cascade-initiating process but the threshold of toxicity may be regulated by several protective mechanisms. Addressing this biochemical defect early may prevent neuronal loss and avoid the more complex secondary consequences precipitated by cell death.
Collapse
Affiliation(s)
- Andrew Jiang
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, 131 Dong'an Road, Shanghai 200032, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, 130 Dong'an Road, Shanghai 200032, China
| | - Renee R Handley
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Victoria Hawkins
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Suzanne J Reid
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Jessie C Jacobsen
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Stefano Patassini
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Skye R Rudiger
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Clive J Mclaughlan
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Jennifer M Kelly
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Paul J Verma
- Aquatic and Livestock Sciences, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - C Simon Bawden
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - James F Gusella
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, United States
- Department of Genetics, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, United States
| | - Marcy E MacDonald
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, United States
- Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, United States
| | - Henry J Waldvogel
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Klaus Lehnert
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Russell G Snell
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
6
|
Deng XH, Liu XY, Wei YH, Wang K, Zhu JR, Zhong JJ, Zheng JY, Guo R, Zhu YF, Ye QH, Wang MD, Chen YJ, He JQ, Chen ZX, Huang SQ, Lv CS, Zheng GQ, Liu SF, Wen L. ErbB4 deficiency exacerbates olfactory dysfunction in an early-stage Alzheimer's disease mouse model. Acta Pharmacol Sin 2024:10.1038/s41401-024-01332-6. [PMID: 38982150 DOI: 10.1038/s41401-024-01332-6] [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: 02/13/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024] Open
Abstract
Olfactory dysfunction is increasingly recognized as an early indicator of Alzheimer's disease (AD). Aberrations in GABAergic function and the excitatory/inhibitory (E/I) balance within the olfactory bulb (OB) have been implicated in olfactory impairment during the initial stages of AD. While the neuregulin 1 (NRG1)/ErbB4 signaling pathway is known to regulate GABAergic transmission in the brain and is associated with various neuropsychiatric disorders, its specific role in early AD-related olfactory impairment remains incompletely understood. This study demonstrated that olfactory dysfunction preceded cognitive decline in young adult APP/PS1 mice and was characterized by reduced levels of NRG1 and ErbB4 in the OB. Further investigation revealed that deletion of ErbB4 in parvalbumin interneurons reduced GABAergic transmission and increased hyperexcitability in mitral and tufted cells (M/Ts) in the OB, thereby accelerating olfactory dysfunction in young adult APP/PS1 mice. Additionally, ErbB4 deficiency was associated with increased accumulation of Aβ and BACE1-mediated cleavage of APP, along with enhanced CDK5 signaling in the OB. NRG1 infusion into the OB was found to enhance GABAergic transmission in M/Ts and alleviate olfactory dysfunction in young adult APP/PS1 mice. These findings underscore the critical role of NRG1/ErbB4 signaling in regulating GABAergic transmission and E/I balance within the OB, contributing to olfactory impairment in young adult APP/PS1 mice, and provide novel insights for early intervention strategies in AD. This work has shown that ErbB4 deficiency increased the burden of Aβ, impaired GABAergic transmission, and disrupted the E/I balance of mitral and tufted cells (M/Ts) in the OB, ultimately resulting in olfactory dysfunction in young adult APP/PS1 mice. NRG1 could enhance GABAergic transmission, rescue E/I imbalance in M/Ts, and alleviate olfactory dysfunction in young adult APP/PS1 mice. OB: olfactory bulb, E/I: excitation/inhibition, Pr: probability of release, PV: parvalbumin interneurons, Aβ: β-amyloid, GABA: gamma-aminobutyric acid.
Collapse
Affiliation(s)
- Xian-Hua Deng
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Xing-Yang Liu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Yi-Hua Wei
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Ke Wang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Jun-Rong Zhu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jia-Jun Zhong
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jing-Yuan Zheng
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Rui Guo
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yi-Fan Zhu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Qiu-Hong Ye
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Meng-Dan Wang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ying-Jie Chen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jian-Quan He
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ze-Xu Chen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Shu-Qiong Huang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Chong-Shan Lv
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Guo-Qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China.
| | - Sui-Feng Liu
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Lei Wen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
7
|
Huang J, Zhang YY, Qiu YY, Yao S, Qiu WT, Peng JL, Li YQ, You QL, Wu CH, Wu EJ, Wang J, Zhou YL, Ning YP, Wang HS, Chen WB, Hu BJ, Liu Y, Sun XD. NRG1-ErbB4 signaling in the medial amygdala controls mating motivation in adult male mice. Cell Rep 2024; 43:113905. [PMID: 38446660 DOI: 10.1016/j.celrep.2024.113905] [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: 12/19/2022] [Revised: 12/12/2023] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Motivation-driven mating is a basic affair for the maintenance of species. However, the underlying molecular mechanisms that control mating motivation are not fully understood. Here, we report that NRG1-ErbB4 signaling in the medial amygdala (MeA) is pivotal in regulating mating motivation. NRG1 expression in the MeA negatively correlates with the mating motivation levels in adult male mice. Local injection and knockdown of MeA NRG1 reduce and promote mating motivation, respectively. Consistently, knockdown of MeA ErbB4, a major receptor for NRG1, and genetic inactivation of its kinase both promote mating motivation. ErbB4 deletion decreases neuronal excitability, whereas chemogenetic manipulations of ErbB4-positive neuronal activities bidirectionally modulate mating motivation. We also identify that the effects of NRG1-ErbB4 signaling on neuronal excitability and mating motivation rely on hyperpolarization-activated cyclic nucleotide-gated channel 3. This study reveals a critical molecular mechanism for regulating mating motivation in adult male mice.
Collapse
Affiliation(s)
- Jie Huang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yan-Yan Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yu-Yang Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shan Yao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wan-Ting Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jin-Lin Peng
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan-Quan Li
- Department of Neurology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Qiang-Long You
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Cui-Hong Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Er-Jian Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jin Wang
- Department of Physiology, Guangxi University of Science and Technology, Liuzhou, China
| | - Yan-Ling Zhou
- Department of Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu-Ping Ning
- Department of Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong-Sheng Wang
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen-Bing Chen
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Bing-Jie Hu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China.
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| | - Xiang-Dong Sun
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Popović L, Wintgens JP, Wu Y, Brankatschk B, Menninger S, Degenhart C, Jensen N, Wichert SP, Klebl B, Rossner MJ, Wehr MC. Profiling of ERBB receptors and downstream pathways reveals selectivity and hidden properties of ERBB4 antagonists. iScience 2024; 27:108839. [PMID: 38303712 PMCID: PMC10831936 DOI: 10.1016/j.isci.2024.108839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
ERBB receptor tyrosine kinases are involved in development and diseases like cancer, cardiovascular, neurodevelopmental, and mental disorders. Although existing drugs target ERBB receptors, the next generation of drugs requires enhanced selectivity and understanding of physiological pathway responses to improve efficiency and reduce side effects. To address this, we developed a multilevel barcoded reporter profiling assay, termed 'ERBBprofiler', in living cells to monitor the activity of all ERBB targets and key physiological pathways simultaneously. This assay helps differentiate on-target therapeutic effects from off-target and off-pathway side effects of ERBB antagonists. To challenge the assay, eight established ERBB antagonists were profiled. Known effects were confirmed, and previously uncharacterized properties were discovered, such as pyrotinib's preference for ERBB4 over EGFR. Additionally, two lead compounds selectively targeting ERBB4 were profiled, showing promise for clinical trials. Taken together, this multiparametric profiling approach can guide early-stage drug development and lead to improved future therapeutic interventions.
Collapse
Affiliation(s)
- Lukša Popović
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Jan P. Wintgens
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Ben Brankatschk
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Sascha Menninger
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Carsten Degenhart
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Niels Jensen
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Sven P. Wichert
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Bert Klebl
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Moritz J. Rossner
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Michael C. Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| |
Collapse
|
9
|
He XY, Huang ZH, Wang F, Chen ZL, Wang SB, Jia FJ, Hou CL. Gene Polymorphisms and Expression of NRG1, DAOA, and DISC1 Genes in a Chinese Han Population with an Ultra-High Risk for Psychosis. Neuropsychiatr Dis Treat 2023; 19:2521-2533. [PMID: 38029052 PMCID: PMC10667082 DOI: 10.2147/ndt.s434856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/05/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Although there is previous evidence supporting that ultra-high risk (UHR) for psychosis transformation is associated with NRG1, DAOA, and DISC1 genes, there have been no relevant studies in the Chinese population. The objective of the current study was to explore the gene polymorphism and expression of NRG1, DAOA, and DISC1 genes in a Han population with UHR for psychosis in China. Methods Eighteen UHR individuals, 61 first-degree relatives of patients with schizophrenia (FDR), 55 first-episode psychosis individuals (FEP), and 61 healthy controls (HC) were enrolled in the study. The genotypes at four loci of the NRG1 gene, four loci of the DAOA gene, and two loci of the DISC1 gene were tested for all subjects, and mRNAs of NRG1 and DISC1 were examined and analyzed in a pairwise comparison among the four groups. Statistical analysis of genetics was performed using snpStats software. For the case-control association analysis, a single site association study, epistatic effect analysis, and haplotype analysis were used to explore the association of the above genes. Results This study found that rs3918341 in the DAOA gene was associated with susceptibility to UHR by single site association analysis. Epistatic effect analysis results showed that the NRG1 gene interacted with the DAOA gene and DISC1 gene in the susceptibility to UHR. Haplotype association analysis showed that all haplotypes were not significantly associated with UHR. NRG1 mRNA was significantly downregulated in the UHR group compared with the HC group as well as the FEP group. Conclusion Our preliminary results show that NRG1, DAOA, and DISC1 genes may play a role in psychosis onset, opening the way to the identification of prognostic biomarkers.
Collapse
Affiliation(s)
- Xiao-Yan He
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
- Psychiatric Rehabilitation Section, The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi Central Rehabilitation Hospital, Wuxi, People’s Republic of China
| | - Zhuo-Hui Huang
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Fei Wang
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Zi-Lang Chen
- Psychiatry Department, Luoding Mental Health Center, Yunfu, People’s Republic of China
| | - Shi-Bin Wang
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Fu-Jun Jia
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Cai-Lan Hou
- Psychological Department, Guangdong Mental Health Center, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
10
|
Vincent B, Maitra S. BACE1-dependent metabolism of neuregulin 1: Bridging the gap in explaining the occurrence of schizophrenia-like symptoms in Alzheimer's disease with psychosis? Ageing Res Rev 2023; 89:101988. [PMID: 37331479 DOI: 10.1016/j.arr.2023.101988] [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: 03/08/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
Alzheimer's disease is a neurodegenerative disease mainly characterized by cortico-neuronal atrophy, impaired memory and other cognitive declines. On the other hand, schizophrenia is a neuro-developmental disorder with an overtly active central nervous system pruning system resulting into abrupt connections with common symptoms including disorganised thoughts, hallucination and delusion. Nevertheless, the fronto-temporal anomaly presents itself as a common denominator for the two pathologies. There is even a strong presumption of increased risk of developing co-morbid dementia for schizophrenic individuals and psychosis for Alzheimer's disease patients, overall leading to a further deteriorated quality of life. However, convincing proofs of how these two disorders, although very distant from each other when considering their aetiology, develop coexisting symptoms is yet to be resolved. At the molecular level, the two primarily neuronal proteins β-amyloid precursor protein and neuregulin 1 have been considered in this relevant context, although the conclusions are for the moment only hypotheses. In order to propose a model for explaining the psychotic schizophrenia-like symptoms that sometimes accompany AD-associated dementia, this review projects out on the similar sensitivity shared by these two proteins regarding their metabolism by the β-site APP cleaving enzyme 1.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560 Valbonne, France.
| | - Subhamita Maitra
- Department of Molecular Biology, Umeå University, Umeå 90736, Sweden
| |
Collapse
|
11
|
Batista-Brito R, Majumdar A, Nuño A, Ward C, Barnes C, Nikouei K, Vinck M, Cardin JA. Developmental loss of ErbB4 in PV interneurons disrupts state-dependent cortical circuit dynamics. Mol Psychiatry 2023; 28:3133-3143. [PMID: 37069344 PMCID: PMC10618960 DOI: 10.1038/s41380-023-02066-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/28/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023]
Abstract
GABAergic inhibition plays an important role in the establishment and maintenance of cortical circuits during development. Neuregulin 1 (Nrg1) and its interneuron-specific receptor ErbB4 are key elements of a signaling pathway critical for the maturation and proper synaptic connectivity of interneurons. Using conditional deletions of the ERBB4 gene in mice, we tested the role of this signaling pathway at two developmental timepoints in parvalbumin-expressing (PV) interneurons, the largest subpopulation of cortical GABAergic cells. Loss of ErbB4 in PV interneurons during embryonic, but not late postnatal development leads to alterations in the activity of excitatory and inhibitory cortical neurons, along with severe disruption of cortical temporal organization. These impairments emerge by the end of the second postnatal week, prior to the complete maturation of the PV interneurons themselves. Early loss of ErbB4 in PV interneurons also results in profound dysregulation of excitatory pyramidal neuron dendritic architecture and a redistribution of spine density at the apical dendritic tuft. In association with these deficits, excitatory cortical neurons exhibit normal tuning for sensory inputs, but a loss of state-dependent modulation of the gain of sensory responses. Together these data support a key role for early developmental Nrg1/ErbB4 signaling in PV interneurons as a powerful mechanism underlying the maturation of both the inhibitory and excitatory components of cortical circuits.
Collapse
Affiliation(s)
- Renata Batista-Brito
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
- Department of Psychiatry and Behavioral Sciences, Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
- Department of Genetics, Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
| | - Antara Majumdar
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Sherrington Road, Oxford, OX1 3PT, England
| | - Alejandro Nuño
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
| | - Claire Ward
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA
| | - Clayton Barnes
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
| | - Kasra Nikouei
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Vinck
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Deutschordenstraße 46, 60528, Frankfurt, Germany
| | - Jessica A Cardin
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
- Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- Wu Tsai Institute, Yale University, 100 College St., New Haven, CT, 06520, USA.
| |
Collapse
|
12
|
Liu Y, Ding M, Pan S, Zhou R, Yao J, Fu R, Yu H, Lu Z. MicroRNA-23a-3p is upregulated in plasma exosomes of bulbar-onset ALS patients and targets ERBB4. Neuroscience 2023:S0306-4522(23)00250-6. [PMID: 37290686 DOI: 10.1016/j.neuroscience.2023.05.030] [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: 01/09/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/10/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease related to the progressive death of motor neurons. Understanding the pathogenesis of ALS continues to provide considerable challenges. Bulbar-onset ALS involves faster functional loss and shorter survival time than spinal cord-onset ALS. However, debate is ongoing regarding typical plasma miRNA changes in ALS patients with bulbar onset. Exosomal miRNAs have not yet been described as a tool for bulbar-onset ALS diagnosis or prognosis prediction. In this study, candidate exosomal miRNAs were identified by small RNA sequencing using samples from patients with bulbar-onset ALS and healthy controls. Potential pathogenic mechanisms were identified through enrichment analysis of target genes for differential miRNAs. Expression of miR-16-5p, miR-23a-3p, miR-22-3p, and miR-93-5p was significantly up-regulated in plasma exosomes from bulbar-onset ALS patients compared with healthy control subjects. Among them, miR-16-5p and miR-23a-3p were significantly lower in spinal-onset ALS patients than those with bulbar-onset. Furthermore, up-regulation of miR-23a-3p in motor neuron-like NSC-34 cells promoted apoptosis and inhibited cell viability. This miRNA was found to directly target ERBB4 and regulate the AKT/GSK3β pathway. Collectively, the above miRNAs and their targets are related to the development of bulbar-onset ALS. Our research indicates that miR-23a-3p might have an effect on motor neuron loss observed in bulbar-onset ALS and may be a novel target for the therapy of ALS in the future.
Collapse
Affiliation(s)
- Yue Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Man Ding
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Sijia Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rumeng Zhou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiajia Yao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Fu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hang Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Zhang Y, Zhu Y, Wang J, Jin L, Guo M, Chen L, Zhang L, Li Y, Wan B, Zhang R, Jia W, Hu C. Neuregulin4 Acts on Hypothalamic ErBb4 to Excite Oxytocin Neurons and Preserve Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204824. [PMID: 37060105 PMCID: PMC10238187 DOI: 10.1002/advs.202204824] [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: 08/22/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Neuregulin 4 (Nrg4) is an adipose tissue-enriched secreted factor that modulates glucose and lipid metabolism. Nrg4 is closely associated with obesity and preserves diet-induced metabolic disorders. However, the specific mechanisms via which Nrg4 regulates metabolic homeostasis remain incompletely understood. Here, this work finds that the Nrg4 receptor, ErbB4, is highly expressed in the hypothalamus, and the phosphorylation of hypothalamic ErbB4 is reduced in diet-induced obesity (DIO) mice. Peripheral Nrg4 can act on ErbB4 via blood circulation and excite neurons in the paraventricular nucleus of hypothalamus (PVN). Central administration of recombinant Nrg4 protein (rNrg4) reduces obesity and related metabolic disorders by influencing energy expenditure and intake. Overexpression of ErbB4 in the PVN protects against obesity, whereas its knock down in oxytocin (Oxt) neuron accelerates obesity. Furthermore, Nrg4-ErbB4 signaling excites Oxt release, and ablation of Oxt neuron considerably attenuates the effect of Nrg4 on energy balance. These data suggest that the hypothalamus is a key target of Nrg4, which partially explains the multifaceted roles of Nrg4 in metabolism.
Collapse
Affiliation(s)
- Yi Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Zhu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| | - Jinghui Wang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of EndocrinologyAffiliated Hospital of Nantong UniversityNantong226001P. R. China
| | - Li Jin
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Liwei Chen
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Lina Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Li
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Baocheng Wan
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Rong Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Weiping Jia
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Cheng Hu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| |
Collapse
|
14
|
Sun W, Xie G, Jiang X, Khaitovich P, Han D, Liu X. Epigenetic regulation of human-specific gene expression in the prefrontal cortex. BMC Biol 2023; 21:123. [PMID: 37226244 DOI: 10.1186/s12915-023-01612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/03/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Changes in gene expression levels during brain development are thought to have played an important role in the evolution of human cognition. With the advent of high-throughput sequencing technologies, changes in brain developmental expression patterns, as well as human-specific brain gene expression, have been characterized. However, interpreting the origin of evolutionarily advanced cognition in human brains requires a deeper understanding of the regulation of gene expression, including the epigenomic context, along the primate genome. Here, we used chromatin immunoprecipitation sequencing (ChIP-seq) to measure the genome-wide profiles of histone H3 lysine 4 trimethylation (H3K4me3) and histone H3 lysine 27 acetylation (H3K27ac), both of which are associated with transcriptional activation in the prefrontal cortex of humans, chimpanzees, and rhesus macaques. RESULTS We found a discrete functional association, in which H3K4me3HP gain was significantly associated with myelination assembly and signaling transmission, while H3K4me3HP loss played a vital role in synaptic activity. Moreover, H3K27acHP gain was enriched in interneuron and oligodendrocyte markers, and H3K27acHP loss was enriched in CA1 pyramidal neuron markers. Using strand-specific RNA sequencing (ssRNA-seq), we first demonstrated that approximately 7 and 2% of human-specific expressed genes were epigenetically marked by H3K4me3HP and H3K27acHP, respectively, providing robust support for causal involvement of histones in gene expression. We also revealed the co-activation role of epigenetic modification and transcription factors in human-specific transcriptome evolution. Mechanistically, histone-modifying enzymes at least partially contribute to an epigenetic disturbance among primates, especially for the H3K27ac epigenomic marker. In line with this, peaks enriched in the macaque lineage were found to be driven by upregulated acetyl enzymes. CONCLUSIONS Our results comprehensively elucidated a causal species-specific gene-histone-enzyme landscape in the prefrontal cortex and highlighted the regulatory interaction that drove transcriptional activation.
Collapse
Affiliation(s)
- Weifen Sun
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, 200063, China
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gangcai Xie
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China
| | - Xi Jiang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China
| | - Philipp Khaitovich
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China.
- Skolkovo Institute of Science and Technology, Moscow, 121205, Russia.
| | - Dingding Han
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China.
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China.
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, 200063, China.
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China.
| |
Collapse
|
15
|
Sun YM, Chen J. Editorial: New insights into schizophrenia-related neural and behavioral phenotypes. Front Cell Neurosci 2023; 17:1202230. [PMID: 37234917 PMCID: PMC10206298 DOI: 10.3389/fncel.2023.1202230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Affiliation(s)
| | - Ji Chen
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China
- Department of Psychiatry, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| |
Collapse
|
16
|
Dysfunction of NRG1/ErbB4 Signaling in the Hippocampus Might Mediate Long-term Memory Decline After Systemic Inflammation. Mol Neurobiol 2023; 60:3210-3226. [PMID: 36840846 DOI: 10.1007/s12035-023-03278-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Abstract
Accumulating evidence has suggested that a great proportion of sepsis survivors suffer from long-term cognitive impairments after hospital discharge, leading to decreased life quality and substantial caregiving burdens for family members. However, the underlying mechanism remains unclear. In the present study, we established a mouse model of systemic inflammation by repeated lipopolysaccharide (LPS) injections. A combination of behavioral tests, biochemical, and in vivo electrophysiology techniques were conducted to test whether abnormal NRG1/ErbB4 signaling, parvalbumin (PV) interneurons, and hippocampal neural oscillations were involved in memory decline after repeated LPS injections. Here, we showed that LPS induced long-term memory decline, which was accompanied by dysfunction of NRG1/ErbB4 signaling and PV interneurons, and decreased theta and gamma oscillations. Notably, NRG1 treatment reversed LPS-induced decreases in p-ErbB4 and PV expressions, abnormalities in theta and gamma oscillations, and long-term memory decline. Together, our study demonstrated that dysfunction of NRG1/ErbB4 signaling in the hippocampus might mediate long-term memory decline in a mouse model of systemic inflammation induced by repeated LPS injections. Thus, targeting NRG1/ErbB4 signaling in the hippocampus may be promising for the prevention and treatment of this long-term memory decline.
Collapse
|
17
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
18
|
von Bohlen Und Halbach O. Neurotrophic Factors and Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:223-254. [PMID: 37962797 DOI: 10.1007/978-3-031-36159-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spines are highly dynamic structures that play important roles in neuronal plasticity. The morphologies and the numbers of dendritic spines are highly variable, and this diversity is correlated with the different morphological and physiological features of this neuronal compartment. Dendritic spines can change their morphology and number rapidly, allowing them to adapt to plastic changes. Neurotrophic factors play important roles in the brain during development. However, these factors are also necessary for a variety of processes in the postnatal brain. Neurotrophic factors, especially members of the neurotrophin family and the ephrin family, are involved in the modulation of long-lasting effects induced by neuronal plasticity by acting on dendritic spines, either directly or indirectly. Thereby, the neurotrophic factors play important roles in processes attributed, for example, to learning and memory.
Collapse
|
19
|
Spahic H, Parmar P, Miller S, Emerson PC, Lechner C, St. Pierre M, Rastogi N, Nugent M, Duck SA, Kirkwood A, Chavez-Valdez R. Dysregulation of ErbB4 Signaling Pathway in the Dorsal Hippocampus after Neonatal Hypoxia-Ischemia and Late Deficits in PV + Interneurons, Synaptic Plasticity and Working Memory. Int J Mol Sci 2022; 24:ijms24010508. [PMID: 36613949 PMCID: PMC9820818 DOI: 10.3390/ijms24010508] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxic-ischemic (HI) injury leads to deficits in hippocampal parvalbumin (PV)+ interneurons (INs) and working memory. Therapeutic hypothermia (TH) does not prevent these deficits. ErbB4 supports maturation and maintenance of PV+ IN. Thus, we hypothesized that neonatal HI leads to persistent deficits in PV+ INs, working memory and synaptic plasticity associated with ErbB4 dysregulation despite TH. P10 HI-injured mice were randomized to normothermia (NT, 36 °C) or TH (31 °C) for 4 h and compared to sham. Hippocampi were studied for α-fodrin, glial fibrillary acidic protein (GFAP), and neuroregulin (Nrg) 1 levels; erb-b2 receptor tyrosine kinase 4 (ErbB4)/ Ak strain transforming (Akt) activation; and PV, synaptotagmin (Syt) 2, vesicular-glutamate transporter (VGlut) 2, Nrg1, and ErbB4 expression in coronal sections. Extracellular field potentials and behavioral testing were performed. At P40, deficits in PV+ INs correlated with impaired memory and coincided with blunted long-term depression (LTD), heightened long-term potentiation (LTP) and increased Vglut2/Syt2 ratio, supporting excitatory-inhibitory (E/I) imbalance. Hippocampal Nrg1 levels were increased in the hippocampus 24 h after neonatal HI, delaying the decline documented in shams. Paradoxically ErbB4 activation decreased 24 h and again 30 days after HI. Neonatal HI leads to persistent deficits in hippocampal PV+ INs, memory, and synaptic plasticity. While acute decreased ErbB4 activation supports impaired maturation and survival after HI, late deficit reemergence may impair PV+ INs maintenance after HI.
Collapse
Affiliation(s)
- Harisa Spahic
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pritika Parmar
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah Miller
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul Casey Emerson
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Charles Lechner
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mark St. Pierre
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Neetika Rastogi
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Nugent
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Ann Duck
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
20
|
Talebi F, Ghorbani S, Alizadeh L, Akhlaghi F, Sadat Moeeni S, Karimzadeh F. Alteration in Neuregulin 1/ERbB4 in Absence Epilepsy: Regulatory Effect on TRPV1 Expression. Basic Clin Neurosci 2022; 13:777-788. [PMID: 37323956 PMCID: PMC10262282 DOI: 10.32598/bcn.2021.731.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/14/2021] [Accepted: 07/05/2021] [Indexed: 06/17/2023] Open
Abstract
Introduction The footprint of Neuregulin 1 (NRG1) / ERbB4 in the pathophysiology of some neurological disorders and TRPV1 regulation has been indicated. The alterations in NRG1 and ErbB4 as well as the TRPV1 signaling pathway were investigated during the development of absence epilepsy in the genetic animal model of absence epilepsy. Methods Male WAG/Rij and Wistar rats were divided into four experimental groups of two and six months of age. The protein levels of NRG1, ERbB4, and TRPV1 were measured in the somatosensory cortex and hippocampus. Results The cortical protein levels of NRG1 and ErbB4 in the 6-month-old WAG/Rij rats were lower than in Wistar rats. Protein levels of TRPV1 were lower in two- and six-month-old WAG/Rij rats compared to age-matched Wistar rats.Hippocampal protein levels of NRG1 in 6-month-old WAG/Rij rats were lower than two-month-old WAG/Rij rats. Low levels of ErbB4 protein in two-month-old and high levels in six-month-old WAG/Rij rats were found compared to Wistar rats. Protein levels of TRPV1 were lower in the two-month-old and higher in the six-month-old WAG/Rij rats compared to age-matched Wistar rats.Furthermore, a high correlation between NRG1/ERbB4 and TRPV1 expressions in the cortex and hippocampus was indicated. The expression of NRG1/ERbB4 and TRPV1 followed a similar pattern during the life span of Wistar and WAG/Rij rats. Conclusion Our findings indicated the potential role of the NRG1/ErbB4 pathway as well as TRPV1 in the pathogenesis of absence epilepsy. The regulatory effect of the ERbB4 receptor on the TRPV1 expression has been suggested following the similar pattern of expression.
Collapse
Affiliation(s)
- Farideh Talebi
- Immunoregulation Research Center, Shahed University, Tehran, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Samira Ghorbani
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Leila Alizadeh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Fatemeh Akhlaghi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Sadat Moeeni
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Karimzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Greenwood TA. Genetic Influences on Cognitive Dysfunction in Schizophrenia. Curr Top Behav Neurosci 2022; 63:291-314. [PMID: 36029459 DOI: 10.1007/7854_2022_388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Schizophrenia is a severe and debilitating psychotic disorder that is highly heritable and relatively common in the population. The clinical heterogeneity associated with schizophrenia is substantial, with patients exhibiting a broad range of deficits and symptom severity. Large-scale genomic studies employing a case-control design have begun to provide some biological insight. However, this strategy combines individuals with clinically diverse symptoms and ignores the genetic risk that is carried by many clinically unaffected individuals. Consequently, the majority of the genetic architecture underlying schizophrenia remains unexplained, and the pathways by which the implicated variants contribute to the clinically observable signs and symptoms are still largely unknown. Parsing the complex, clinical phenotype of schizophrenia into biologically relevant components may have utility in research aimed at understanding the genetic basis of liability. Cognitive dysfunction is a hallmark symptom of schizophrenia that is associated with impaired quality of life and poor functional outcome. Here, we examine the value of quantitative measures of cognitive dysfunction to objectively target the underlying neurobiological pathways and identify genetic variants and gene networks contributing to schizophrenia risk. For a complex disorder, quantitative measures are also more efficient than diagnosis, allowing for the identification of associated genetic variants with fewer subjects. Such a strategy supplements traditional analyses of schizophrenia diagnosis, providing the necessary biological insight to help translate genetic findings into actionable treatment targets. Understanding the genetic basis of cognitive dysfunction in schizophrenia may thus facilitate the development of novel pharmacological and procognitive interventions to improve real-world functioning.
Collapse
Affiliation(s)
- Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
22
|
Shiosaka S. Kallikrein 8: A key sheddase to strengthen and stabilize neural plasticity. Neurosci Biobehav Rev 2022; 140:104774. [PMID: 35820483 DOI: 10.1016/j.neubiorev.2022.104774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Neural networks are modified and reorganized throughout life, even in the matured brain. Synapses in the networks form, change, or disappear dynamically in the plasticity state. The pre- and postsynaptic signaling, transmission, and structural dynamics have been studied considerably well. However, not many studies have shed light on the events in the synaptic cleft and intercellular space. Neural activity-dependent protein shedding is a phenomenon in which (1) presynaptic excitation evokes secretion or activation of sheddases, (2) sheddases are involved not only in cleavage of membrane- or matrix-bound proteins but also in mechanical modulation of cell-to-cell connectivity, and (3) freed activity domains of protein factors play a role in receptor-mediated or non-mediated biological actions. Kallikrein 8/neuropsin (KLK8) is a kallikrein family serine protease rich in the mammalian limbic brain. Accumulated evidence has suggested that KLK8 is an important modulator of neural plasticity and consequently, cognition. Insufficiency, as well as excess of KLK8 may have detrimental effects on limbic functions.
Collapse
Affiliation(s)
- Sadao Shiosaka
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka Prefectural Hospital Organization, Miyanosaka 3-16-21, Hirakata-shi, Osaka 573-0022, Japan.
| |
Collapse
|
23
|
Vrillon A, Mouton-Liger F, Martinet M, Cognat E, Hourregue C, Dumurgier J, Bouaziz-Amar E, Brinkmalm A, Blennow K, Zetterberg H, Hugon J, Paquet C. Plasma neuregulin 1 as a synaptic biomarker in Alzheimer's disease: a discovery cohort study. Alzheimers Res Ther 2022; 14:71. [PMID: 35606871 PMCID: PMC9125890 DOI: 10.1186/s13195-022-01014-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Synaptic dysfunction is an early core feature of Alzheimer's disease (AD), closely associated with cognitive symptoms. Neuregulin 1 (NRG1) is a growth and differentiation factor with a key role in the development and maintenance of synaptic transmission. Previous reports have shown that changes in cerebrospinal fluid (CSF) NRG1 concentration are associated with cognitive status and biomarker evidence of AD pathology. Plasma biomarkers reflecting synaptic impairment would be of great clinical interest. OBJECTIVE To measure plasma NRG1 concentration in AD patients in comparison with other neurodegenerative disorders and neurological controls (NC) and to study its association with cerebrospinal fluid (CSF) core AD and synaptic biomarkers. METHODS This retrospective study enrolled 127 participants including patients with AD at mild cognitive impairment stage (AD-MCI, n = 27) and at dementia stage (n = 35), non-AD dementia (n = 26, Aβ-negative), non-AD MCI (n = 19), and neurological controls (n=20). Plasma and CSF NRG1, as well as CSF core AD biomarkers (Aβ 42/Aβ 40 ratio, phospho-tau, and total tau), were measured using ELISA. CSF synaptic markers were measured using ELISA for GAP-43 and neurogranin and through immunoprecipitation mass spectrometry for SNAP-25. RESULTS Plasma NRG1 concentration was higher in AD-MCI and AD dementia patients compared with neurological controls (respectively P = 0.005 and P < 0.001). Plasma NRG1 differentiated AD MCI patients from neurological controls with an area under the curve of 88.3%, and AD dementia patients from NC with an area under the curve of 87.3%. Plasma NRG1 correlated with CSF NRG1 (β = 0.372, P = 0.0056, adjusted on age and sex). Plasma NRG1 was associated with AD CSF core biomarkers in the whole cohort and in Aβ-positive patients (β = -0.197-0.423). Plasma NRG1 correlated with CSF GAP-43, neurogranin, and SNAP-25 (β = 0.278-0.355). Plasma NRG1 concentration correlated inversely with MMSE in the whole cohort and in Aβ-positive patients (all, β = -0.188, P = 0.038; Aβ+: β = -0.255, P = 0.038). CONCLUSION Plasma NRG1 concentration is increased in AD patients and correlates with CSF core AD and synaptic biomarkers and cognitive status. Thus, plasma NRG1 is a promising non-invasive biomarker to monitor synaptic impairment in AD.
Collapse
Affiliation(s)
- Agathe Vrillon
- Université Paris Cité, Inserm U1144, Paris, France.
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France.
| | | | - Matthieu Martinet
- Université Paris Cité, Inserm U1144, Paris, France
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| | - Emmanuel Cognat
- Université Paris Cité, Inserm U1144, Paris, France
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| | - Claire Hourregue
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| | - Julien Dumurgier
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| | - Elodie Bouaziz-Amar
- Université Paris Cité, Inserm U1144, Paris, France
- Université Paris Cité, Department of Biochemistry, APHP GHU Nord Lariboisière-Fernand Widal, Paris, France
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, The Salhgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, The Salhgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Salhgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jacques Hugon
- Université Paris Cité, Inserm U1144, Paris, France
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| | - Claire Paquet
- Université Paris Cité, Inserm U1144, Paris, France
- Université Paris Cité, Center of Cognitive Neurology, Lariboisière Fernand-Widal Hospital, APHP, 200 rue du Faubourg Saint-Denis, 75010, Paris, France
| |
Collapse
|
24
|
Differential expression of miR-148b, miR-129-2 and miR-296 in animal models of schizophrenia-Relevance to NMDA receptor hypofunction. Neuropharmacology 2022; 210:109024. [PMID: 35276119 DOI: 10.1016/j.neuropharm.2022.109024] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/29/2022] [Accepted: 03/05/2022] [Indexed: 12/26/2022]
Abstract
Hypofunction of N-methyl-d-aspartate receptors (NMDAR) is a key component in the pathophysiology of schizophrenia. Alterations in the regulation of NMDARs by microRNAs (miRNAs) are possible since numerous miRNAs are differentially expressed in post mortem schizophrenia brain samples. We screened the miRNAs that are altered in schizophrenia against the targets, Grin2A and Grin2B subunits of NMDAR using bioinformatic tools. Among the predicted miRNAs some interacted with the 3'-UTR sequences of Grin2A (miR-296, miR-148b, miR-129-2, miR-137) and Grin2B (miR-296, miR-148b, miR-129-2, miR-223) in dual luciferase assays. This was supported by downregulation of the GluN2B protein in primary hippocampal neurons upon overexpressing Grin2B targeting miRNAs. In two models of schizophrenia-pharmacological MK-801 model and neurodevelopmental methylazoxymethanol acetate (MAM) model which showed cognitive deficits - protein levels of GluN2A and GluN2B were downregulated but their transcript levels were upregulated. miR-296-3p, miR-148b-5p and miR-137 levels showed upregulation in both models which could have interacted with Grin2A/Grin2B transcripts resulting in translational arrest. In MAM model, reciprocal changes in the expression of the 3p and 5p forms of miR-148b and miR-137 were observed. Expression of some genes implicated in schizophrenia such as Neuregulin 1 (NRG1), BDNF and CaMKIIα, were also altered in these models. This is the first report showing downregulation of GluN2A and GluN2B by miR-296, miR-148b and miR-129-2 in vitro and association between them in animal models. Mining miRNAs regulating NMDA receptors might give insights into the pathophysiology of this disorder, providing avenues in therapeutics.
Collapse
|
25
|
Rey CC, Robert V, Bouisset G, Loisy M, Lopez S, Cattaud V, Lejards C, Piskorowski RA, Rampon C, Chevaleyre V, Verret L. Altered inhibitory function in hippocampal CA2 contributes in social memory deficits in Alzheimer’s mouse model. iScience 2022; 25:103895. [PMID: 35243253 PMCID: PMC8873612 DOI: 10.1016/j.isci.2022.103895] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/07/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Parvalbumin (PV)-expressing interneurons which are often associated with the specific extracellular matrix perineuronal net (PNN) play a critical role in the alteration of brain activity and memory performance in Alzheimer’s disease (AD). The integrity of these neurons is crucial for normal functioning of the hippocampal subfield CA2, and hence, social memory formation. Here, we find that social memory deficits of mouse models of AD are associated with decreased presence of PNN around PV cells and long-term synaptic plasticity in area CA2. Furthermore, single local injection of the growth factor neuregulin-1 (NRG1) is sufficient to restore both PV/PNN levels and social memory performance of these mice. Thus, the PV/PNN disruption in area CA2 could play a causal role in social memory deficits of AD mice, and activating PV cell pro-maturation pathways may be sufficient to restore social memory. Tg2576 mouse model of AD have normal sociability, but cannot form social memory Tg2576 mice have less detectable PV interneurons and PNN in hippocampal area CA2 PV-dependent long-term plasticity is altered in CA2 of Tg2576 mice NRG1 in CA2 increases PV/PNN and restores social memory of these AD mice
Collapse
|
26
|
Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 2022; 27:731-743. [PMID: 34163013 PMCID: PMC8695640 DOI: 10.1038/s41380-021-01196-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The neurobiology of schizophrenia involves multiple facets of pathophysiology, ranging from its genetic basis over changes in neurochemistry and neurophysiology, to the systemic level of neural circuits. Although the precise mechanisms associated with the neuropathophysiology remain elusive, one essential aspect is the aberrant maturation and connectivity of the prefrontal cortex that leads to complex symptoms in various stages of the disease. Here, we focus on how early developmental dysfunction, especially N-methyl-D-aspartate receptor (NMDAR) development and hypofunction, may lead to the dysfunction of both local circuitry within the prefrontal cortex and its long-range connectivity. More specifically, we will focus on an "all roads lead to Rome" hypothesis, i.e., how NMDAR hypofunction during development acts as a convergence point and leads to local gamma-aminobutyric acid (GABA) deficits and input-output dysconnectivity in the prefrontal cortex, which eventually induce cognitive and social deficits. Many outstanding questions and hypothetical mechanisms are listed for future investigations of this intriguing hypothesis that may lead to a better understanding of the aberrant maturation and connectivity associated with the prefrontal cortex.
Collapse
|
27
|
Insulin-like growth factor I mitigates post-traumatic stress by inhibiting AMP-kinase in orexin neurons. Mol Psychiatry 2022; 27:2182-2196. [PMID: 35115701 PMCID: PMC9126821 DOI: 10.1038/s41380-022-01442-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022]
Abstract
Maladaptive coping behaviors are probably involved in post-traumatic stress disorders (PTSD), but underlying mechanisms are incompletely understood. We now report that mice lacking functional insulin-like growth factor I (IGF-I) receptors in orexin neurons of the lateral hypothalamus (Firoc mice) are unresponsive to the anxiolytic actions of IGF-I and develop PTSD-like behavior that is ameliorated by inhibition of orexin neurons. Conversely, systemic IGF-I treatment ameliorated PTSD-like behavior in a wild-type mouse model of PTSD (PTSD mice). Further, systemic IGF-I modified the GABA/Glutamate synaptic structure in orexin neurons of naïve wild-type mice by increasing the dephosphorylation of GABA(B) receptor subunit through inhibition of AMP-kinase (AMPK). Significantly, pharmacological inhibition of AMPK mimicked IGF-I, normalizing fear behavior in PTSD mice. Thus, we suggest that IGF-I enables coping behaviors by balancing E/I input onto orexin neurons in a context-dependent manner. These observations provide a novel therapeutic approach to PTSD through modulation of AMPK.
Collapse
|
28
|
Fernández-Teruel A, Oliveras I, Cañete T, Rio-Álamos C, Tapias-Espinosa C, Sampedro-Viana D, Sánchez-González A, Sanna F, Torrubia R, González-Maeso J, Driscoll P, Morón I, Torres C, Aznar S, Tobeña A, Corda MG, Giorgi O. Neurobehavioral and neurodevelopmental profiles of a heuristic genetic model of differential schizophrenia- and addiction-relevant features: The RHA vs. RLA rats. Neurosci Biobehav Rev 2021; 131:597-617. [PMID: 34571119 DOI: 10.1016/j.neubiorev.2021.09.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022]
Abstract
The Roman High- (RHA) and Low-(RLA) avoidance rat lines/strains were generated through bidirectional selective breeding for rapid (RHA) vs. extremely poor (RLA) two-way active avoidance acquisition. Compared with RLAs and other rat strains/stocks, RHAs are characterized by increased impulsivity, deficits in social behavior, novelty-induced hyper-locomotion, impaired attentional/cognitive abilities, vulnerability to psychostimulant sensitization and drug addiction. RHA rats also exhibit decreased function of the prefrontal cortex (PFC) and hippocampus, increased functional activity of the mesolimbic dopamine system and a dramatic deficit of central metabotropic glutamate-2 (mGlu2) receptors (due to a stop codon mutation at cysteine 407 in Grm2 -cys407*-), along with increased density of 5-HT2A receptors in the PFC, alterations of several synaptic markers and increased density of pyramidal "thin" (immature) dendrític spines in the PFC. These characteristics suggest an immature brain of RHA rats, and are reminiscent of schizophrenia features like hypofrontality and disruption of the excitation/inhibition cortical balance. RHA rats represent a promising heuristic model of neurodevelopmental schizophrenia-relevant features and comorbidity with drug addiction vulnerability.
Collapse
Affiliation(s)
- Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | | | - Carles Tapias-Espinosa
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Daniel Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Ana Sánchez-González
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Francesco Sanna
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy
| | - Rafael Torrubia
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | - Ignacio Morón
- Department of Psychobiology and Centre of Investigation of Mind, Brain, and Behaviour (CIMCYC), University of Granada, Spain
| | - Carmen Torres
- Department of Psychology, University of Jaén, 23071, Jaén, Spain.
| | - Susana Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400, Copenhagen, Denmark.
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Maria G Corda
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy.
| | - Osvaldo Giorgi
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy.
| |
Collapse
|
29
|
Connecting the Neurobiology of Developmental Brain Injury: Neuronal Arborisation as a Regulator of Dysfunction and Potential Therapeutic Target. Int J Mol Sci 2021; 22:ijms22158220. [PMID: 34360985 PMCID: PMC8348801 DOI: 10.3390/ijms22158220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Neurodevelopmental disorders can derive from a complex combination of genetic variation and environmental pressures on key developmental processes. Despite this complex aetiology, and the equally complex array of syndromes and conditions diagnosed under the heading of neurodevelopmental disorder, there are parallels in the neuropathology of these conditions that suggest overlapping mechanisms of cellular injury and dysfunction. Neuronal arborisation is a process of dendrite and axon extension that is essential for the connectivity between neurons that underlies normal brain function. Disrupted arborisation and synapse formation are commonly reported in neurodevelopmental disorders. Here, we summarise the evidence for disrupted neuronal arborisation in these conditions, focusing primarily on the cortex and hippocampus. In addition, we explore the developmentally specific mechanisms by which neuronal arborisation is regulated. Finally, we discuss key regulators of neuronal arborisation that could link to neurodevelopmental disease and the potential for pharmacological modification of arborisation and the formation of synaptic connections that may provide therapeutic benefit in the future.
Collapse
|
30
|
Navarro-Gonzalez C, Carceller H, Benito Vicente M, Serra I, Navarrete M, Domínguez-Canterla Y, Rodríguez-Prieto Á, González-Manteiga A, Fazzari P. Nrg1 haploinsufficiency alters inhibitory cortical circuits. Neurobiol Dis 2021; 157:105442. [PMID: 34246770 DOI: 10.1016/j.nbd.2021.105442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 05/19/2021] [Accepted: 06/30/2021] [Indexed: 11/19/2022] Open
Abstract
Neuregulin 1 (NRG1) and its receptor ERBB4 are schizophrenia (SZ) risk genes that control the development of both excitatory and inhibitory cortical circuits. Most studies focused on the characterization ErbB4 deficient mice. However, ErbB4 deletion concurrently perturbs the signaling of Nrg1 and Neuregulin 3 (Nrg3), another ligand expressed in the cortex. In addition, NRG1 polymorphisms linked to SZ locate mainly in non-coding regions and they may partially reduce Nrg1 expression. Here, to study the relevance of Nrg1 partial loss-of-function in cortical circuits we characterized a recently developed haploinsufficient mouse model of Nrg1 (Nrg1tm1Lex). These mice display SZ-like behavioral deficits. The cellular and molecular underpinnings of the behavioral deficits in Nrg1tm1Lex mice remain to be established. With multiple approaches including Magnetic Resonance Spectroscopy (MRS), electrophysiology, quantitative imaging and molecular analysis we found that Nrg1 haploinsufficiency impairs the inhibitory cortical circuits. We observed changes in the expression of molecules involved in GABAergic neurotransmission, decreased density of Vglut1 excitatory buttons onto Parvalbumin interneurons and decreased frequency of spontaneous inhibitory postsynaptic currents. Moreover, we found a decreased number of Parvalbumin positive interneurons in the cortex and altered expression of Calretinin. Interestingly, we failed to detect other alterations in excitatory neurons that were previously reported in ErbB4 null mice suggesting that the Nrg1 haploinsufficiency does not entirely phenocopies ErbB4 deletions. Altogether, this study suggests that Nrg1 haploinsufficiency primarily affects the cortical inhibitory circuits in the cortex and provides new insights into the structural and molecular synaptic impairment caused by NRG1 hypofunction in a preclinical model of SZ.
Collapse
Affiliation(s)
- Carmen Navarro-Gonzalez
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Héctor Carceller
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Marina Benito Vicente
- Laboratorio Resonancia Magnética de Investigación, Hospital Nacional de Parapléjicos, Toledo, Spain.
| | - Irene Serra
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto Cajal, Madrid, Spain.
| | - Marta Navarrete
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto Cajal, Madrid, Spain.
| | - Yaiza Domínguez-Canterla
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Ángela Rodríguez-Prieto
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Ana González-Manteiga
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Pietro Fazzari
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain; Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa, Madrid, Spain.
| |
Collapse
|
31
|
Extreme Glycemic Fluctuations Debilitate NRG1, ErbB Receptors and Olig1 Function: Association with Regeneration, Cognition and Mood Alterations During Diabetes. Mol Neurobiol 2021; 58:4727-4744. [PMID: 34165684 DOI: 10.1007/s12035-021-02455-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/16/2021] [Indexed: 12/28/2022]
Abstract
Neuronal regeneration is crucial for maintaining intact neural interactions for perpetuation of cognitive and emotional functioning. The NRG1-ErbB receptor signaling is a key pathway for regeneration in adult brain and also associated with learning and mood stabilization by modulating synaptic transmission. Extreme glycemic stress is known to affect NRG1-ErbB-mediated regeneration in brain; yet, it remains unclear how the ErbB receptor subtypes are differentially affected due to such metabolic variations. Here, we assessed the alterations in NRG1, ErbB receptor subtypes to study the regenerative potential, both in rodents as well as in neuronal and glial cell models of hyperglycemia and hypoglycemic insults during hyperglycemia. The pro-oxidant and anti-oxidant status leading to degenerative changes in brain regions were determined. The spatial memory and anxiogenic behaviour of experimental rodents were tested using 'T' maze and Elevated Plus Maze. Our data revealed that the extreme glycemic discrepancies during diabetes and recurrent hypoglycemia lead to altered expression of NRG1, ErbB receptor subtypes, Syntaxin1 and Olig1 that shows association with impaired regeneration, synaptic dysfunction, demyelination, cognitive deficits and anxiety.
Collapse
|
32
|
Yoo JY, Kim HB, Baik TK, Lee JH, Woo RS. Neuregulin 1/ErbB4/Akt signaling attenuates cytotoxicity mediated by the APP-CT31 fragment of amyloid precursor protein. Exp Mol Pathol 2021; 120:104622. [PMID: 33684392 DOI: 10.1016/j.yexmp.2021.104622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Han-Byeol Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon 34520, Republic of Korea.
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea.
| |
Collapse
|
33
|
Sánchez-González A, Thougaard E, Tapias-Espinosa C, Cañete T, Sampedro-Viana D, Saunders JM, Toneatti R, Tobeña A, Gónzalez-Maeso J, Aznar S, Fernández-Teruel A. Increased thin-spine density in frontal cortex pyramidal neurons in a genetic rat model of schizophrenia-relevant features. Eur Neuropsychopharmacol 2021; 44:79-91. [PMID: 33485732 PMCID: PMC7902438 DOI: 10.1016/j.euroneuro.2021.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/09/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
The cellular mechanisms altered during brain wiring leading to cognitive disturbances in neurodevelopmental disorders remain unknown. We have previously reported altered cortical expression of neurodevelopmentally regulated synaptic markers in a genetic animal model of schizophrenia-relevant behavioral features, the Roman-High Avoidance rat strain (RHA-I). To further explore this phenotype, we looked at dendritic spines in cortical pyramidal neurons, as changes in spine density and morphology are one of the main processes taking place during adolescence. An HSV-viral vector carrying green fluorescent protein (GFP) was injected into the frontal cortex (FC) of a group of 11 RHA-I and 12 Roman-Low Avoidance (RLA-I) male rats. GFP labeled dendrites from pyramidal cells were 3D reconstructed and number and types of spines quantified. We observed an increased spine density in the RHA-I, corresponding to a larger fraction of immature thin spines, with no differences in stubby and mushroom spines. Glia cells, parvalbumin (PV) and somatostatin (SST) interneurons and surrounding perineuronal net (PNN) density are known to participate in FC and pyramidal neuron dendritic spine maturation. We determined by stereological-based quantification a significantly higher number of GFAP-positive astrocytes in the FC of the RHA-I strain, with no difference in microglia (Iba1-positive cells). The number of inhibitory PV, SST interneurons or PNN density, on the contrary, was unchanged. Results support our belief that the RHA-I strain presents a more immature FC, with some structural features like those observed during adolescence, adding construct validity to this strain as a genetic behavioral model of neurodevelopmental disorders.
Collapse
Affiliation(s)
- A Sánchez-González
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - E Thougaard
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400 Copenhagen, Denmark
| | - C Tapias-Espinosa
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - T Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - D Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - J M Saunders
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - R Toneatti
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - A Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - J Gónzalez-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - S Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400 Copenhagen, Denmark; Copenhagen Center for Translational Research, Bispebjerg Copenhagen University Hospital, Copenhagen, Denmark.
| | - A Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
34
|
Seo HJ, Park JE, Choi SM, Kim T, Cho SH, Lee KH, Song WK, Song J, Jeong HS, Kim DH, Kim BC. Inhibitory Neural Network's Impairments at Hippocampal CA1 LTP in an Aged Transgenic Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22020698. [PMID: 33445678 PMCID: PMC7828160 DOI: 10.3390/ijms22020698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a rapid accumulation of amyloid β (Aβ) protein in the hippocampus, which impairs synaptic structures and neuronal signal transmission, induces neuronal loss, and diminishes memory and cognitive functions. The present study investigated the impact of neuregulin 1 (NRG1)-ErbB4 signaling on the impairment of neural networks underlying hippocampal long-term potentiation (LTP) in 5xFAD mice, a model of AD with greater symptom severity than that of TG2576 mice. Specifically, we observed parvalbumin (PV)-containing hippocampal interneurons, the effect of NRG1 on hippocampal LTP, and the functioning of learning and memory. We found a significant decrease in the number of PV interneurons in 11-month-old 5xFAD mice. Moreover, synaptic transmission in the 5xFAD mice decreased at 6 months of age. The 11-month-old transgenic AD mice showed fewer inhibitory PV neurons and impaired NRG1-ErbB4 signaling than did wild-type mice, indicating that the former exhibit the impairment of neuronal networks underlying LTP in the hippocampal Schaffer-collateral pathway. In conclusion, this study confirmed the impaired LTP in 5xFAD mice and its association with aberrant NRG1-ErbB signaling in the neuronal network.
Collapse
Affiliation(s)
- Hyeon Jeong Seo
- Department of Biomedical Sciences, Graduate School, Chonnam National University, Gwangju 61186, Korea;
| | - Jung Eun Park
- Department of Biomedical Science, College of Natural Sciences, Chosun University, Gwangju 61452, Korea;
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Seong-Min Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Taekyoung Kim
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School & Hwasun Hospital, Hwasun 58128, Korea;
| | - Woo Keun Song
- Cell Logistics and Silver Health Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Dong Hyun Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49236, Korea
- Correspondence: (D.H.K.); (B.C.K.); Tel.: +82-51-200-7583 (D.H.K.); +82-62-220-6123 (B.C.K.)
| | - Byeong C. Kim
- Department of Biomedical Sciences, Graduate School, Chonnam National University, Gwangju 61186, Korea;
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
- Correspondence: (D.H.K.); (B.C.K.); Tel.: +82-51-200-7583 (D.H.K.); +82-62-220-6123 (B.C.K.)
| |
Collapse
|
35
|
Ou GY, Lin WW, Zhao WJ. Neuregulins in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:662474. [PMID: 33897409 PMCID: PMC8064692 DOI: 10.3389/fnagi.2021.662474] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), are typically characterized by progressive neuronal loss and neurological dysfunctions in the nervous system, affecting both memory and motor functions. Neuregulins (NRGs) belong to the epidermal growth factor (EGF)-like family of extracellular ligands and they play an important role in the development, maintenance, and repair of both the central nervous system (CNS) and peripheral nervous system (PNS) through the ErbB signaling pathway. They also regulate multiple intercellular signal transduction and participate in a wide range of biological processes, such as differentiation, migration, and myelination. In this review article, we summarized research on the changes and roles of NRGs in neurodegenerative diseases, especially in AD. We elaborated on the structural features of each NRG subtype and roles of NRG/ErbB signaling networks in neurodegenerative diseases. We also discussed the therapeutic potential of NRGs in the symptom remission of neurodegenerative diseases, which may offer hope for advancing related treatment.
Collapse
Affiliation(s)
- Guan-yong Ou
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wen-wen Lin
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Wei-jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Wei-jiang Zhao
| |
Collapse
|
36
|
Zhang KX, Zhao JJ, Chai W, Chen JY. Synaptic remodeling in mouse motor cortex after spinal cord injury. Neural Regen Res 2021; 16:744-749. [PMID: 33063737 PMCID: PMC8067930 DOI: 10.4103/1673-5374.295346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury dramatically blocks information exchange between the central nervous system and the peripheral nervous system. The resulting fate of synapses in the motor cortex has not been well studied. To explore synaptic reorganization in the motor cortex after spinal cord injury, we established mouse models of T12 spinal cord hemi-section and then monitored the postsynaptic dendritic spines and presynaptic axonal boutons of pyramidal neurons in the hindlimb area of the motor cortex in vivo. Our results showed that spinal cord hemi-section led to the remodeling of dendritic spines bilaterally in the motor cortex and the main remodeling regions changed over time. It made previously stable spines unstable and eliminated spines more unlikely to be re-emerged. There was a significant increase in new spines in the contralateral motor cortex. However, the low survival rate of the new spines demonstrated that new spines were still fragile. Observation of presynaptic axonal boutons found no significant change. These results suggest the existence of synapse remodeling in motor cortex after spinal cord hemi-section and that spinal cord hemi-section affected postsynaptic dendritic spines rather than presynaptic axonal boutons. This study was approved by the Ethics Committee of Chinese PLA General Hospital, China (approval No. 201504168S) on April 16, 2015.
Collapse
Affiliation(s)
- Ke-Xue Zhang
- Department of Pediatric Surgery, Chinese PLA General Hospital, Beijing, China
| | - Jia-Jia Zhao
- Department of Anesthesiology, Shunyi District Hospital, Beijing, China
| | - Wei Chai
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ji-Ying Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
37
|
Colàs-Campàs L, Farre J, Mauri-Capdevila G, Molina-Seguín J, Aymerich N, Ois Á, Roquer J, Tur S, García-Carreira MDC, Martí-Fàbregas J, Cruz-Culebras A, Segura T, Arque G, Purroy F. Inflammatory Response of Ischemic Tolerance in Circulating Plasma: Preconditioning-Induced by Transient Ischemic Attack (TIA) Phenomena in Acute Ischemia Patients (AIS). Front Neurol 2020; 11:552470. [PMID: 33192985 PMCID: PMC7658473 DOI: 10.3389/fneur.2020.552470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/30/2020] [Indexed: 11/15/2022] Open
Abstract
Introduction: Ischemic tolerance (IT) refers to a state where cells are resistant to the damaging effects caused by periods of ischemia. In a clinical scenario, the IT phenomenon would be activated by a recent transient ischemic attack (TIA) before an ischemic stroke (IS). The characterization of inflammatory protein expression patterns will contribute to improved understanding of IT. Methods: A total of 477 IS patients from nine hospitals, recruited between January 2011 and January 2016, were included in the current study and divided in three groups: 438 (91.9%) patients without previous TIA (group 1), 22 (4.6%) patients who suffered TIA 24 h before IS (group 2), and 17 (3.5%) patients who suffered TIA between 24 h and 7 days prior to IS (group 3). An inflammatory biomarker panel (IL-6, NT-proBNP, hsCRP, hs-Troponin, NSE, and S-100b) on plasma and a cytokine antibody array was performed to achieve the preconditioning signature potentially induced by TIA phenomena. Primary outcome was modified rankin scale (mRs) score at 90 days. Results: Recent previous TIA was associated with better clinical outcome at 90 days (median mRS of group 1: 2.0 [1.0–4.0]; group 2: 2.0 [0.0–3.0]; group 3: 1.0 [0–2.5]; p = 0.086) and smaller brain lesion (group 1: 3.7 [0.7–18.3]; group 2: 0.8 [0.3–8.9]; group 3: 0.6 [0.1–5.5] mL; p = 0.006). All inflammation biomarkers were down regulated in the groups of recent TIA prior to IS compared to those who did not suffer a TIA events. Moreover, a cytokine antibody array revealed 30 differentially expressed proteins between the three groups. Among them, HRG1-alpha (Fold change 74.4 between group 1 and 2; 74.2 between group 1 and 3) and MAC-1 (Fold change 0.05 between group 1 and 2; 0.06 between group 1 and 3) expression levels would better stratify patients with TIA 7 days before IS. These two proteins showed an earlier inflammation profile that was not detectable by the biomarker panel. Conclusion: Inflammatory pathways were activated by transient ischemic attack, however the period of time between this event and a further ischemic stroke could be determined by a protein signature that would contribute to define the role of ischemic tolerance induced by TIA.
Collapse
Affiliation(s)
- Laura Colàs-Campàs
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Joan Farre
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Medical Laboratory, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Gerard Mauri-Capdevila
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Jessica Molina-Seguín
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | | | | | | | - Silvia Tur
- Hospital Son Espases, Palma de Mallorca, Spain
| | | | | | | | - Tomás Segura
- Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Gloria Arque
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Francisco Purroy
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
38
|
Yang JM, Shen CJ, Chen XJ, Kong Y, Liu YS, Li XW, Chen Z, Gao TM, Li XM. erbb4 Deficits in Chandelier Cells of the Medial Prefrontal Cortex Confer Cognitive Dysfunctions: Implications for Schizophrenia. Cereb Cortex 2020; 29:4334-4346. [PMID: 30590426 DOI: 10.1093/cercor/bhy316] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/18/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022] Open
Abstract
erbb4 is a known susceptibility gene for schizophrenia. Chandelier cells (ChCs, also known as axo-axonic cells) are a distinct GABAergic interneuron subtype that exclusively target the axonal initial segment, which is the site of pyramidal neuron action potential initiation. ChCs are a source of ErbB4 expression and alterations in ChC-pyramidal neuron connectivity occur in the medial prefrontal cortex (mPFC) of schizophrenic patients and animal models of schizophrenia. However, the contribution of ErbB4 in mPFC ChCs to the pathogenesis of schizophrenia remains unknown. By conditional deletion or knockdown of ErbB4 from mPFC ChCs, we demonstrated that ErbB4 deficits led to impaired ChC-pyramidal neuron connections and cognitive dysfunctions. Furthermore, the cognitive dysfunctions were normalized by L-838417, an agonist of GABAAα2 receptors enriched in the axonal initial segment. Given that cognitive dysfunctions are a core symptom of schizophrenia, our results may provide a new perspective for understanding the etiology of schizophrenia and suggest that GABAAα2 receptors may be potential pharmacological targets for its treatment.
Collapse
Affiliation(s)
- Jian-Ming Yang
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chen-Jie Shen
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Juan Chen
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Kong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Si Liu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Li
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhong Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tian-Ming Gao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Ming Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Stock R, Jeckel P, Kraushaar U, Wüst R, Fallgatter A, Volkmer H. The potential of induced pluripotent stem cells for discriminating neurodevelopmental disorders. Stem Cells Transl Med 2020; 10:50-56. [PMID: 32864861 PMCID: PMC7780807 DOI: 10.1002/sctm.20-0206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
Studying human disease‐specific processes and mechanisms in vitro is limited by a lack of valid human test systems. Induced pluripotent stem cells (iPSCs) evolve as an important and promising tool to better understand the molecular pathology of neurodevelopmental disorders. Patient‐derived iPSCs enable analysis of unique disease mechanisms and may also serve for preclinical drug development. Here, we review the current knowledge on iPSC models for schizophrenia and autism spectrum disorders with emphasis on the discrimination between them. It appears that transcriptomic analyses and functional read‐outs are the most promising approaches to uncover specific disease mechanisms in vitro.
Collapse
Affiliation(s)
- Ricarda Stock
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Pauline Jeckel
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Udo Kraushaar
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, University of Tübingen, Tübingen, Germany
| | | | - Hansjürgen Volkmer
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
40
|
Schoonover KE, Dienel SJ, Lewis DA. Prefrontal cortical alterations of glutamate and GABA neurotransmission in schizophrenia: Insights for rational biomarker development. Biomark Neuropsychiatry 2020; 3. [PMID: 32656540 PMCID: PMC7351254 DOI: 10.1016/j.bionps.2020.100015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Certain cognitive deficits in schizophrenia, such as impaired working memory, are thought to reflect alterations in the neural circuitry of the dorsolateral prefrontal cortex (DLPFC). Gamma oscillations in the DLPFC appear to be a neural corollary of working memory function, and the power of these oscillations during working memory tasks is lower in individuals with schizophrenia. Thus, gamma oscillations represent a potentially useful biomarker to index dysfunction in the DLPFC circuitry responsible for working memory in schizophrenia. Postmortem studies, by identifying the cellular basis of DLPFC dysfunction, can help inform the utility of biomarker measures obtained in vivo. Given that gamma oscillations reflect network activity of excitatory pyramidal neurons and inhibitory GABA neurons, we review postmortem findings of alterations to both cell types in the DLPFC and discuss how these findings might inform future biomarker development and use.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States
| | - Samuel J Dienel
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States.,Medical Scientist Training Program, University of Pittsburgh, United States.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.,Center for the Neural Basis of Cognition, Carnegie Mellon University, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.,Center for the Neural Basis of Cognition, Carnegie Mellon University, United States
| |
Collapse
|
41
|
Sokolenko E, Nithianantharajah J, Jones NC. MK-801 impairs working memory on the Trial-Unique Nonmatch-to-Location test in mice, but this is not exclusively mediated by NMDA receptors on PV+ interneurons or forebrain pyramidal cells. Neuropharmacology 2020; 171:108103. [DOI: 10.1016/j.neuropharm.2020.108103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/11/2020] [Accepted: 04/06/2020] [Indexed: 01/13/2023]
|
42
|
Kalev-Zylinska ML, Hearn JI, Makhro A, Bogdanova A. N-Methyl-D-Aspartate Receptors in Hematopoietic Cells: What Have We Learned? Front Physiol 2020; 11:577. [PMID: 32625106 PMCID: PMC7311790 DOI: 10.3389/fphys.2020.00577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/08/2020] [Indexed: 12/24/2022] Open
Abstract
The N-methyl-D-aspartate receptor (NMDAR) provides a pathway for glutamate-mediated inter-cellular communication, best known for its role in the brain but with multiple examples of functionality in non-neuronal cells. Data previously published by others and us provided ex vivo evidence that NMDARs regulate platelet and red blood cell (RBC) production. Here, we summarize what is known about these hematopoietic roles of the NMDAR. Types of NMDAR subunits expressed in megakaryocytes (platelet precursors) and erythroid cells are more commonly found in the developing rather than adult brain, suggesting trophic functions. Nevertheless, similar to their neuronal counterparts, hematopoietic NMDARs function as ion channels, and are permeable to calcium ions (Ca2+). Inhibitors that block open NMDAR (memantine and MK-801) interfere with megakaryocytic maturation and proplatelet formation in primary culture. The effect on proplatelet formation appears to involve Ca2+ influx-dependent regulation of the cytoskeletal remodeling. In contrast to normal megakaryocytes, NMDAR effects in leukemic Meg-01 cells are diverted away from differentiation to increase proliferation. NMDAR hypofunction triggers differentiation of Meg-01 cells with the bias toward erythropoiesis. The underlying mechanism involves changes in the intracellular Ca2+ homeostasis, cell stress pathways, and hematopoietic transcription factors that upon NMDAR inhibition shift from the predominance of megakaryocytic toward erythroid regulators. This ability of NMDAR to balance both megakaryocytic and erythroid cell fates suggests receptor involvement at the level of a bipotential megakaryocyte-erythroid progenitor. In human erythroid precursors and circulating RBCs, NMDAR regulates intracellular Ca2+ homeostasis. NMDAR activity supports survival of early proerythroblasts, and in mature RBCs NMDARs impact cellular hydration state, hemoglobin oxygen affinity, and nitric oxide synthase activity. Overexcitation of NMDAR in mature RBCs leads to Ca2+ overload, K+ loss, RBC dehydration, and oxidative stress, which may contribute to the pathogenesis of sickle cell disease. In summary, there is growing evidence that glutamate-NMDAR signaling regulates megakaryocytic and erythroid cells at different stages of maturation, with some intriguing differences emerging in NMDAR expression and function between normal and diseased cells. NMDAR signaling may provide new therapeutic opportunities in hematological disease, but in vivo applicability needs to be confirmed.
Collapse
Affiliation(s)
- Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Pathology and Laboratory Medicine, LabPlus Haematology, Auckland City Hospital, Auckland, New Zealand
| | - James I. Hearn
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Asya Makhro
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
43
|
Shi L, Bergson CM. Neuregulin 1: an intriguing therapeutic target for neurodevelopmental disorders. Transl Psychiatry 2020; 10:190. [PMID: 32546684 PMCID: PMC7297728 DOI: 10.1038/s41398-020-00868-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/14/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodevelopmental psychiatric disorders including schizophrenia (Sz) and attention deficit hyperactivity disorder (ADHD) are chronic mental illnesses, which place costly and painful burdens on patients, their families and society. In recent years, the epidermal growth factor (EGF) family member Neuregulin 1 (NRG1) and one of its receptors, ErbB4, have received considerable attention due to their regulation of inhibitory local neural circuit mechanisms important for information processing, attention, and cognitive flexibility. Here we examine an emerging body of work indicating that either decreasing NRG1-ErbB4 signaling in fast-spiking parvalbumin positive (PV+) interneurons or increasing it in vasoactive intestinal peptide positive (VIP+) interneurons could reactivate cortical plasticity, potentially making it a future target for gene therapy in adults with neurodevelopmental disorders. We propose preclinical studies to explore this model in prefrontal cortex (PFC), but also review the many challenges in pursuing cell type and brain-region-specific therapeutic approaches for the NRG1 system.
Collapse
Affiliation(s)
- Liang Shi
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Boulevard, Augusta, GA 30912 USA ,grid.189967.80000 0001 0941 6502Present Address: Department of Cell Biology, Emory University School of Medicine, Atlanta, GA USA
| | - Clare M. Bergson
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Boulevard, Augusta, GA 30912 USA
| |
Collapse
|
44
|
Neuregulins 1, 2, and 3 Promote Early Neurite Outgrowth in ErbB4-Expressing Cortical GABAergic Interneurons. Mol Neurobiol 2020; 57:3568-3588. [PMID: 32542595 DOI: 10.1007/s12035-020-01966-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022]
Abstract
The neuregulins (Nrgs 1-4) are a family of signaling molecules that play diverse roles in the nervous system. Nrg1 has been implicated in the formation of synapses and in synaptic plasticity. Previous studies have shown Nrg1 can affect neurite outgrowth in several neuronal populations, while the role of Nrg2 and Nrg3 in this process has remained understudied. The Nrgs can bind and activate the ErbB4 receptor tyrosine kinase which is preferentially expressed in GABAergic interneurons in the rodent hippocampus and cerebral cortex. In the present study, we evaluated the effects of Nrgs 1, 2, and 3 on neurite outgrowth of dissociated rat cortical ErbB4-positive (+)/GABA+ interneurons in vitro. All three Nrgs were able to promote neurite outgrowth during the first 2 days in vitro, with increases detected for both the axon (116-120%) and other neurites (100-120%). Increases in the average number of primary and secondary neurites were also observed. Treatment with the Nrgs for an additional 3 days promoted an increase in axonal length (86-96%), with only minimal effects on the remaining neurites (8-13%). ErbB4 expression persisted throughout the dendritic arbor and cell soma at all stages examined, while its expression in the axon was transient and declined with cell maturation. ErbB4 overexpression in GABAergic neurons promoted neurite outgrowth, an effect that was potentiated by Nrg treatment. These results show that Nrgs 1, 2, and 3 are each capable of influencing dendritic and axonal growth at early developmental stages in GABAergic neurons grown in vitro.
Collapse
|
45
|
Paterson C, Cumming B, Law AJ. Temporal Dynamics of the Neuregulin-ErbB Network in the Murine Prefrontal Cortex across the Lifespan. Cereb Cortex 2020; 30:3325-3339. [PMID: 31897479 DOI: 10.1093/cercor/bhz312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Neuregulin-ErbB signaling is essential for numerous functions in the developing, adult, and aging brain, particularly in the prefrontal cortex (PFC). Mouse models with disrupted Nrg and/or ErbB genes are relevant to psychiatric, developmental, and age-related disorders, displaying a range of abnormalities stemming from cortical circuitry impairment. Many of these models display nonoverlapping phenotypes dependent upon the gene target and timing of perturbation, suggesting that cortical expression of the Nrg-ErbB network undergoes temporal regulation across the lifespan. Here, we report a comprehensive temporal expression mapping study of the Nrg-ErbB signaling network in the mouse PFC across postnatal development through aging. We find that Nrg and ErbB genes display distinct expression profiles; moreover, splice isoforms of these genes are differentially expressed across the murine lifespan. We additionally find a developmental switch in ErbB4 splice isoform expression potentially mediated through coregulation of the lncRNA Miat expression. Our results are the first to comprehensively and quantitatively map the expression patterns of the Nrg-ErbB network in the mouse PFC across the postnatal lifespan and may help disentangle the pathway's involvement in normal cortical sequences of events across the lifespan, as well as shedding light on the pathophysiological mechanisms of abnormal Nrg-ErbB signaling in neurological disease.
Collapse
Affiliation(s)
- Clare Paterson
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooke Cumming
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amanda J Law
- Department of Psychiatry, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Cell and Developmental Biology, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Medicine, University of Colorado, School of Medicine Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
46
|
Interneuron NMDA Receptor Ablation Induces Hippocampus-Prefrontal Cortex Functional Hypoconnectivity after Adolescence in a Mouse Model of Schizophrenia. J Neurosci 2020; 40:3304-3317. [PMID: 32205341 DOI: 10.1523/jneurosci.1897-19.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 12/13/2022] Open
Abstract
Although the etiology of schizophrenia is still unknown, it is accepted to be a neurodevelopmental disorder that results from the interaction of genetic vulnerabilities and environmental insults. Although schizophrenia's pathophysiology is still unclear, postmortem studies point toward a dysfunction of cortical interneurons as a central element. It has been suggested that alterations in parvalbumin-positive interneurons in schizophrenia are the consequence of a deficient signaling through NMDARs. Animal studies demonstrated that early postnatal ablation of the NMDAR in corticolimbic interneurons induces neurobiochemical, physiological, behavioral, and epidemiological phenotypes related to schizophrenia. Notably, the behavioral abnormalities emerge only after animals complete their maturation during adolescence and are absent if the NMDAR is deleted during adulthood. This suggests that interneuron dysfunction must interact with development to impact on behavior. Here, we assess in vivo how an early NMDAR ablation in corticolimbic interneurons impacts on mPFC and ventral hippocampus functional connectivity before and after adolescence. In juvenile male mice, NMDAR ablation results in several pathophysiological traits, including increased cortical activity and decreased entrainment to local gamma and distal hippocampal theta rhythms. In addition, adult male KO mice showed reduced ventral hippocampus-mPFC-evoked potentials and an augmented low-frequency stimulation LTD of the pathway, suggesting that there is a functional disconnection between both structures in adult KO mice. Our results demonstrate that early genetic abnormalities in interneurons can interact with postnatal development during adolescence, triggering pathophysiological mechanisms related to schizophrenia that exceed those caused by NMDAR interneuron hypofunction alone.SIGNIFICANCE STATEMENT NMDAR hypofunction in cortical interneurons has been linked to schizophrenia pathophysiology. How a dysfunction of GABAergic cortical interneurons interacts with maturation during adolescence has not been clarified yet. Here, we demonstrate in vivo that early postnatal ablation of the NMDAR in corticolimbic interneurons results in an overactive but desynchronized PFC before adolescence. Final postnatal maturation during this stage outspreads the impact of the genetic manipulation toward a functional disconnection of the ventral hippocampal-prefrontal pathway, probably as a consequence of an exacerbated propensity toward hippocampal-evoked depotentiation plasticity. Our results demonstrate a complex interaction between genetic and developmental factors affecting cortical interneurons and PFC function.
Collapse
|
47
|
Zhang YM, Yang ZD, Yu YF. Effect of neuregulin-1 on the auditory cortex in adult C57BL/6J mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:362-367. [PMID: 32440323 PMCID: PMC7229506 DOI: 10.22038/ijbms.2020.33656.8026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 07/27/2019] [Indexed: 01/23/2023]
Abstract
OBJECTIVES We sought to explore whether neuregulin-1(NRG1) would have a protective effect on the auditory cortices of adult C57BL/6J mice. MATERIALS AND METHODS We used RTPCR and Western blot (WB) to detect the expression of NRG1 and ERBB4 (the receptor of NRG1) in the auditory cortices of C57BL/6J mice of different ages (6-8 weeks and 42-44 weeks). Three groups of 42-44 week-old C57BL/6J mice were intraperitoneally injected with mouse neurotrophic factor (m-NGF), NRG1, or saline for two months. We observed the ultrastructures of the auditory cortices of adult mice after treatment using transmission electron microscopy. Additionally, we observed expression of NRG1 in the auditory cortices by immunohistochemistry. RESULTS Expression of NRG1 and ERBB4 in the auditory cortices of C57BL/6J mice at the age of 42-44 weeks was lower compared with 6-8 week-old mice. The ultra-structures of the auditory cortices, including the neurons and myelin sheaths, as revealed by transmission electron microscopy were healthier in the m-NGF and NRG1 treatment groups than those in the saline group. We found that expression of NRG1 in the auditory cortices after treatment in the m-NGF and NRG1 groups, especially in the NRG1 group, was higher than that in the saline group. CONCLUSION We concluded that with increasing age, NRG1 in the auditory cortices of C57BL/6J mice gradually decreased, and that NRG1 had a protective effect on the auditory cortices in adult C57BL/J mice.
Collapse
Affiliation(s)
- Yun-Mei Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhen-Dong Yang
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ya-Feng Yu
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
48
|
Xie R, Hong S, Ye Y, Wang X, Chen F, Yang L, Yan Y, Liao L. Ketamine Affects the Expression of ErbB4 in the Hippocampus and Prefrontal Cortex of Rats. J Mol Neurosci 2020; 70:962-967. [PMID: 32096126 DOI: 10.1007/s12031-020-01502-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a severe chronic neuropsychiatric disorder, and its exact pathogenesis remains unclear. This study investigated the effect of ketamine on the expression of ErbB4 (considered a schizophrenia candidate gene) in the hippocampus and prefrontal cortex of rats. Rats were randomly divided into four groups: control, low-dose, medium-dose and high-dose groups. The low-dose, medium-dose and high-dose groups were intraperitoneally injected with 15 mg/kg, 30 mg/kg and 60 mg/kg ketamine, respectively, twice a day (9:00 a.m. and 9:00 p.m.); the control group was administered normal saline. The treatment lasted 7 days. After treatment, rats were euthanized, and their brain tissues were collected and then analyzed by immunohistochemistry. The results of immunohistochemistry staining demonstrated that the ErbB4 protein was expressed exclusively in the CA3 region of the hippocampus and the Cg1 region of the prefrontal cortex. Ketamine administration significantly decreased the expression of ErbB4 in a dose-dependent manner. The high-dose ketamine treatment was found to be optimal for establishing a rat model for schizophrenia. Ketamine induced symptoms similar to schizophrenia in humans. The ketamine-induced rat model for schizophrenia constructed in this study provides novel insights to better understand the pathogenic mechanisms of schizophrenia and aid in drug discovery.
Collapse
Affiliation(s)
- Runfang Xie
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, People's Republic of China
| | - Shijun Hong
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, People's Republic of China
| | - Yi Ye
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xueyan Wang
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fan Chen
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lin Yang
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Youyi Yan
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Linchuan Liao
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
49
|
Transcriptome analysis of fibroblasts from schizophrenia patients reveals differential expression of schizophrenia-related genes. Sci Rep 2020; 10:630. [PMID: 31959813 PMCID: PMC6971273 DOI: 10.1038/s41598-020-57467-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/19/2019] [Indexed: 01/05/2023] Open
Abstract
Schizophrenia is a complex neurodevelopmental disorder with high rate of morbidity and mortality. While the heritability rate is high, the precise etiology is still unknown. Although schizophrenia is a central nervous system disorder, studies using peripheral tissues have also been established to search for patient specific biomarkers and to increase understanding of schizophrenia etiology. Among all peripheral tissues, fibroblasts stand out as they are easy to obtain and culture. Furthermore, they keep genetic stability for long period and exhibit molecular similarities to cells from nervous system. Using a unique set of fibroblast samples from a genetically isolated population in northern Sweden, we performed whole transcriptome sequencing to compare differentially expressed genes in seven controls and nine patients. We found differential fibroblast expression between cases and controls for 48 genes, including eight genes previously implicated in schizophrenia or schizophrenia related pathways; HGF, PRRT2, EGR1, EGR3, C11orf87, TLR3, PLEKHH2 and PIK3CD. Weighted gene correlation network analysis identified three differentially co-expressed networks of genes significantly-associated with schizophrenia. All three modules were significantly suppressed in patients compared to control, with one module highly enriched in genes involved in synaptic plasticity, behavior and synaptic transmission. In conclusion, our results support the use of fibroblasts for identification of differentially expressed genes in schizophrenia and highlight dysregulation of synaptic networks as an important mechanism in schizophrenia.
Collapse
|
50
|
Yoo JY, Kim HB, Yoo SY, Yoo HI, Song DY, Baik TK, Lee JH, Woo RS. Neuregulin 1/ErbB4 signaling attenuates neuronal cell damage under oxygen-glucose deprivation in primary hippocampal neurons. Anat Cell Biol 2019; 52:462-468. [PMID: 31949986 PMCID: PMC6952697 DOI: 10.5115/acb.19.210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/25/2022] Open
Abstract
The hippocampus is one of the most important brain areas of cognition. This region is particularly sensitive to hypoxia and ischemia. Neuregulin-1 (NRG1) has been shown to be able to protect against focal cerebral ischemia. The aim of the present study was to investigate the neuroprotective effect of NRG1 in primary hippocampal neurons and its underlying mechanism. Our data showed oxygen-glucose deprivation (OGD)-induced cytotoxicity and overexpression of ErbB4 in primary hippocampal neurons. Moreover, pretreatment with NRG1 could inhibit OGD-induced overexpression of ErbB4. In addition, NRG1 significantly attenuated neuronal death induced by OGD. The neuroprotective effect of NRG1 was blocked in ischemic neurons after pretreatment with AG1478, an inhibitor of ErbB4, but not after pretreatment with AG879, an inhibitor of ErbB2. These results indicate an important role of ErbB4 in NRG1-mediated neuroprotection, suggesting that endogenous ErbB4 might serve as a valuable therapeutic target for treating global cerebral ischemia.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Han-Byeol Kim
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Seung-Yeon Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Hong-Il Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Dae-Yong Song
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| |
Collapse
|