1
|
Pantovic-Stefanovic M, Velimirovic M, Jurisic V, Puric M, Gostiljac M, Dodic S, Minic I, Nesic M, Nikolic T, Petronijevic N, Ivkovic M, Dunjic-Kostic B. Exploring the role of TNF-α, TGF-β, and IL-6 serum levels in categorical and noncategorical models of mood and psychosis. Sci Rep 2024; 14:23117. [PMID: 39367011 PMCID: PMC11452617 DOI: 10.1038/s41598-024-73937-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
Psychotic and mood disorders are discussed as part of the same continuum. The potential role of immune dysregulation in defining their clinical presentations, however, remains unclear. Differences in TNF-α, IL-6 and TGF-β levels were investigated in 143 patients with schizophrenia (SCH = 63) and bipolar disorder (BD = 80), in remission. Cytokines were evaluated against the dimensional assessment of psychosis and affective symptoms using the schizo-bipolar scale, together with the severity of the same symptom domains measured by the brief psychiatric rating scale (BPRS). Lower TGF-β was associated with more lifetime episodes, family risk for psychosis, and more severe mood and psychotic symptoms in all patients. BPRS Affect symptoms domain correlated with lower TGF-β levels in BD, and higher TGF-β levels in SCH patients. Using moderated mediation analysis, TGF-β was a relevant predictor only in the setting of non-categorical symptom distribution, with familial risk for psychosis confirmed as a significant moderator. Severity of BPRS Affect symptoms domain was an independent predictor of inclination towards the psychosis spectrum. The underlying immune dysregulation may be shared by the disorders, rather than a unique characteristic of each, having significant implications for our understanding of the continuum vs. categorical approach to psychosis and mood disorders.
Collapse
Affiliation(s)
- Maja Pantovic-Stefanovic
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia.
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia.
| | - Milica Velimirovic
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia.
- Institute of Medical and Clinical Biochemistry, Pasterova 2, 11000, Belgrade, Serbia.
| | - Vladimir Jurisic
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 11000, Kragujevac, Serbia
| | - Marija Puric
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
| | - Marta Gostiljac
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
| | - Sara Dodic
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
| | - Ivana Minic
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
| | - Milica Nesic
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
| | - Tatjana Nikolic
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Pasterova 2, 11000, Belgrade, Serbia
| | - Natasa Petronijevic
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Pasterova 2, 11000, Belgrade, Serbia
| | - Maja Ivkovic
- Department for Bipolar Disorders, Clinic for Psychiatry, University Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
| | - Bojana Dunjic-Kostic
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
- Institute of Mental Health, Milana Kasanina 3, 11000, Belgrade, Serbia
| |
Collapse
|
2
|
Okur Z, Schlauri N, Bitsikas V, Panopoulou M, Ortiz R, Schwaiger M, Karmakar K, Schreiner D, Scheiffele P. Control of neuronal excitation-inhibition balance by BMP-SMAD1 signalling. Nature 2024; 629:402-409. [PMID: 38632412 PMCID: PMC11078759 DOI: 10.1038/s41586-024-07317-z] [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: 02/23/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024]
Abstract
Throughout life, neuronal networks in the mammalian neocortex maintain a balance of excitation and inhibition, which is essential for neuronal computation1,2. Deviations from a balanced state have been linked to neurodevelopmental disorders, and severe disruptions result in epilepsy3-5. To maintain balance, neuronal microcircuits composed of excitatory and inhibitory neurons sense alterations in neural activity and adjust neuronal connectivity and function. Here we identify a signalling pathway in the adult mouse neocortex that is activated in response to increased neuronal network activity. Overactivation of excitatory neurons is signalled to the network through an increase in the levels of BMP2, a growth factor that is well known for its role as a morphogen in embryonic development. BMP2 acts on parvalbumin-expressing (PV) interneurons through the transcription factor SMAD1, which controls an array of glutamatergic synapse proteins and components of perineuronal nets. PV-interneuron-specific disruption of BMP2-SMAD1 signalling is accompanied by a loss of glutamatergic innervation in PV cells, underdeveloped perineuronal nets and decreased excitability. Ultimately, this impairment of the functional recruitment of PV interneurons disrupts the cortical excitation-inhibition balance, with mice exhibiting spontaneous epileptic seizures. Our findings suggest that developmental morphogen signalling is repurposed to stabilize cortical networks in the adult mammalian brain.
Collapse
Affiliation(s)
- Zeynep Okur
- Biozentrum, University of Basel, Basel, Switzerland
| | - Nadia Schlauri
- Biozentrum, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Raul Ortiz
- Biozentrum, University of Basel, Basel, Switzerland
| | - Michaela Schwaiger
- Swiss Institute of Bioinformatics, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Kajari Karmakar
- Biozentrum, University of Basel, Basel, Switzerland
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | | |
Collapse
|
3
|
Bhuiyan P, Sun Z, Khan MA, Hossain MA, Rahman MH, Qian Y. System biology approaches to identify hub genes linked with ECM organization and inflammatory signaling pathways in schizophrenia pathogenesis. Heliyon 2024; 10:e25191. [PMID: 38322840 PMCID: PMC10844262 DOI: 10.1016/j.heliyon.2024.e25191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 12/18/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Schizophrenia (SZ) is a chronic and devastating mental illness that affects around 20 million individuals worldwide. Cognitive deficits and structural and functional changes of the brain, abnormalities of brain ECM components, chronic neuroinflammation, and devastating clinical manifestation during SZ are likely etiological factors shown by affected individuals. However, the pathophysiological events associated with multiple regulatory pathways involved in the brain of this complex disorder are still unclear. This study aimed to develop a pipeline based on bioinformatics and systems biology approaches for identifying potential therapeutic targets involving possible biological mechanisms from SZ patients and healthy volunteers. About 420 overlapping differentially expressed genes (DEGs) from three RNA-seq datasets were identified. Gene ontology (GO), and pathways analysis showed several biological mechanisms enriched by the commonly shared DEGs, including extracellular matrix organization (ECM) organization, collagen fibril organization, integrin signaling pathway, inflammation mediated by chemokines and cytokines signaling pathway, and GABA-B receptor II and IL4 mediated signaling. Besides, 15 hub genes (FN1, COL1A1, COL3A1, COL1A2, COL5A1, COL2A1, COL6A2, COL6A3, MMP2, THBS1, DCN, LUM, HLA-A, HLA-C, and FBN1) were discovered by comprehensive analysis, which was mainly involved in the ECM organization and inflammatory signaling pathway. Furthermore, the miRNA target of the hub genes was analyzed with the random-forest-based approach software miRTarBase. In addition, the transcriptional factors and protein kinases regulating overlapping DEGs in SZ, namely, SUZ12, EZH2, TRIM28, TP53, EGR1, CSNK2A1, GSK3B, CDK1, and MAPK14, were also identified. The results point to a new understanding that the hub genes (fibronectin 1, collagen, matrix metalloproteinase-2, and lumican) in the ECM organization and inflammatory signaling pathways may be involved in the SZ occurrence and pathogenesis.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
| | - Zhaochu Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Md Arif Khan
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
- Bio-Bio-1 Bioinformatics Research Foundation, Dhaka, Bangladesh
| | - Md Arju Hossain
- Department of Microbiology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Islamic University, Kushtia-7003, Bangladesh
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Nagasubramanian K, Gupta K. Interactome analysis implicates class II transactivator (CIITA) in depression and other neuroinflammatory disorders. Int J Neurosci 2023:1-19. [PMID: 37933915 DOI: 10.1080/00207454.2023.2279502] [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: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Inappropriate inflammatory responses within the nervous system (neuroinflammation) have been implicated in several neurological conditions. Class II transactivator (CIITA), a principal regulator of the major histocompatibility complex II (MHCII), is known to play essential roles in inflammation. Hence, CIITA and its interactors could be potentially involved in multiple neurological disorders. However, the molecular mechanisms underlying CIITA-mediated neuroinflammation (NI) are yet to be understood. MATERIALS AND METHODS In this regard, we analyzed the potential involvement of CIITA and its interactome in the regulation of neuroinflammation. In the present study, using various computational tools, we aimed (1) to identify NI-related proteins, (2) to filter the critical interactors in the CIITA-NI network, and (3) to analyze the protein-disease interactions and the associated molecular pathways through which CIITA could influence neuroinflammation. RESULTS CIITA was found to interact with P T GS2, GSK3B, and NR3C1 and may influence depressive disorders. Further, the IL4/IL13 pathway was found to be potentially underlying the CIITA-interactomemediated effects on neurological disorders. Moreover, CIITA was found to be connected to genes associated with depressive disorder through IL4, wherein CIITA was found to be potentially involved in depressive disorders through IL-4/IL-13 and hippo pathways. However, the present study is based on the existing data on protein interactomes and could be re-evaluated as newer interactions are discovered. Also, the functional mechanisms of CIITA's roles in neuroinflammation must be evaluated further. CONCLUSION Notwithstanding these limitations, the results presented here, could form a basis for further experimental studies to assess CIITA as a potential therapeutic target in managing depression and other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Kishore Nagasubramanian
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Krishnakant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
- NCCS, Pune, India
| |
Collapse
|
5
|
Fitzgerald E, Arcego DM, Shen MJ, O'Toole N, Wen X, Nagy C, Mostafavi S, Craig K, Silveira PP, Rayan NA, Diorio J, Meaney MJ, Zhang TY. Sex and cell-specific gene expression in corticolimbic brain regions associated with psychiatric disorders revealed by bulk and single-nuclei RNA sequencing. EBioMedicine 2023; 95:104749. [PMID: 37549631 PMCID: PMC10432187 DOI: 10.1016/j.ebiom.2023.104749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/28/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND There are sex-specific differences in the prevalence, symptomology and course of psychiatric disorders. However, preclinical models have primarily used males, such that the molecular mechanisms underlying sex-specific differences in psychiatric disorders are not well established. METHODS In this study, we compared transcriptome-wide gene expression profiles in male and female rats within the corticolimbic system, including the cingulate cortex, nucleus accumbens medial shell (NAcS), ventral dentate gyrus and the basolateral amygdala (n = 22-24 per group/region). FINDINGS We found over 3000 differentially expressed genes (DEGs) in the NAcS between males and females. Of these DEGs in the NAcS, 303 showed sex-dependent conservation DEGs in humans and were significantly enriched for gene ontology terms related to blood vessel morphogenesis and regulation of cell migration. Single nuclei RNA sequencing in the NAcS of male and female rats identified widespread sex-dependent expression, with genes upregulated in females showing a notable enrichment for synaptic function. Female upregulated genes in astrocytes, Drd3+MSNs and oligodendrocyte were also enriched in several psychiatric genome-wide association studies (GWAS). INTERPRETATION Our data provide comprehensive evidence of sex- and cell-specific molecular profiles in the NAcS. Importantly these differences associate with anxiety, bipolar disorder, schizophrenia, and cross-disorder, suggesting an intrinsic molecular basis for sex-based differences in psychiatric disorders that strongly implicates the NAcS. FUNDING This work was supported by funding from the Hope for Depression Research Foundation (MJM).
Collapse
Affiliation(s)
- Eamon Fitzgerald
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Mo Jun Shen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Xianglan Wen
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Corina Nagy
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Sara Mostafavi
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 185 E Stevens Way NE, Seattle, WA 9819, USA
| | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Patricia Pelufo Silveira
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nirmala Arul Rayan
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences and Brain - Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Michael J Meaney
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences and Brain - Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tie-Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada.
| |
Collapse
|
6
|
Jaric I, Voelkl B, Clerc M, Schmid MW, Novak J, Rosso M, Rufener R, von Kortzfleisch VT, Richter SH, Buettner M, Bleich A, Amrein I, Wolfer DP, Touma C, Sunagawa S, Würbel H. The rearing environment persistently modulates mouse phenotypes from the molecular to the behavioural level. PLoS Biol 2022; 20:e3001837. [PMID: 36269766 PMCID: PMC9629646 DOI: 10.1371/journal.pbio.3001837] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/02/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
The phenotype of an organism results from its genotype and the influence of the environment throughout development. Even when using animals of the same genotype, independent studies may test animals of different phenotypes, resulting in poor replicability due to genotype-by-environment interactions. Thus, genetically defined strains of mice may respond differently to experimental treatments depending on their rearing environment. However, the extent of such phenotypic plasticity and its implications for the replicability of research findings have remained unknown. Here, we examined the extent to which common environmental differences between animal facilities modulate the phenotype of genetically homogeneous (inbred) mice. We conducted a comprehensive multicentre study, whereby inbred C57BL/6J mice from a single breeding cohort were allocated to and reared in 5 different animal facilities throughout early life and adolescence, before being transported to a single test laboratory. We found persistent effects of the rearing facility on the composition and heterogeneity of the gut microbial community. These effects were paralleled by persistent differences in body weight and in the behavioural phenotype of the mice. Furthermore, we show that environmental variation among animal facilities is strong enough to influence epigenetic patterns in neurons at the level of chromatin organisation. We detected changes in chromatin organisation in the regulatory regions of genes involved in nucleosome assembly, neuronal differentiation, synaptic plasticity, and regulation of behaviour. Our findings demonstrate that common environmental differences between animal facilities may produce facility-specific phenotypes, from the molecular to the behavioural level. Furthermore, they highlight an important limitation of inferences from single-laboratory studies and thus argue that study designs should take environmental background into account to increase the robustness and replicability of findings. The phenotype of an organism results not only from its genotype but also the influence of its environment throughout development. This study shows that common environmental differences between animal facilities can induce substantial variation in the phenotype of mice, thereby highlighting an important limitation of inferences from single-laboratory studies in animal research.
Collapse
Affiliation(s)
- Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail: (IJ); (HW)
| | - Bernhard Voelkl
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Melanie Clerc
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | | | - Janja Novak
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marianna Rosso
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | | | - S. Helene Richter
- Department of Behavioural Biology, University of Münster, Münster, Germany
| | - Manuela Buettner
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Irmgard Amrein
- Institute of Anatomy, Division of Functional Neuroanatomy, University of Zürich, Zürich, Switzerland; Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - David P. Wolfer
- Institute of Anatomy, Division of Functional Neuroanatomy, University of Zürich, Zürich, Switzerland; Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Chadi Touma
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany
| | - Shinichi Sunagawa
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail: (IJ); (HW)
| |
Collapse
|
7
|
Gibbons AS, Hoyer D, Dean B. SMAD4 protein is decreased in the dorsolateral prefrontal and anterior cingulate cortices in schizophrenia. World J Biol Psychiatry 2021; 22:70-77. [PMID: 32081064 DOI: 10.1080/15622975.2020.1733081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Signal transduction through the mothers against decapentaplegic (SMAD) is a family of signal transduction factors that mediate signalling of the transforming growth factor B (TGFB)-superfamily of cell regulatory proteins. A recent transcriptomic analysis of post-mortem, cortical tissue from subjects with schizophrenia found decreased mRNA expression of SMAD2 and SMAD4 in the dorsolateral prefrontal cortex (DLPFC) associated with the disorder. To expand this initial finding, we sought to determine whether SMAD2 and SMAD4 protein were also altered in the cortex from subjects with schizophrenia. METHODS Western blotting was used to measure SMAD2 and SMAD4 protein levels in DLPFC and anterior cingulate cortex (ACC) taken post-mortem from subjects with schizophrenia (n = 20) and matched control (n = 20) subjects. RESULTS Compared to controls, levels of SMAD4 were 25% lower in the DLPFC and 38% lower in the ACC from subjects with schizophrenia. By contrast, SMAD2 levels were not altered in either DLPFC or ACC. CONCLUSIONS Our finding of lower SMAD4 protein in the cortex suggests there are likely to be abnormalities in cortical TGFB-superfamily signalling in schizophrenia.
Collapse
Affiliation(s)
- Andrew S Gibbons
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,The Department of Psychiatry, Monash University, Clayton, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Brian Dean
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,Brain and Psychological Sciences Research Centre, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
8
|
Izsak J, Vizlin-Hodzic D, Iljin M, Strandberg J, Jadasz J, Olsson Bontell T, Theiss S, Hanse E, Ågren H, Funa K, Illes S. TGF-β1 Suppresses Proliferation and Induces Differentiation in Human iPSC Neural in vitro Models. Front Cell Dev Biol 2020; 8:571332. [PMID: 33195202 PMCID: PMC7655796 DOI: 10.3389/fcell.2020.571332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Persistent neural stem cell (NSC) proliferation is, among others, a hallmark of immaturity in human induced pluripotent stem cell (hiPSC)-based neural models. TGF-β1 is known to regulate NSCs in vivo during embryonic development in rodents. Here we examined the role of TGF-β1 as a potential candidate to promote in vitro differentiation of hiPSCs-derived NSCs and maturation of neuronal progenies. We present that TGF-β1 is specifically present in early phases of human fetal brain development. We applied confocal imaging and electrophysiological assessment in hiPSC-NSC and 3D neural in vitro models and demonstrate that TGF-β1 is a signaling protein, which specifically suppresses proliferation, enhances neuronal and glial differentiation, without effecting neuronal maturation. Moreover, we demonstrate that TGF-β1 is equally efficient in enhancing neuronal differentiation of human NSCs as an artificial synthetic small molecule. The presented approach provides a proof-of-concept to replace artificial small molecules with more physiological signaling factors, which paves the way to improve the physiological relevance of human neural developmental in vitro models.
Collapse
Affiliation(s)
- Julia Izsak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Dzeneta Vizlin-Hodzic
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Margarita Iljin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Joakim Strandberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Janusz Jadasz
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Thomas Olsson Bontell
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stephan Theiss
- Result Medical GmbH, Düsseldorf, Germany.,Medical Faculty, Institute of Clinical Neuroscience and Medical Psychology, Heinrich Heine University, Düsseldorf, Germany
| | - Eric Hanse
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Hans Ågren
- Section of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Keiko Funa
- Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Illes
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Rozés-Salvador V, Wilson C, Olmos C, Gonzalez-Billault C, Conde C. Fine-Tuning the TGFβ Signaling Pathway by SARA During Neuronal Development. Front Cell Dev Biol 2020; 8:550267. [PMID: 33015054 PMCID: PMC7494740 DOI: 10.3389/fcell.2020.550267] [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: 04/08/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Neural development is a complex process that involves critical events, including cytoskeleton dynamics and selective trafficking of proteins to defined cellular destinations. In this regard, Smad Anchor for Receptor Activation (SARA) is an early endosome resident protein, where perform trafficking- associated functions. In addition, SARA is also involved in cell signaling, including the TGFβ-dependent pathway. Accordingly, SARA, and TGFβ signaling are required for proper axonal specification and migration of cortical neurons, unveiling a critical role for neuronal development. However, the cooperative action between the TGFβ pathway and SARA to this process has remained understudied. In this work, we show novel evidence suggesting a cross-talk between SARA and TGFβ pathway needed for proper polarization, axonal specification, growth and cortical migration of central neurons both in vitro and in vivo. Using microscopy tools and cultured hippocampal neurons, we show a local interaction between SARA and TβRI (TGFβ I receptor) at endosomes. In addition, SARA loss of function, induced by the expression of the dominant-negative SARA-F728A, over-activates the TGFβ pathway, most likely by preserving phosphorylated TβRI. Consequently, SARA-mediated activation of TGFβ pathway impacts on neuronal development, promoting axonal growth and cortical migration of neurons during brain development. Moreover, our data suggests that SARA basally prevents the activation of TβRI through the recruitment of the inhibitory complex PP1c/GADD34 in polarizing neurons. Together, these results propose that SARA is a negative regulator of the TGFβ pathway, being critical for a proper orchestration for neuronal development.
Collapse
Affiliation(s)
- Victoria Rozés-Salvador
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina.,Instituto de Ciencias Básicas, Universidad Nacional de Villa María (UNVM), Córdoba, Argentina
| | - Carlos Wilson
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina.,Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina
| | - Cristina Olmos
- Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Christian Gonzalez-Billault
- Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States
| | - Cecilia Conde
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina
| |
Collapse
|
10
|
Corsi-Zuelli F, Loureiro CM, Shuhama R, Fachim HA, Menezes PR, Louzada-Junior P, Mondelli V, Del-Ben CM. Cytokine profile in first-episode psychosis, unaffected siblings and community-based controls: the effects of familial liability and childhood maltreatment. Psychol Med 2020; 50:1139-1147. [PMID: 31064423 DOI: 10.1017/s0033291719001016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Inflammation is a possible biological mechanism underlying the association between childhood maltreatment and psychosis. Previous investigations on this regard were mainly conducted on chronic schizophrenia and lacked control for confounders. We aim to investigate the role of familial liability, childhood maltreatment and recent stress in determining cytokine abnormalities at the onset of psychosis. METHODS We recruited 114 first-episode psychosis (FEP) patients, 57 unaffected biological siblings of FEP patients, and 251 community-based controls. Plasma cytokines (IL-1β, IL-6, TNF-α, IFN-γ, IL-4, IL-10 and TGF-β) were measured and differences across the groups analysed after adjusting for potential confounders. RESULTS FEP had a higher pro- and anti-inflammatory cytokine profile (IL-1β, IL-6, TNF-α, IL-10 and TGF-β), which was not observed in unaffected siblings. Siblings presented decreased IL-1β when compared with patients and controls. Childhood maltreatment was associated with higher levels of TGF-β in both patients and siblings when compared with controls. Physical childhood abuse was associated with increased levels of TGF-β in FEP patients but with decreased levels in controls. Other childhood maltreatment subtypes or recent stressors did not affect cytokine levels in any of the groups. CONCLUSIONS Normal or reduced cytokines in siblings represent possibly a protective factor and suggest that the identified inflammatory profile in FEP can be a real pathophysiological component of psychosis. Experience of childhood maltreatment may contribute as long-term immune priming for the TGF-β pathway, and increased levels of this cytokine in both patients and siblings exposed to childhood maltreatment point to a possible biological candidate of familial risk for psychosis.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Camila Marcelino Loureiro
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Rosana Shuhama
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Helene Aparecida Fachim
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Paulo Rossi Menezes
- Department of Preventive Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paulo Louzada-Junior
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Cristina Marta Del-Ben
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Nenov MN, Malkov AE, Konakov MV, Levin SG. Interleukin-10 and transforming growth factor-β1 facilitate long-term potentiation in CA1 region of hippocampus. Biochem Biophys Res Commun 2019; 518:486-491. [PMID: 31434608 DOI: 10.1016/j.bbrc.2019.08.072] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022]
Abstract
It has been shown that pro-inflammatory cytokines preferentially attenuate long-term potentiation (LTP), at the same time the effect of anti-inflammatory cytokines on synaptic plasticity has not been fully studied yet. Here we studied the effect of two anti-inflammatory cytokines - interleukin-10 (IL-10) and transforming growth factor-β1 (TGF-β1) on long-term potentiation. It was found that exogenously added IL-10 as well as TGF-β1 were able to effectively facilitate LTP evoked with ether high frequency or theta burst stimulation protocols in CA1 area of hippocampus. Effectiveness of IL-10 and TGF-β1 on LTP varied depending on the concentration of used cytokine and type of tetanic stimulation protocol used for LTP induction. Overall the positive effect of studied cytokines on LTP was associated with their ability to increase basal synaptic strength at Schaffer collateral - CA1 synapse. At the same time IL-10 and TGF-β1 did not have any effect on short-term plasticity. Our results provide new evidence upon the modulatory effects that anti-inflammatory cytokines exert on synaptic plasticity further highlighting their potency as modulators of neuronal function.
Collapse
Affiliation(s)
- Miroslav N Nenov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| | - Anton E Malkov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Maxim V Konakov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Sergey G Levin
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
12
|
Vieira JS, Cunha EJ, de Souza JF, Sant'Ana RD, Zielak JC, Costa-Casagrande TA, Giovanini AF. Alendronate induces postnatal maxillary bone growth by stimulating intramembranous ossification and preventing premature cartilage mineralization in the midpalatal suture of newborn rats. Int J Oral Maxillofac Surg 2019; 48:1494-1503. [PMID: 31054875 DOI: 10.1016/j.ijom.2019.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/25/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Cleft palate is a common malformation of craniofacial development, and postnatal deficiencies in palate formation may occur. The aim of this study was to determine whether alendronate treatment could induce maxillary mineralization and thus reduce the need for surgical procedures. The effects of alendronate on maxillary bone development, the midpalatal suture, and the levels of transforming growth factor beta-1 (TGF-β1), bone morphogenetic protein 2 (BMP-2), collagen I and II, and V-ATPase were evaluated in newborn rats. Thirty newborn rats were placed in a control group and 30 in a group that received intraperitoneal alendronate (2.5 mg/kg/day). The animals were euthanized on day 7 or 12, and the heads were subjected to histological and immunohistochemical analyses. Specimens from rats that received alendronate presented larger bone matrix deposition in areas of intramembranous ossification of the maxillary bone when compared to controls. Furthermore, higher levels of TGF-β1, BMP-2, and collagen I were observed, whereas osteoclasts showed no V-ATPase. The alendronate group also showed higher levels of TGF-β1 and collagen II in the midpalatal suture, whereas BMP-2 levels were lower than in controls. These results coincided with an expansion of the chondroid. In conclusion, alendronate increased the intramembranous ossification in the maxillary bone in association with increased expression of TGF-β1, BMP-2, and collagen I and decreased V-ATPase. The drug induced an expansion of chondrocytes and a decrease in mineral bone deposition despite the high levels of TGF-β1 in this area. Alendronate may therefore be useful in the treatment of diseases affecting bone growth.
Collapse
Affiliation(s)
- J S Vieira
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - E J Cunha
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - J F de Souza
- Department of Stomatology, School of Dentistry, Federal University of Paraná, UFPR, Paraná, Brazil
| | - R D Sant'Ana
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - J C Zielak
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - T A Costa-Casagrande
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - A F Giovanini
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil.
| |
Collapse
|
13
|
The role of anti-inflammatory cytokines in memory processing in a healthy brain. Behav Brain Res 2019; 367:111-116. [PMID: 30943419 DOI: 10.1016/j.bbr.2019.03.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 11/21/2022]
Abstract
The purpose of the work was to study the role of anti-inflammatory cytokines in memory processing in a healthy brain. Wistar rats were trained to perform a task with positive (food) reinforcement; and then the task performance was tested after intraventricular injection of IL-10 or TGF-β1. A microinjection into the brain of either of the two cytokines did not affect the performance of the task and did not have an anti-amnesic effect when the retrieval was deteriorated with scopolamine. In addition, endogenous levels of IL-10 and TGF-β1 were determine in the prefrontal cortex and in the hippocampus after one and two training sessions, consisting of 10 runs each. The level of IL-10 did not change after training both in the prefrontal cortex and in the hippocampus. Endogenous level of TGF-β1 increased in the neocortex after the first training session, the second session, and recovered to the normal level three days after training. In contrast, in the hippocampus, the level of TGF-β1 was decreased: maximally after the first training session in the right hippocampus and after the second training session in the left one. Given the role of the prefrontal cortex in memory processing, we assume that a specific increase of TGF-β1 in the prefrontal cortex may indicate involvement in memory trace consolidation.
Collapse
|
14
|
Amoli MM, Khatami F, Arzaghi SM, Enayati S, Nejatisafa AA. Over-expression of TGF-β1 gene in medication free Schizophrenia. Psychoneuroendocrinology 2019; 99:265-270. [PMID: 30389222 DOI: 10.1016/j.psyneuen.2018.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 10/13/2018] [Accepted: 10/14/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Immunological pathways play a crucial role in developing and precipitating neuropsychiatric disorders. Although the exact pathogenesis of schizophrenia is unknown, the possible role of genetic and biomarker involvement of the immune system is gaining attention. Here we quantified the mRNA expression of cytokines as a key role player of the immune system from the peripheral blood mononuclear cells of patients with schizophrenia and healthy controls to identify the differentially expressed genes. METHODS Sixteen medication-free schizophrenia patients and 16 healthy subjects were enrolled in the current study. To investigate the desired expression level of mRNAs including TGF-β1, IL-1β, IL-23, TNF-α, NF-κB, and BDNF, quantitative real-time PCR was performed using specific oligonucleotide primers and the Applied Bio systems StepOne™ real time PCR system. DNA methylation was also analyzed through methylation-specific polymerase chain reaction (MSP). RESULTS TGF-β1 was significantly up-regulated in peripheral blood mononuclear cells of patients vs. healthy individuals (P value = 0.03). In addition, we found a significant correlation between the positive symptom scale and TGF-β1 gene overexpression (r = 0.536, P = 0.039). However, we did not observe any statistically significant differences for the methylation status of CpG Islands 1 and 2 between the patients and normal group. No statistical significance was found either for gene expression of IL-1β (P = 0.32), IL-23 (P = 0.12), TNF-α (P = 0.87), NF-κB (P = 0.07), and BDNF (P = 0.33). CONCLUSIONS Although the number of medication-free schizophrenia patients is extremely limited, our data highlighted the potential role of TGF-β1 as a regulatory cytokine in complex inflammatory mechanism involved in medication-free schizophrenia. In addition, we observed that increased level of TGF-β1 mRNA in this disease might not be under methylation as an epigenetic control element at the genomic level.
Collapse
Affiliation(s)
- Mahsa M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Masoud Arzaghi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Samaneh Enayati
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Nejatisafa
- Psychiatry & Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Astrocytes and the TGF-β1 Pathway in the Healthy and Diseased Brain: a Double-Edged Sword. Mol Neurobiol 2018; 56:4653-4679. [DOI: 10.1007/s12035-018-1396-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/14/2018] [Indexed: 12/14/2022]
|
16
|
Joseph AT, Bhardwaj SK, Srivastava LK. Role of Prefrontal Cortex Anti- and Pro-inflammatory Cytokines in the Development of Abnormal Behaviors Induced by Disconnection of the Ventral Hippocampus in Neonate Rats. Front Behav Neurosci 2018; 12:244. [PMID: 30459571 PMCID: PMC6232928 DOI: 10.3389/fnbeh.2018.00244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Neonatal disconnection of ventral hippocampus (VH) outputs in rats has been reported to lead to post-pubertal behavioral and synaptic changes of relevance to schizophrenia. Increased oxidative and inflammatory load in the prefrontal cortex (PFC) has been suggested to mediate some of the effects of neonatal VH lesion (NVHL). In this study, we hypothesized that developmental imbalance of anti- and pro-inflammatory factors within the PFC might affect synaptic development thus contributing to the adult NVHL-induced behavioral deficits. Ibotenic acid-induced excitotoxic NVHL was performed in postnatal day (PD) 7 male Sprague-Dawley rats and the mRNA levels of select pro- and anti-inflammatory cytokines were measured in the medial PFC (mPFC) at two developmental time points (PD15 and PD60). We observed a development-specific increase of pro-inflammatory cytokine, interleukin (IL)-1β mRNA at PD15, and an overall reduction in the expression and signaling of transforming growth factor beta 1 (TGF-β1), an anti-inflammatory cytokine, at both PD15 and PD60 in the NVHL animals. These cytokine changes were not seen in the somatosensory cortex (SSC) or tissue surrounding the lesion site. Daily administration of systemic recombinant TGF-β1 from PD7-14 prevented the appearance of hyperlocomotion, deficits in prepulse inhibition (PPI) of startle and social interaction (SI) in post-pubertal (PD60) NVHL rats. Neonatal supplementation of TGF-β1 was also able to attenuate dendritic spine loss in the layer 3 mPFC pyramidal neurons of NVHL animals. These results suggest that early damage of the VH has long-lasting inflammatory consequences in distant connected structures, and that TGF-β1 has potential to confer protection against the deleterious effects of developmental hippocampal damage.
Collapse
Affiliation(s)
- Antoneta T Joseph
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Sanjeev K Bhardwaj
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Lalit K Srivastava
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
17
|
Canonical TGF-β Signaling Negatively Regulates Neuronal Morphogenesis through TGIF/Smad Complex-Mediated CRMP2 Suppression. J Neurosci 2018; 38:4791-4810. [PMID: 29695415 DOI: 10.1523/jneurosci.2423-17.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 11/21/2022] Open
Abstract
Functional neuronal connectivity requires proper neuronal morphogenesis and its dysregulation causes neurodevelopmental diseases. Transforming growth factor-β (TGF-β) family cytokines play pivotal roles in development, but little is known about their contribution to morphological development of neurons. Here we show that the Smad-dependent canonical signaling of TGF-β family cytokines negatively regulates neuronal morphogenesis during brain development. Mechanistically, activated Smads form a complex with transcriptional repressor TG-interacting factor (TGIF), and downregulate the expression of a neuronal polarity regulator, collapsin response mediator protein 2. We also demonstrate that TGF-β family signaling inhibits neurite elongation of human induced pluripotent stem cell-derived neurons. Furthermore, the expression of TGF-β receptor 1, Smad4, or TGIF, which have mutations found in patients with neurodevelopmental disorders, disrupted neuronal morphogenesis in both mouse (male and female) and human (female) neurons. Together, these findings suggest that the regulation of neuronal morphogenesis by an evolutionarily conserved function of TGF-β signaling is involved in the pathogenesis of neurodevelopmental diseases.SIGNIFICANCE STATEMENT Canonical transforming growth factor-β (TGF-β) signaling plays a crucial role in multiple organ development, including brain, and mutations in components of the signaling pathway associated with several human developmental disorders. In this study, we found that Smads/TG-interacting factor-dependent canonical TGF-β signaling regulates neuronal morphogenesis through the suppression of collapsin response mediator protein-2 (CRMP2) expression during brain development, and that function of this signaling is evolutionarily conserved in the mammalian brain. Mutations in canonical TGF-β signaling factors identified in patients with neurodevelopmental disorders disrupt the morphological development of neurons. Thus, our results suggest that proper control of TGF-β/Smads/CRMP2 signaling pathways is critical for the precise execution of neuronal morphogenesis, whose impairment eventually results in neurodevelopmental disorders.
Collapse
|
18
|
De Sanctis C, Bellenchi GC, Viggiano D. A meta-analytic approach to genes that are associated with impaired and elevated spatial memory performance. Psychiatry Res 2018; 261:508-516. [PMID: 29395873 DOI: 10.1016/j.psychres.2018.01.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Abstract
Spatial memory deficits are a common hallmark of psychiatric conditions, possibly due to a genetic predisposition. Thus, unravelling the relationship between genes and memory might suggest novel therapeutic targets and pathogenetic pathways. Genetic deletions are known to lead to memory deficits (post-deletion "forgetfulness" genes, PDF), or, in few instances to improve spatial memory (post-deletion "hypermnesic" genes, PDH). To assess this topic, we performed a meta-analytic approach on memory behavior in knock-out mice. We screened 300 studies from PubMed and retrieved 87 genes tested for possible effects on spatial memory. This database was crossed with the Allen Brain Atlas (brain distribution) and the Enrichr (gene function) databases. The results show that PDF genes have higher expression level in several ventral brain structures, particularly the encephalic trunk and in the hypothalamus. Moreover, part of these genes are implicated in synaptic functions. Conversely, the PDH genes are associated to G-protein coupled receptors downstream signalling. Some candidate drugs were also found to interfere with some of the PDH genes, further suggesting that this approach might help in identifying drugs to improve memory performance in psychiatric conditions.
Collapse
Affiliation(s)
- Claudia De Sanctis
- IRCCS Neuromed, Pozzilli, IS 86077, Italy; Department of Medicine and Health Sciences, University of Molise, Via De Sanctis, Campobasso 86100, Italy
| | | | - Davide Viggiano
- Department of Medicine and Health Sciences, University of Molise, Via De Sanctis, Campobasso 86100, Italy.
| |
Collapse
|
19
|
Liu Z, Huo X, Zhao S, Yang J, Shi W, Jing L, Li W, Li Y, Ma L, Gao Y, Diao A. Low density lipoprotein receptor class A domain containing 4 (LDLRAD4) promotes tumorigenesis of hepatic cancer cells. Exp Cell Res 2017; 360:189-198. [PMID: 28888937 DOI: 10.1016/j.yexcr.2017.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/03/2017] [Accepted: 09/02/2017] [Indexed: 02/08/2023]
Abstract
LDLRAD4 was previously identified and shown to be connected with psychiatric disorders. The structure of LDLRAD4 protein is similar to that of TMEPAI protein, which is overexpressed in many tumors. However, it is still unknown whether LDLRAD4 is involved in tumorigenesis. In this study, the potential role of LDLRAD4 in tumorigenesis was investigated. LDLRAD4 is elevated in hepatic cancer cells and tumor tissues, and expression of LDLRAD4 promotes hepatic cancer cell HepG2 and SMMC-7721 proliferation and migration. LDLRAD4 interacts Nedd4 to promote cell proliferation and migration and negatively regulates the TGF-β signaling. Furthermore, immunofluorescence microscopy analysis indicates that LDLRAD4 is localized to the lysosome and association with Nedd4 is necessary for its intracellular transport to the lysosome. In addition, depletion of LDLRAD4 in HepG2 liver cancer cells inhibited tumorigenesis in nude mice. These results reveal an oncogenic role of LDLRAD4 in tumorigenesis through its association with Nedd4.
Collapse
Affiliation(s)
- Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Xin Huo
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Shuang Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Jingjing Yang
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Wenxia Shi
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, No. 83, Jintang Road, Hedong District, Tianjin 300170, China
| | - Lei Jing
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Wei Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Long Ma
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Yingtang Gao
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, No. 83, Jintang Road, Hedong District, Tianjin 300170, China.
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China.
| |
Collapse
|
20
|
Astrocyte Transforming Growth Factor Beta 1 Protects Synapses against Aβ Oligomers in Alzheimer's Disease Model. J Neurosci 2017; 37:6797-6809. [PMID: 28607171 DOI: 10.1523/jneurosci.3351-16.2017] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive decline, increasingly attributed to neuronal dysfunction induced by amyloid-β oligomers (AβOs). Although the impact of AβOs on neurons has been extensively studied, only recently have the possible effects of AβOs on astrocytes begun to be investigated. Given the key roles of astrocytes in synapse formation, plasticity, and function, we sought to investigate the impact of AβOs on astrocytes, and to determine whether this impact is related to the deleterious actions of AβOs on synapses. We found that AβOs interact with astrocytes, cause astrocyte activation and trigger abnormal generation of reactive oxygen species, which is accompanied by impairment of astrocyte neuroprotective potential in vitro We further show that both murine and human astrocyte conditioned media (CM) increase synapse density, reduce AβOs binding, and prevent AβO-induced synapse loss in cultured hippocampal neurons. Both a neutralizing anti-transforming growth factor-β1 (TGF-β1) antibody and siRNA-mediated knockdown of TGF-β1, previously identified as an important synaptogenic factor secreted by astrocytes, abrogated the protective action of astrocyte CM against AβO-induced synapse loss. Notably, TGF-β1 prevented hippocampal dendritic spine loss and memory impairment in mice that received an intracerebroventricular infusion of AβOs. Results suggest that astrocyte-derived TGF-β1 is part of an endogenous mechanism that protects synapses against AβOs. By demonstrating that AβOs decrease astrocyte ability to protect synapses, our results unravel a new mechanism underlying the synaptotoxic action of AβOs in AD.SIGNIFICANCE STATEMENT Alzheimer's disease is characterized by progressive cognitive decline, mainly attributed to synaptotoxicity of the amyloid-β oligomers (AβOs). Here, we investigated the impact of AβOs in astrocytes, a less known subject. We show that astrocytes prevent synapse loss induced by AβOs, via production of transforming growth factor-β1 (TGF-β1). We found that AβOs trigger morphological and functional alterations in astrocytes, and impair their neuroprotective potential. Notably, TGF-β1 reduced hippocampal dendritic spine loss and memory impairment in mice that received intracerebroventricular infusions of AβOs. Our results describe a new mechanism underlying the toxicity of AβOs and indicate novel therapeutic targets for Alzheimer's disease, mainly focused on TGF-β1 and astrocytes.
Collapse
|
21
|
Dixit AB, Banerjee J, Srivastava A, Tripathi M, Sarkar C, Kakkar A, Jain M, Chandra PS. RNA-seq analysis of hippocampal tissues reveals novel candidate genes for drug refractory epilepsy in patients with MTLE-HS. Genomics 2016; 107:178-88. [DOI: 10.1016/j.ygeno.2016.04.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 01/12/2016] [Accepted: 04/05/2016] [Indexed: 12/13/2022]
|
22
|
Muñoz MD, Antolín-Vallespín M, Tapia-González S, Sánchez-Capelo A. Smad3 deficiency inhibits dentate gyrus LTP by enhancing GABAA neurotransmission. J Neurochem 2016; 137:190-9. [PMID: 26826552 DOI: 10.1111/jnc.13558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/07/2016] [Accepted: 01/20/2016] [Indexed: 11/29/2022]
Abstract
Transforming growth factor-β signaling through intracellular Smad3 has been implicated in Parkinson's disease (PD) and it fulfills an important role in the neurogenesis and synaptic plasticity that occurs in the adult dentate gyrus (DG). The long-term potentiation (LTP) induced in the DG by high-frequency stimulation of the medial perforant pathway is abolished in the DG of Smad3-deficient mice, but not in the CA1 hippocampal region. Here, we show that NMDA- and AMPA-type glutamate receptors do not participate in the inhibition of LTP associated with Smad3 deficiency. Moreover, there is no difference in the hippocampal GAD65 and GAD67 content, suggesting that GABA biosynthesis remains unaffected. Increased conductance and higher action potential firing thresholds were evident in intracellular recordings of granule cells from Smad3 deficient mice. Interestingly, phasic and tonic GABAA receptor (GABAA R)-mediated neurotransmission is enhanced in the DG of Smad3-deficient mice, and LTP induction can be rescued by inhibiting GABAA R with picrotoxin. Hence, Smad3 signaling in the DG appears to be necessary to induce LTP by regulating GABAA neurotransmission, suggesting a central role of this intracellular signaling pathway in the hippocampal brain plasticity related to learning and memory. Smad3 deficient mice represent a new and interesting model of Parkinson's disease, displaying hippocampal dysfunctions that include decreased neurogenesis and the failure to induce LTP in the dentate gyrus. Here we show that Smad3 deficiency inhibits LTP induction by enhancing phasic and tonic GABAA receptor-mediated neurotransmission, while LTP induction can be rescued with a GABAA receptor antagonist. Alteration of GABA neurotransmission is thought to produce hippocampal cognitive dysfunction in Down's syndrome or Alzheimer's disease, and here we provide new insights into the hippocampal changes in an animal model of Parkinson's disease.
Collapse
Affiliation(s)
- M Dolores Muñoz
- Unidad de Neurología Experimental, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Mónica Antolín-Vallespín
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Silvia Tapia-González
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Amelia Sánchez-Capelo
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| |
Collapse
|
23
|
Forebrain-Specific Loss of BMPRII in Mice Reduces Anxiety and Increases Object Exploration. PLoS One 2015; 10:e0139860. [PMID: 26444546 PMCID: PMC4596878 DOI: 10.1371/journal.pone.0139860] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/16/2015] [Indexed: 12/14/2022] Open
Abstract
To investigate the role of Bone Morphogenic Protein Receptor Type II (BMPRII) in learning, memory, and exploratory behavior in mice, a tissue-specific knockout of BMPRII in the post-natal hippocampus and forebrain was generated. We found that BMPRII mutant mice had normal spatial learning and memory in the Morris water maze, but showed significantly reduced swimming speeds with increased floating behavior. Further analysis using the Porsolt Swim Test to investigate behavioral despair did not reveal any differences in immobility between mutants and controls. In the Elevated Plus Maze, BMPRII mutants and Smad4 mutants showed reduced anxiety, while in exploratory tests, BMPRII mutants showed more interest in object exploration. These results suggest that loss of BMPRII in the mouse hippocampus and forebrain does not disrupt spatial learning and memory encoding, but instead impacts exploratory and anxiety-related behaviors.
Collapse
|
24
|
Berretta S, Heckers S, Benes FM. Searching human brain for mechanisms of psychiatric disorders. Implications for studies on schizophrenia. Schizophr Res 2015; 167:91-7. [PMID: 25458567 PMCID: PMC4427537 DOI: 10.1016/j.schres.2014.10.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022]
Abstract
In the past 25years, research on the human brain has been providing a clear path toward understanding the pathophysiology of psychiatric illnesses. The successes that have been accrued are matched by significant difficulties identifying and controlling a large number of potential confounding variables. By systematically and effectively accounting for unwanted variance in data from imaging and postmortem human brain studies, meaningful and reliable information regarding the pathophysiology of human brain disorders can be obtained. This perspective paper focuses on postmortem investigations to discuss some of the most challenging sources of variance, including diagnosis, comorbidity, substance abuse and pharmacological treatment, which confound investigations of the human brain.
Collapse
Affiliation(s)
- Sabina Berretta
- Translational Neuroscience Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA.
| | - Stephan Heckers
- Department of Psychiatry, Vanderbilt University. 161 21st Ave S. #T1217 Nashville, TN, USA
| | - Francine M. Benes
- Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA,Program in Neuroscience, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA,Program in Structural and Molecular Neuroscience, 115 Mill St. Belmont MA, 02478, USA
| |
Collapse
|
25
|
Dixit AB, Tripathi M, Chandra PS, Banerjee J. Molecular biomarkers in drug-resistant epilepsy: Facts & possibilities. Int J Surg 2015; 36:483-491. [PMID: 26306771 DOI: 10.1016/j.ijsu.2015.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/03/2015] [Indexed: 01/08/2023]
Abstract
Despite great advances in our understanding of the process of epileptogenesis we are yet to develop reliable biomarkers that have the potential to accurately localize the epileptogenic zone (EZ), and to resolve the issue of heterogeneity in epilepsy surgery outcome. Inability to precisely localize the epileptogenic foci is one of the reason why more than 30% of these DRE patients are not benefited. Molecular and cellular biomarkers in combination with imaging and electrical investigations will provide a more specific platform for defining epileptogenic zone. Potential molecular biomarkers of epileptogenesis including markers of inflammation, synaptic alterations and neurodegeneration may also have the potential for localizing EZ. At molecular level components derived from epileptogenic tissues, such as metabolites, proteins, mRNAs and miRNAs that are significantly altered can serve as biomarkers and can be clubbed with existing techniques to preoperatively localize the EZ. Neurosurgeons across the world face problems while defining the margins of the epileptogenic tissues to be resected during surgery. In this review we discuss molecular biomarkers reported so far in the context of epileptogenesis and some of the unexplored markers which may have the potential to localize EZ during surgery. We also discuss "Intelligent knife" technique that couples electrosurgery and mass spectrometry allowing near-real-time characterization of human tissue and may prove to be instrumental in defining the margins of the epileptogenic zone during surgery.
Collapse
Affiliation(s)
- Aparna Banerjee Dixit
- Center of Excellence for Epilepsy, National Brain Research Centre, Manesar, 122051, India
| | - Manjari Tripathi
- Dept. of Neurology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - P Sarat Chandra
- Dept. of Neurosurgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jyotirmoy Banerjee
- Center of Excellence for Epilepsy, National Brain Research Centre, Manesar, 122051, India.
| |
Collapse
|
26
|
McGehee AM, Moss BJ, Juo P. The DAF-7/TGF-β signaling pathway regulates abundance of the Caenorhabditis elegans glutamate receptor GLR-1. Mol Cell Neurosci 2015; 67:66-74. [PMID: 26054666 DOI: 10.1016/j.mcn.2015.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/03/2015] [Indexed: 11/24/2022] Open
Abstract
Transforming growth factor-β (TGF-β) family signaling pathways have roles in both neuronal development and the regulation of synaptic function. Here we identify a novel role for the Caenorhabditis elegans DAF-7/TGF-β signaling pathway in the regulation of the AMPA-type glutamate receptor GLR-1. We found that the abundance of GLR-1 increases at synapses in the ventral nerve cord (VNC) of animals with loss-of-function mutations in multiple DAF-7/TGF-β pathway components including the TGF-β ligand DAF-7, the type I receptor DAF-1, and the Smads DAF-8 and DAF-14. The GLR-1 defect can be rescued by expression of daf-8 specifically in glr-1-expressing interneurons. The effect on GLR-1 was specific for the DAF-7 pathway because mutations in the DBL-1/TGF-β family pathway did not increase GLR-1 levels in the VNC. Immunoblot analysis indicates that total levels of GLR-1 protein are increased in neurons of DAF-7/TGF-β pathway mutants. The increased abundance of GLR-1 in the VNC of daf-7 pathway mutants is dependent on the transcriptional regulator DAF-3/Smad suggesting that DAF-3-dependent transcription controls GLR-1 levels. Furthermore, we found that glr-1 transcription is increased in daf-7 mutants based on a glr-1 transcriptional reporter. Together these results suggest that the DAF-7/TGF-β signaling pathway functions in neurons and negatively regulates the abundance of GLR-1, in part, by controlling transcription of the receptor itself. Finally, DAF-7/TGF-β pathway mutants exhibit changes in spontaneous locomotion that are dependent on endogenous GLR-1 and consistent with increased glutamatergic signaling. These results reveal a novel mechanism by which TGF-β signaling functions in the nervous system to regulate behavior.
Collapse
Affiliation(s)
- Annette M McGehee
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA; Department of Biology, Suffolk University, Boston, MA 02114, USA.
| | - Benjamin J Moss
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Peter Juo
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
27
|
Yamasaki A, Kasai A, Toi A, Kurita M, Kimoto S, Hayata-Takano A, Nakazawa T, Nagayasu K, Shintani N, Hashimoto R, Ito A, Meltzer HY, Ago Y, Waschek JA, Onaka Y, Matsuda T, Baba A, Hashimoto H. Identification of the role of bone morphogenetic protein (BMP) and transforming growth factor-β (TGF-β) signaling in the trajectory of serotonergic differentiation in a rapid assay in mouse embryonic stem cells in vitro. J Neurochem 2015; 132:418-28. [PMID: 25421849 DOI: 10.1111/jnc.12999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/18/2014] [Accepted: 11/15/2014] [Indexed: 12/25/2022]
Abstract
The mechanism by which extracellular molecules control serotonergic cell fate remains elusive. Recently, we showed that noggin, which inactivates bone morphogenetic proteins (BMPs), induces serotonergic differentiation of mouse embryonic (ES) and induced pluripotent stem cells with coordinated gene expression along the serotonergic lineage. Here, we created a rapid assay for serotonergic induction by generating knock-in ES cells expressing a naturally secreted Gaussia luciferase driven by the enhancer of Pet-1/Fev, a landmark of serotonergic differentiation. Using these cells, we performed candidate-based screening and identified BMP type I receptor kinase inhibitors LDN-193189 and DMH1 as activators of luciferase. LDN-193189 induced ES cells to express the genes encoding Pet-1, tryptophan hydroxylase 2, and the serotonin transporter, and increased serotonin release without altering dopamine release. In contrast, TGF-β receptor inhibitor SB-431542 selectively inhibited serotonergic differentiation, without changing overall neuronal differentiation. LDN-193189 inhibited expression of the BMP signaling target gene Id, and induced the TGF-β target gene Lefty, whereas the opposite effect was observed with SB-431542. This study thus provides a new tool to investigate serotonergic differentiation and suggests that inhibition of BMP type I receptors and concomitant activation of TGF-β receptor signaling are implicated in serotonergic differentiation. Candidate-based screening for serotonergic induction using a rapid assay in mouse embryonic stem cells revealed that the bone morphogenetic protein (BMP) type I receptor kinase inhibitors selectively induce serotonergic differentiation, whereas the TGF-β receptor inhibitor SB-431542 inhibits the differentiation. These results suggest that inhibition of BMP type I receptors and concomitant activation of transforming growth factor-β (TGF-β) receptor signaling are involved in the early trajectory of serotonergic differentiation.
Collapse
Affiliation(s)
- Atsushi Yamasaki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Diniz LP, Matias ICP, Garcia MN, Gomes FCA. Astrocytic control of neural circuit formation: highlights on TGF-beta signaling. Neurochem Int 2014; 78:18-27. [PMID: 25125369 DOI: 10.1016/j.neuint.2014.07.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/18/2014] [Accepted: 07/28/2014] [Indexed: 01/23/2023]
Abstract
Brain function depends critically on the coordinated activity of presynaptic and postsynaptic signals derived from both neurons and non-neuronal elements such as glial cells. A key role for astrocytes in neuronal differentiation and circuitry formation has emerged within the last decade. Although the function of glial cells in synapse formation, elimination and efficacy has greatly increased, we are still very far from deeply understanding the molecular and cellular mechanism underlying these events. The present review discusses the mechanisms driving astrocytic control of excitatory and inhibitory synapse formation in the central nervous system, especially the mechanisms mediated by soluble molecules, particularly those from the TGF-β family. Further, we discuss whether and how human astrocytes might contribute to the acquisition of human cognition. We argue that understanding how astrocytic signals regulate synaptic development might offer new insights into human perception, learning, memory, and cognition and, ultimately, provide new targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isadora C Pereira Matias
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Matheus Nunes Garcia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
29
|
Diniz LP, Tortelli V, Garcia MN, Araújo APB, Melo HM, Silva GSSD, Felice FGD, Alves-Leon SV, Souza JMD, Romão LF, Castro NG, Gomes FCA. Astrocyte transforming growth factor beta 1 promotes inhibitory synapse formation via CaM kinase II signaling. Glia 2014; 62:1917-31. [PMID: 25042347 DOI: 10.1002/glia.22713] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/16/2014] [Accepted: 06/18/2014] [Indexed: 11/09/2022]
Abstract
The balance between excitatory and inhibitory synaptic inputs is critical for the control of brain function. Astrocytes play important role in the development and maintenance of neuronal circuitry. Whereas astrocytes-derived molecules involved in excitatory synapses are recognized, molecules and molecular mechanisms underlying astrocyte-induced inhibitory synapses remain unknown. Here, we identified transforming growth factor beta 1 (TGF-β1), derived from human and murine astrocytes, as regulator of inhibitory synapse in vitro and in vivo. Conditioned media derived from human and murine astrocytes induce inhibitory synapse formation in cerebral cortex neurons, an event inhibited by pharmacologic and genetic manipulation of the TGF-β pathway. TGF-β1-induction of inhibitory synapse depends on glutamatergic activity and activation of CaM kinase II, which thus induces localization and cluster formation of the synaptic adhesion protein, Neuroligin 2, in inhibitory postsynaptic terminals. Additionally, intraventricular injection of TGF-β1 enhanced inhibitory synapse number in the cerebral cortex. Our results identify TGF-β1/CaMKII pathway as a novel molecular mechanism underlying astrocyte control of inhibitory synapse formation. We propose here that the balance between excitatory and inhibitory inputs might be provided by astrocyte signals, at least partly achieved via TGF-β1 downstream pathways. Our work contributes to the understanding of the GABAergic synapse formation and may be of relevance to further the current knowledge on the mechanisms underlying the development of various neurological disorders, which commonly involve impairment of inhibitory synapse transmission.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nakano N, Maeyama K, Sakata N, Itoh F, Akatsu R, Nakata M, Katsu Y, Ikeno S, Togawa Y, Vo Nguyen TT, Watanabe Y, Kato M, Itoh S. C18 ORF1, a novel negative regulator of transforming growth factor-β signaling. J Biol Chem 2014; 289:12680-92. [PMID: 24627487 DOI: 10.1074/jbc.m114.558981] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor (TGF)-β signaling is deliberately regulated at multiple steps in its pathway from the extracellular microenvironment to the nucleus. However, how TGF-β signaling is activated or attenuated is not fully understood. We recently identified transmembrane prostate androgen-induced RNA (TMEPAI), which is involved in a negative feedback loop of TGF-β signaling. When we searched for a family molecule(s) for TMEPAI, we found C18ORF1, which, like TMEPAI, possesses two PY motifs and one Smad-interacting motif (SIM) domain. As expected, C18ORF1 could block TGF-β signaling but not bone morphogenetic protein signaling. C18ORF1 bound to Smad2/3 via its SIM and competed with the Smad anchor for receptor activation for Smad2/3 binding to attenuate recruitment of Smad2/3 to the TGF-β type I receptor (also termed activin receptor-like kinase 5 (ALK5)), in a similar fashion to TMEPAI. Knockdown of C18ORF1 prolonged duration of TGF-β-induced Smad2 phosphorylation and concomitantly potentiated the expression of JunB, p21, and TMEPAI mRNAs induced by TGF-β. Consistently, TGF-β-induced cell migration was enhanced by the knockdown of C18ORF1. These results indicate that the inhibitory function of C18ORF1 on TGF-β signaling is similar to that of TMEPAI. However, in contrast to TMEPAI, C18ORF1 was not induced upon TGF-β signaling. Thus, we defined C18ORF1 as a surveillant of steady state TGF-β signaling, whereas TMEPAI might help C18ORF1 to inhibit TGF-β signaling in a coordinated manner when cells are stimulated with high levels of TGF-β.
Collapse
Affiliation(s)
- Naoko Nakano
- From the Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tapia-González S, Muñoz MD, Cuartero MI, Sánchez-Capelo A. Smad3 is required for the survival of proliferative intermediate progenitor cells in the dentate gyrus of adult mice. Cell Commun Signal 2013; 11:93. [PMID: 24330661 PMCID: PMC4029396 DOI: 10.1186/1478-811x-11-93] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/06/2013] [Indexed: 12/16/2022] Open
Abstract
Background New neurons are continuously being generated in the adult hippocampus, a phenomenon that is regulated by external stimuli, such as learning, memory, exercise, environment or stress. However, the molecular mechanisms underlying neuron production and how they are integrated into existing circuits under such physiological conditions remain unclear. Indeed, the intracellular modulators that transduce the extracellular signals are not yet fully understood. Results We show that Smad3, an intracellular molecule involved in the transforming growth factor (TGF)-β signaling cascade, is strongly expressed by granule cells in the dentate gyrus (DG) of adult mice, although the loss of Smad3 in null mutant mice does not affect their survival. Smad3 is also expressed by adult progenitor cells in the subgranular zone (SGZ) and more specifically, it is first expressed by Type 2 cells (intermediate progenitor cells). Its expression persists through the distinct cell stages towards that of the mature neuron. Interestingly, proliferative intermediate progenitor cells die in Smad3 deficiency, which is associated with a large decrease in the production of newborn neurons in Smad3 deficient mice. Smad3 signaling appears to influence adult neurogenesis fulfilling distinct roles in the rostral and mid-caudal regions of the DG. In rostral areas, Smad3 deficiency increases proliferation and promotes the cell cycle exit of undifferentiated progenitor cells. By contrast, Smad3 deficiency impairs the survival of newborn neurons in the mid-caudal region of the DG at early proliferative stages, activating apoptosis of intermediate progenitor cells. Furthermore, long-term potentiation (LTP) after high frequency stimulation (HFS) to the medial perforant path (MPP) was abolished in the DG of Smad3-deficient mice. Conclusions These data show that endogenous Smad3 signaling is central to neurogenesis and LTP induction in the adult DG, these being two forms of hippocampal brain plasticity related to learning and memory that decline with aging and as a result of neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Amelia Sánchez-Capelo
- CIBERNED - Ser, Neurobiología-Investigación, Hospital Universitario Ramón y Cajal-IRYCIS, Ctra, Colmenar Viejo Km 9, 28034 Madrid, Spain.
| |
Collapse
|
32
|
Poon VY, Choi S, Park M. Growth factors in synaptic function. Front Synaptic Neurosci 2013; 5:6. [PMID: 24065916 PMCID: PMC3776238 DOI: 10.3389/fnsyn.2013.00006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/29/2013] [Indexed: 12/15/2022] Open
Abstract
Synapses are increasingly recognized as key structures that malfunction in disorders like schizophrenia, mental retardation, and neurodegenerative diseases. The importance and complexity of the synapse has fuelled research into the molecular mechanisms underlying synaptogenesis, synaptic transmission, and plasticity. In this regard, neurotrophic factors such as netrin, Wnt, transforming growth factor-β (TGF-β), tumor necrosis factor-α (TNF-α), and others have gained prominence for their ability to regulate synaptic function. Several of these factors were first implicated in neuroprotection, neuronal growth, and axon guidance. However, their roles in synaptic development and function have become increasingly clear, and the downstream signaling pathways employed by these factors have begun to be elucidated. In this review, we will address the role of these factors and their downstream effectors in synaptic function in vivo and in cultured neurons.
Collapse
Affiliation(s)
- Vivian Y Poon
- Neuroscience and Behavioral Disorders Program, Duke-NUS Graduate Medical School Singapore, Singapore
| | | | | |
Collapse
|
33
|
Pisansky MT, Wickham RJ, Su J, Fretham S, Yuan LL, Sun M, Gewirtz JC, Georgieff MK. Iron deficiency with or without anemia impairs prepulse inhibition of the startle reflex. Hippocampus 2013; 23:952-62. [PMID: 23733517 DOI: 10.1002/hipo.22151] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2013] [Indexed: 12/29/2022]
Abstract
Iron deficiency (ID) during early life causes long-lasting detrimental cognitive sequelae, many of which are linked to alterations in hippocampus function, dopamine synthesis, and the modulation of dopaminergic circuitry by the hippocampus. These same features have been implicated in the origins of schizophrenia, a neuropsychiatric disorder with significant cognitive impairments. Deficits in sensorimotor gating represent a reliable endophenotype of schizophrenia that can be measured by prepulse inhibition (PPI) of the acoustic startle reflex. Using two rodent model systems, we investigated the influence of early-life ID on PPI in adulthood. To isolate the role of hippocampal iron in PPI, our mouse model utilized a timed (embryonic day 18.5), hippocampus-specific knockout of Slc11a2, a gene coding an important regulator of cellular iron uptake, the divalent metal transport type 1 protein (DMT-1). Our second model used a classic rat dietary-based global ID during gestation, a condition that closely mimics human gestational ID anemia (IDA). Both models exhibited impaired PPI in adulthood. Furthermore, our DMT-1 knockout model displayed reduced long-term potentiation (LTP) and elevated paired-pulse facilitation (PPF), electrophysiological results consistent with previous findings in the IDA rat model. These results, in combination with previous findings demonstrating impaired hippocampus functioning and altered dopaminergic and glutamatergic neurotransmission, suggest that iron availability within the hippocampus is critical for the neurodevelopmental processes underlying sensorimotor gating. Ultimately, evidence of reduced PPI in both of our models may offer insights into the roles of fetal ID and the hippocampus in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Marc T Pisansky
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Liu Y, Li Z, Zhang M, Deng Y, Yi Z, Shi T. Exploring the pathogenetic association between schizophrenia and type 2 diabetes mellitus diseases based on pathway analysis. BMC Med Genomics 2013; 6 Suppl 1:S17. [PMID: 23369358 PMCID: PMC3552677 DOI: 10.1186/1755-8794-6-s1-s17] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Schizophrenia (SCZ) and type 2 diabetes mellitus (T2D) are both complex diseases. Accumulated studies indicate that schizophrenia patients are prone to present the type 2 diabetes symptoms, but the potential mechanisms behind their association remain unknown. Here we explored the pathogenetic association between SCZ and T2D based on pathway analysis and protein-protein interaction. RESULTS With sets of prioritized susceptibility genes for SCZ and T2D, we identified significant pathways (with adjusted p-value < 0.05) specific for SCZ or T2D and for both diseases based on pathway enrichment analysis. We also constructed a network to explore the crosstalk among those significant pathways. Our results revealed that some pathways are shared by both SCZ and T2D diseases through a number of susceptibility genes. With 382 unique susceptibility proteins for SCZ and T2D, we further built a protein-protein interaction network by extracting their nearest interacting neighbours. Among 2,104 retrieved proteins, 364 of them were found simultaneously interacted with susceptibility proteins of both SCZ and T2D, and proposed as new candidate risk factors for both diseases. Literature mining supported the potential association of partial new candidate proteins with both SCZ and T2D. Moreover, some proteins were hub proteins with high connectivity and interacted with multiple proteins involved in both diseases, implying their pleiotropic effects for the pathogenic association. Some of these hub proteins are the components of our identified enriched pathways, including calcium signaling, g-secretase mediated ErbB4 signaling, adipocytokine signaling, insulin signaling, AKT signaling and type II diabetes mellitus pathways. Through the integration of multiple lines of information, we proposed that those signaling pathways, which contain susceptibility genes for both diseases, could be the key pathways to bridge SCZ and T2D. AKT could be one of the important shared components and may play a pivotal role to link both of the pathogenetic processes. CONCLUSIONS Our study is the first network and pathway-based systematic analysis for SCZ and T2D, and provides the general pathway-based view of pathogenetic association between two diseases. Moreover, we identified a set of candidate genes potentially contributing to the linkage between these two diseases. This research offers new insights into the potential mechanisms underlying the co-occurrence of SCZ and T2D, and thus, could facilitate the inference of novel hypotheses for the co-morbidity of the two diseases. Some etiological factors that exert pleiotropic effects shared by the significant pathways of two diseases may have important implications for the diseases and could be therapeutic intervention targets.
Collapse
Affiliation(s)
- Yanli Liu
- Center for Bioinformatics and Computational Biology, and The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Zezhi Li
- Department of Neurology, Shanghai Changhai Hospital, Secondary Military Medical University, 168 Changhai Road, Shanghai, China
| | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University 37 Guoxuexiang, Chengdu, Sichuan, 610041, China
| | - Youping Deng
- Rush University Cancer Center, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Zhenghui Yi
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wan Ping Nan Road, Shanghai 200030, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology, and The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| |
Collapse
|
35
|
Diniz LP, Almeida JC, Tortelli V, Vargas Lopes C, Setti-Perdigão P, Stipursky J, Kahn SA, Romão LF, de Miranda J, Alves-Leon SV, de Souza JM, Castro NG, Panizzutti R, Gomes FCA. Astrocyte-induced synaptogenesis is mediated by transforming growth factor β signaling through modulation of D-serine levels in cerebral cortex neurons. J Biol Chem 2012; 287:41432-45. [PMID: 23055518 PMCID: PMC3510841 DOI: 10.1074/jbc.m112.380824] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Assembly of synapses requires proper coordination between pre- and postsynaptic elements. Identification of cellular and molecular events in synapse formation and maintenance is a key step to understand human perception, learning, memory, and cognition. A key role for astrocytes in synapse formation and function has been proposed. Here, we show that transforming growth factor β (TGF-β) signaling is a novel synaptogenic pathway for cortical neurons induced by murine and human astrocytes. By combining gain and loss of function approaches, we show that TGF-β1 induces the formation of functional synapses in mice. Further, TGF-β1-induced synaptogenesis involves neuronal activity and secretion of the co-agonist of the NMDA receptor, d-serine. Manipulation of d-serine signaling, by either genetic or pharmacological inhibition, prevented the TGF-β1 synaptogenic effect. Our data show a novel molecular mechanism that might impact synaptic function and emphasize the evolutionary aspect of the synaptogenic property of astrocytes, thus shedding light on new potential therapeutic targets for synaptic deficit diseases.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-590 Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
In primary sensory neocortical areas of mammals, the distribution of sensory receptors is mapped with topographic precision and amplification in proportion to the peripheral receptor density. The visual, somatosensory and auditory cortical maps are established during a critical period in development. Throughout this window in time, the developing cortical maps are vulnerable to deleterious effects of sense organ damage or sensory deprivation. The rodent barrel cortex offers an invaluable model system with which to investigate the mechanisms underlying the formation of topographic maps and their plasticity during development. Five rows of mystacial vibrissa (whisker) follicles on the snout and an array of sinus hairs are represented by layer IV neural modules ('barrels') and thalamocortical axon terminals in the primary somatosensory cortex. Perinatal damage to the whiskers or the sensory nerve innervating them irreversibly alters the structural organization of the barrels. Earlier studies emphasized the role of the sensory periphery in dictating whisker-specific brain maps and patterns. Recent advances in molecular genetics and analyses of genetically altered mice allow new insights into neural pattern formation in the neocortex and the mechanisms underlying critical period plasticity. Here, we review the development and patterning of the barrel cortex and the critical period plasticity.
Collapse
Affiliation(s)
- Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201-1075, USA.
| | | |
Collapse
|
37
|
Li LY, Li JL, Zhang HM, Yang WM, Wang K, Fang Y, Wang Y. TGFβ1 treatment reduces hippocampal damage, spontaneous recurrent seizures, and learning memory deficits in pilocarpine-treated rats. J Mol Neurosci 2012; 50:109-23. [PMID: 22936246 DOI: 10.1007/s12031-012-9879-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 08/15/2012] [Indexed: 11/28/2022]
Abstract
Studies have demonstrated the neuroprotective activity of transforming growth factor beta-1 (TGFβ1), protecting neurons against different kinds of insults. However, the role of exogenous TGFβ1 in the neuronal damage following status epilepticus (SE) and the related spontaneous recurrent seizures (SRS) is unknown. The present study aimed to determine the effect of intranasal TGFβ1 administration on SRS and cognitive function following lithium-pilocarpine-induced SE and associated hippocampal damage. We found that intranasal TGFβ1 significantly attenuated the hippocampal insults marked by hematoxylin and eosin, terminal deoxynucleotidyl transferase dUTP nick end labeling, and Fluoro-Jade B staining by 24, 48, and 72 h after SE was induced. The expression of the apoptosis-suppressing protein, Bcl-2, was elevated, whereas the expression of the apoptosis-promoting proteins, Bax and Caspase-3, was suppressed in TGFβ1-treated rats compared to rats without TGFβ1 treatment by 24, 48, and 72 h following induction of SE. The seizure number, severity, and duration of SRS over a 1-month period of monitoring starting 15 days after SE induction as well as the cognitive deficits detected 45 days after SE induction were significantly reduced in TGFβ1-treated rats compared to those without TGFβ1 treatment. Our results indicate that intranasal delivery of TGFβ1 immediately after SE induction not only protected against SRS but also improved cognitive function. The anti-epileptogenic properties of TGFβ1 may be related to its effect of neuroprotection or to its effect of apoptosis pathway changes.
Collapse
Affiliation(s)
- Liang-Yong Li
- Epilepsy and Headache Group, Department of Neurology, the First Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Chacón PJ, Rodríguez-Tébar A. Increased expression of the homologue of enhancer-of-split 1 protects neurons from beta amyloid neurotoxicity and hints at an alternative role for transforming growth factor beta1 as a neuroprotector. ALZHEIMERS RESEARCH & THERAPY 2012; 4:31. [PMID: 22849569 PMCID: PMC3506945 DOI: 10.1186/alzrt134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/11/2012] [Accepted: 07/31/2012] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the deposition of β-amyloid (Aβ) in the brain, which produces progressive neuronal loss and dementia. We recently demonstrated that the noxious effects of Aβ on cultured hippocampal neurons are in part provoked by the antagonism of nerve growth factor (NGF) signalling, which impairs the activation of nuclear factor κB (NF-κB) by impeding the tyrosine phosphorylation of I-κBα. As a result, the expression of the homologue of Enhancer-of split 1 (Hes1) gene is downregulated and ultimately, gamma-aminobutyric acid (GABA)-ergic connectivity is lost. METHODS Hes1 activity was promoted in cultured hippocampal neurons by overexpressing a Hes1-encoding plasmid or by upregulating this gene by activating NF-κB through different approaches (overexpressing either the I-κB kinaseβ, or p65/RelA/NF-κB). Alternatively neurons were exposed to TGFβ1. Dendrite patterning, GABAergic connectivity and cell survival were analyzed by immunofluorescence microscopy. Hes1 expression was determined by real-time PCR. NF-κB activation was measured using the dual-luciferase reporter assay. RESULTS The expression of Hes1 abolished the effects of Aβ on dendritic patterning and GABAergic input, and it prevented the death of the cultured neurons. TGFβ1, a known neuroprotector, could counteract the deleterious effects of Aβ by inducing NF-κB activation following the serine phosphorylation of I-κBα. Indeed, the number of GABAergic terminals generated by inducing Hes1 expression was doubled. CONCLUSION Our data define some of the mechanisms involved in Aβ-mediated cell death and they point to potential means to counteract this noxious activity.
Collapse
Affiliation(s)
- Pedro J Chacón
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Americo Vespucio s/n, Isla de la Cartuja, 41092 Seville, Spain.
| | | |
Collapse
|
39
|
Martins-de-Souza D, Guest PC, Harris LW, Vanattou-Saifoudine N, Webster MJ, Rahmoune H, Bahn S. Identification of proteomic signatures associated with depression and psychotic depression in post-mortem brains from major depression patients. Transl Psychiatry 2012; 2:e87. [PMID: 22832852 PMCID: PMC3309534 DOI: 10.1038/tp.2012.13] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [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
Major depressive disorder (MDD) is a leading cause of disability worldwide and results tragically in the loss of almost one million lives in Western societies every year. This is due to poor understanding of the disease pathophysiology and lack of empirical medical tests for accurate diagnosis or for guiding antidepressant treatment strategies. Here, we have used shotgun proteomics in the analysis of post-mortem dorsolateral prefrontal cortex brain tissue from 24 MDD patients and 12 matched controls. Brain proteomes were pre-fractionated by gel electrophoresis and further analyzed by shotgun data-independent label-free liquid chromatography-mass spectrometry. This led to identification of distinct proteome fingerprints between MDD and control subjects. Some of these differences were validated by Western blot or selected reaction monitoring mass spectrometry. This included proteins associated with energy metabolism and synaptic function and we also found changes in the histidine triad nucleotide-binding protein 1 (HINT1), which has been implicated recently in regulation of mood and behavior. We also found differential proteome profiles in MDD with (n=11) and without (n=12) psychosis. Interestingly, the psychosis fingerprint showed a marked overlap to changes seen in the brain proteome of schizophrenia patients. These findings suggest that it may be possible to contribute to the disease understanding by distinguishing different subtypes of MDD based on distinct brain proteomic profiles.
Collapse
Affiliation(s)
- D Martins-de-Souza
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| | - P C Guest
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - L W Harris
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - N Vanattou-Saifoudine
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - M J Webster
- Stanley Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD, USA
| | - H Rahmoune
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - S Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK,Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands,Department of Chemical Engineering and Biotechnology, University of Cambridge, Tennis Court Road, Cambridge, Cambridgeshire CB2 1QT, UK. E-mails: or
| |
Collapse
|
40
|
Powell SB, Weber M, Geyer MA. Genetic models of sensorimotor gating: relevance to neuropsychiatric disorders. Curr Top Behav Neurosci 2012; 12:251-318. [PMID: 22367921 PMCID: PMC3357439 DOI: 10.1007/7854_2011_195] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sensorimotor gating, or the ability of a sensory event to suppress a motor response, can be measured operationally via prepulse inhibition (PPI) of the startle response. PPI is deficient in schizophrenia patients as well as other neuropsychiatric disorders, can be measured across species, and has been used widely as a translational tool in preclinical neuropharmacological and genetic research. First developed to assess drug effects in pharmacological and developmental models, PPI has become one of the standard behavioral measures in genetic models of schizophrenia and other neuropsychiatric disorders that exhibit PPI deficits. In this chapter we review the literature on genetic models of sensorimotor gating and discuss the utility of PPI as a tool in phenotyping mutant mouse models. We highlight the approaches to genetic mouse models of neuropsychiatric disease, discuss some of the important caveats to these approaches, and provide a comprehensive table covering the more recent genetic models that have evaluated PPI.
Collapse
Affiliation(s)
- Susan B. Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Martin Weber
- Department of Neuroscience, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Mark A. Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
41
|
Krieglstein K, Zheng F, Unsicker K, Alzheimer C. More than being protective: functional roles for TGF-β/activin signaling pathways at central synapses. Trends Neurosci 2011; 34:421-9. [PMID: 21742388 DOI: 10.1016/j.tins.2011.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
It is becoming increasingly clear that members of the transforming growth factor-β (TGF-β) family have roles in the central nervous system that extend beyond their well-established roles as neurotrophic and neuroprotective factors. Recent findings have indicated that the TGF-β signaling pathways are involved in the modulation of both excitatory and inhibitory synaptic transmission in the adult mammalian brain. In this review, we discuss how TGF-β, bone morphogenetic protein and activin signaling at central synapses modulate synaptic plasticity, cognition and affective behavior. We also discuss the implications of these findings for the molecular understanding and potential treatment of neuropsychiatric diseases, such as anxiety, depression and other neurological disorders.
Collapse
Affiliation(s)
- Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | |
Collapse
|
42
|
Lu HL, Tanguy S, Rispe C, Gauthier JP, Walsh T, Gordon K, Edwards O, Tagu D, Chang CC, Jaubert-Possamai S. Expansion of genes encoding piRNA-associated argonaute proteins in the pea aphid: diversification of expression profiles in different plastic morphs. PLoS One 2011; 6:e28051. [PMID: 22162754 PMCID: PMC3230593 DOI: 10.1371/journal.pone.0028051] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 10/31/2011] [Indexed: 12/04/2022] Open
Abstract
Piwi-interacting RNAs (piRNAs) are known to regulate transposon activity in germ cells of several animal models that propagate sexually. However, the role of piRNAs during asexual reproduction remains almost unknown. Aphids that can alternate sexual and asexual reproduction cycles in response to seasonal changes of photoperiod provide a unique opportunity to study piRNAs and the piRNA pathway in both reproductive modes. Taking advantage of the recently sequenced genome of the pea aphid Acyrthosiphon pisum, we found an unusually large lineage-specific expansion of genes encoding the Piwi sub-clade of Argonaute proteins. In situ hybridisation showed differential expressions between the duplicated piwi copies: while Api-piwi2 and Api-piwi6 are "specialised" in germ cells their most closely related copy, respectively Api-piwi5 and Api-piwi3, are expressed in the somatic cells. The differential expression was also identified in duplicated ago3: Api-ago3a in germ cells and Api-ago3b in somatic cells. Moreover, analyses of expression profiles of the expanded piwi and ago3 genes by semi-quantitative RT-PCR showed that expressions varied according to the reproductive types. These specific expression patterns suggest that expanded aphid piwi and ago3 genes have distinct roles in asexual and sexual reproduction.
Collapse
Affiliation(s)
- Hsiao-ling Lu
- Department of Entomology/Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
- Research Centre for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Sylvie Tanguy
- UMR (Unité Mixte de Recherche) 1099 BiO3P (Biologie des Organismes et des Population appliquée à la Protection des Plantes) INRA (Institut National de la Rechercher Agronomique) – Agrocampus – Université Rennes1, Le Rheu, France
| | - Claude Rispe
- UMR (Unité Mixte de Recherche) 1099 BiO3P (Biologie des Organismes et des Population appliquée à la Protection des Plantes) INRA (Institut National de la Rechercher Agronomique) – Agrocampus – Université Rennes1, Le Rheu, France
| | - Jean-Pierre Gauthier
- UMR (Unité Mixte de Recherche) 1099 BiO3P (Biologie des Organismes et des Population appliquée à la Protection des Plantes) INRA (Institut National de la Rechercher Agronomique) – Agrocampus – Université Rennes1, Le Rheu, France
| | - Tom Walsh
- CSIRO (Commonwealth Scientific and Industrial Research Organisation) Ecosystem Sciences, Canberra, Australia
| | - Karl Gordon
- CSIRO (Commonwealth Scientific and Industrial Research Organisation) Ecosystem Sciences, Canberra, Australia
| | | | - Denis Tagu
- UMR (Unité Mixte de Recherche) 1099 BiO3P (Biologie des Organismes et des Population appliquée à la Protection des Plantes) INRA (Institut National de la Rechercher Agronomique) – Agrocampus – Université Rennes1, Le Rheu, France
| | - Chun-che Chang
- Department of Entomology/Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
- Research Centre for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| | - Stéphanie Jaubert-Possamai
- UMR (Unité Mixte de Recherche) 1099 BiO3P (Biologie des Organismes et des Population appliquée à la Protection des Plantes) INRA (Institut National de la Rechercher Agronomique) – Agrocampus – Université Rennes1, Le Rheu, France
| |
Collapse
|
43
|
Depino AM, Lucchina L, Pitossi F. Early and adult hippocampal TGF-β1 overexpression have opposite effects on behavior. Brain Behav Immun 2011; 25:1582-91. [PMID: 21640817 DOI: 10.1016/j.bbi.2011.05.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 04/29/2011] [Accepted: 05/16/2011] [Indexed: 10/18/2022] Open
Abstract
TGF-β1 is an anti-inflammatory cytokine that is augmented in the brain of autistic patients and that can affect brain development. In this work, we studied the effects of overexpressing TGF-β1 in the dentate gyrus of adult or young mice on behavior. TGF-β1 overexpression during postnatal development led to a long-term decrease in social interaction and to long-term increases in self-grooming and depression-related behaviors. Our analysis shows that these behavioral changes correlate with the long-term downregulation of TGF-β1 and IL-6 expression in the dentate gyrus, as well as to decreases in the mRNA levels of the synaptic protein neuroligin 3 and in the number of Reelin-positive neurons in the dentate gyrus. In contrast, chronic expression of TGF-β1 during adulthood led to transient opposite effects on these behaviors. These results show a central role of hippocampal TGF-β1 in the programming and modulation of social interaction, repetitive behavior and depression-related behavior. Finally, our data suggest a role of hippocampal TGF-β1 and early-life neuroinflammation in the development of the behavioral alterations observed in autism spectrum disorders.
Collapse
Affiliation(s)
- Amaicha Mara Depino
- Institute for Physiology, Molecular Biology and Neurosciences, CONICET-UBA, C1428EHA Buenos Aires, Argentina.
| | | | | |
Collapse
|
44
|
Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. TGFβ signaling in the brain increases with aging and signals to astrocytes and innate immune cells in the weeks after stroke. J Neuroinflammation 2010; 7:62. [PMID: 20937129 PMCID: PMC2958905 DOI: 10.1186/1742-2094-7-62] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 10/11/2010] [Indexed: 01/18/2023] Open
Abstract
Background TGFβ is both neuroprotective and a key immune system modulator and is likely to be an important target for future stroke therapy. The precise function of increased TGF-β1 after stroke is unknown and its pleiotropic nature means that it may convey a neuroprotective signal, orchestrate glial scarring or function as an important immune system regulator. We therefore investigated the time course and cell-specificity of TGFβ signaling after stroke, and whether its signaling pattern is altered by gender and aging. Methods We performed distal middle cerebral artery occlusion strokes on 5 and 18 month old TGFβ reporter mice to get a readout of TGFβ responses after stroke in real time. To determine which cell type is the source of increased TGFβ production after stroke, brain sections were stained with an anti-TGFβ antibody, colocalized with markers for reactive astrocytes, neurons, and activated microglia. To determine which cells are responding to TGFβ after stroke, brain sections were double-labelled with anti-pSmad2, a marker of TGFβ signaling, and markers of neurons, oligodendrocytes, endothelial cells, astrocytes and microglia. Results TGFβ signaling increased 2 fold after stroke, beginning on day 1 and peaking on day 7. This pattern of increase was preserved in old animals and absolute TGFβ signaling in the brain increased with age. Activated microglia and macrophages were the predominant source of increased TGFβ after stroke and astrocytes and activated microglia and macrophages demonstrated dramatic upregulation of TGFβ signaling after stroke. TGFβ signaling in neurons and oligodendrocytes did not undergo marked changes. Conclusions We found that TGFβ signaling increases with age and that astrocytes and activated microglia and macrophages are the main cell types that undergo increased TGFβ signaling in response to post-stroke increases in TGFβ. Therefore increased TGFβ after stroke likely regulates glial scar formation and the immune response to stroke.
Collapse
Affiliation(s)
- Kristian P Doyle
- Department of Neurology and Neurological Sciences, Stanford University Medical School, Stanford, CA 94305-5489, USA
| | | | | | | |
Collapse
|