1
|
Kochmanski J, Virani M, Kuhn NC, Boyd SL, Becker K, Adams M, Bernstein AI. Developmental origins of Parkinson's disease risk: perinatal exposure to the organochlorine pesticide dieldrin leads to sex-specific DNA modifications in critical neurodevelopmental pathways in the mouse midbrain. Toxicol Sci 2024; 201:263-281. [PMID: 38995845 PMCID: PMC11424889 DOI: 10.1093/toxsci/kfae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Abstract
Epidemiological studies show that exposure to the organochlorine pesticide dieldrin is associated with an increased risk of Parkinson's disease (PD). Animal studies support a link between developmental dieldrin exposure and increased neuronal susceptibility in the α-synuclein preformed fibril and MPTP models in adult male C57BL/6 mice. In a previous study, we showed that developmental dieldrin exposure was associated with sex-specific changes in DNA modifications within genes related to dopaminergic neuron development and maintenance at 12 wk of age. Here, we used capture hybridization-sequencing with custom baits to interrogate DNA modifications across the entire genetic loci of the previously identified genes at multiple time points-birth, 6, 12, and 36 wk old. We identified largely sex-specific dieldrin-induced changes in DNA modifications at each time point that annotated to pathways important for neurodevelopment, potentially related to critical steps in early neurodevelopment, dopaminergic neuron differentiation, synaptogenesis, synaptic plasticity, and glial-neuron interactions. Despite large numbers of age-specific DNA modifications, longitudinal analysis identified a small number of differential modification of cytosines with dieldrin-induced deflection of epigenetic aging. The sex-specificity of these results adds to evidence that sex-specific responses to PD-related exposures may underly sex-specific differences in disease. Overall, these data support the idea that developmental dieldrin exposure leads to changes in epigenetic patterns that persist after the exposure period and disrupt critical neurodevelopmental pathways, thereby impacting risk of late-life diseases, including PD.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Sierra L Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Katelyn Becker
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI 49503, United States
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI 49503, United States
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
- Environmental and Occupational Health Sciences Institute, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| |
Collapse
|
2
|
Hansson C, Hadžibajramović E, Svensson PA, Jonsdottir IH. Increased plasma levels of neuro-related proteins in patients with stress-related exhaustion: A longitudinal study. Psychoneuroendocrinology 2024; 167:107091. [PMID: 38964018 DOI: 10.1016/j.psyneuen.2024.107091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
Exhaustion disorder (ED) is a stress-related disorder characterized by physical and mental symptoms of exhaustion. Recent data suggest that pathophysiological processes in the central nervous system are involved in the biological mechanisms underlying ED. The aims of this study were to investigate if plasma levels of neuro-related proteins differ between patients with ED and healthy controls, and, if so, to investigate if these differences persist over time. Using the Olink Neuro Exploratory panel, we quantified the plasma levels of 92 neuro-related proteins in 163 ED patients at the time of diagnosis (baseline), 149 patients at long-term follow-up (7-12 years later, median follow-up time 9 years and 5 months), and 100 healthy controls. We found that the plasma levels of 40 proteins were significantly higher in the ED group at baseline compared with the control group. Out of these, the plasma levels of 36 proteins were significantly lower in the ED group at follow-up compared with the same group at baseline and the plasma levels of four proteins did not significantly differ between the groups. At follow-up, the plasma levels of two proteins were significantly lower in the ED group compared with the control group. These data support the hypothesis that pathophysiological processes in the central nervous system are involved in the biological mechanisms underlying ED.
Collapse
Affiliation(s)
- Caroline Hansson
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Emina Hadžibajramović
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingibjörg H Jonsdottir
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
3
|
Yuan R, Li Y, Li X, Fu Y, Ning A, Wang D, Zhang R, Yu S, Xu Q. Transcriptome analysis to explore the mechanism of downregulated TNIK influencing the effect of risperidone. Front Pharmacol 2024; 15:1431923. [PMID: 39268461 PMCID: PMC11391209 DOI: 10.3389/fphar.2024.1431923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Risperidone is one of the most reliable and effective antipsychotics for schizophrenia treatment. However, the mechanism of action of risperidone is not yet fully understood. Traf2 and Nck-interacting protein kinase (TNIK), a schizophrenia susceptibility gene, is associated with risperidone treatment response. Our previous in vitro experiments confirmed that downregulated TNIK affected the effect of risperidone on downstream targets. However, the effect of downregulated TNIK on risperidone-induced molecular expression remains to be further explored. Methods Transcriptome analysis was performed on U251 cells subjected to risperidone, TNIK siRNA, and no treatment, respectively. Compared to the no-treatment group, two groups of DEGs were screened out and then intersected with the schizophrenia-related genes to screen the cross-talk genes. Those DEGs were analyzed using GO and KEGG. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network for the cross-talk gene. Results The results showed that the parathyroid hormone synthesis, secretion, and action were significantly enriched after risperidone treatment. Downregulated TNIK could have an impact on the collagen-containing extracellular matrix, signaling receptor activator activity, and PI3K-Akt signaling pathway. Interestingly, bone mineralization function and calcium signaling pathway were enriched in the cross-talk genes. Additionally, FGFR2, FGF1, and FGFR might be the potential targets for TNIK affecting the effects of risperidone. Conclusion The study indicated that risperidone primarily influences functions and/or pathways associated with bone metabolism, potentially contributing to the adverse effect of osteoporosis. Our study may offer a novel perspective on investigating the mechanisms underlying the adverse effects of risperidone.
Collapse
Affiliation(s)
- Ruixue Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaojing Li
- 958 Hospital of PLA ARMY, Chongqing, China
| | - Xiangyi Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yingmei Fu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ailing Ning
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongxiang Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunying Yu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Kochmanski J, Virani M, Kuhn NC, Boyd SL, Becker K, Adams M, Bernstein AI. Developmental origins of Parkinson's disease risk: perinatal exposure to the organochlorine pesticide dieldrin leads to sex-specific DNA modifications in critical neurodevelopmental pathways in the mouse midbrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590998. [PMID: 38746441 PMCID: PMC11092502 DOI: 10.1101/2024.04.26.590998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Epidemiological studies show that exposure to the organochlorine pesticide dieldrin is associated with increased risk of Parkinson's disease (PD). Animal studies support a link between developmental dieldrin exposure and increased neuronal susceptibility in the α-synuclein preformed fibril (α-syn PFF) and MPTP models in adult male C57BL/6 mice. In a previous study, we showed that developmental dieldrin exposure was associated with sex-specific changes in DNA modifications within genes related to dopaminergic neuron development and maintenance at 12 weeks of age. Here, we used capture hybridization-sequencing with custom baits to interrogate DNA modifications across the entire genetic loci of the previously identified genes at multiple time points - birth, 6 weeks, 12 weeks, and 36 weeks old. We identified largely sex-specific dieldrin-induced changes in DNA modifications at each time point that annotated to pathways important for neurodevelopment, potentially related to critical steps in early neurodevelopment, dopaminergic neuron differentiation, synaptogenesis, synaptic plasticity, and glial-neuron interactions. Despite large numbers of age-specific DNA modifications, longitudinal analysis identified a small number of DMCs with dieldrin-induced deflection of epigenetic aging. The sex-specificity of these results adds to evidence that sex-specific responses to PD-related exposures may underly sex-specific differences in disease. Overall, these data support the idea that developmental dieldrin exposure leads to changes in epigenetic patterns that persist after the exposure period and disrupt critical neurodevelopmental pathways, thereby impacting risk of late life diseases, including PD.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Nathan C. Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Sierra L. Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Katelyn Becker
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Alison I. Bernstein
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Environmental and Occupational Health Sciences Institute, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| |
Collapse
|
5
|
Stevens HE, Scuderi S, Collica SC, Tomasi S, Horvath TL, Vaccarino FM. Neonatal loss of FGFR2 in astroglial cells affects locomotion, sociability, working memory, and glia-neuron interactions in mice. Transl Psychiatry 2023; 13:89. [PMID: 36906620 PMCID: PMC10008554 DOI: 10.1038/s41398-023-02372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023] Open
Abstract
Fibroblast growth factor receptor 2 (FGFR2) is almost exclusively expressed in glial cells in postnatal mouse brain, but its impact in glia for brain behavioral functioning is poorly understood. We compared behavioral effects from FGFR2 loss in both neurons and astroglial cells and from FGFR2 loss in astroglial cells by using either the pluripotent progenitor-driven hGFAP-cre or the tamoxifen-inducible astrocyte-driven GFAP-creERT2 in Fgfr2 floxed mice. When FGFR2 was eliminated in embryonic pluripotent precursors or in early postnatal astroglia, mice were hyperactive, and had small changes in working memory, sociability, and anxiety-like behavior. In contrast, FGFR2 loss in astrocytes starting at 8 weeks of age resulted only in reduced anxiety-like behavior. Therefore, early postnatal loss of FGFR2 in astroglia is critical for broad behavioral dysregulation. Neurobiological assessments demonstrated that astrocyte-neuron membrane contact was reduced and glial glutamine synthetase expression increased only by early postnatal FGFR2 loss. We conclude that altered astroglial cell function dependent on FGFR2 in the early postnatal period may result in impaired synaptic development and behavioral regulation, modeling childhood behavioral deficits like attention deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Hanna E Stevens
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Psychiatry, Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA.
| | - Soraya Scuderi
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Sarah C Collica
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Simone Tomasi
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Tamas L Horvath
- Department of Neuroscience, Yale University, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Department of Obstetrics and Gynecology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Flora M Vaccarino
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Neuroscience, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
6
|
Poletti S, Paolini M, Ernst J, Bollettini I, Melloni E, Vai B, Harrington Y, Bravi B, Calesella F, Lorenzi C, Zanardi R, Benedetti F. Long-term effect of childhood trauma: Role of inflammation and white matter in mood disorders. Brain Behav Immun Health 2022; 26:100529. [PMID: 36237478 PMCID: PMC9550612 DOI: 10.1016/j.bbih.2022.100529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 10/27/2022] Open
Abstract
Bipolar disorder (BD) and major depressive disorder (MDD) are severe psychiatric illnesses that share among their environmental risk factors the exposure to adverse childhood experiences (ACE). Exposure to ACE has been associated with long-term changes in brain structure and the immune response. In the lasts decades, brain abnormalities including alterations of white matter (WM) microstructure and higher levels of peripheral immune/inflammatory markers have been reported in BD and MDD and an association between inflammation and WM microstructure has been shown. However, differences in these measures have been reported by comparing the two diagnostic groups. The aim of the present study was to investigate the interplay between ACE, inflammation, and WM in BD and MDD. We hypothesize that inflammation will mediate the association between ACE and WM and that this will be different in the two groups. A sample of 200 patients (100 BD, 100 MDD) underwent 3T MRI scan and ACE assessment through Childhood Trauma Questionnaire. A subgroup of 130 patients (75 MDD and 55 BD) underwent blood sampling for the assessment of immune/inflammatory markers. We observed that ACE associated with higher peripheral levels of IL-2, IL-17, bFGF, IFN-γ, TNF-α, CCL3, CCL4, CCL5, and PDGF-BB only in the BD group. Further, higher levels of CCL3 and IL-2 associated with lower FA in BD. ACE were found to differently affect WM microstructure in the two diagnostic groups and to be negatively associated with FA and AD in BD patients. Mediation analyses showed a significant indirect effect of ACE on WM microstructure mediated by IL-2. Our findings suggest that inflammation may mediate the detrimental effect of early experiences on brain structure and different mechanisms underlying brain alterations in BD and MDD.
Collapse
Affiliation(s)
- Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy,Corresponding author. San Raffaele Turro, Via Stamira d’Ancona 20, 20127, Milano, Italy.
| | - Marco Paolini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Julia Ernst
- Vita-Salute San Raffaele University, Milano, Italy
| | - Irene Bollettini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy
| | - Elisa Melloni
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Benedetta Vai
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Yasmin Harrington
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Beatrice Bravi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Federico Calesella
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milano, Italy,Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
7
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
8
|
Lago SG, Bahn S. The druggable schizophrenia genome: from repurposing opportunities to unexplored drug targets. NPJ Genom Med 2022; 7:25. [PMID: 35338153 PMCID: PMC8956592 DOI: 10.1038/s41525-022-00290-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/04/2022] [Indexed: 12/04/2022] Open
Abstract
There have been no new drugs for the treatment of schizophrenia in several decades and treatment resistance represents a major unmet clinical need. The drugs that exist are based on serendipitous clinical observations rather than an evidence-based understanding of disease pathophysiology. In the present review, we address these bottlenecks by integrating common, rare, and expression-related schizophrenia risk genes with knowledge of the druggability of the human genome as a whole. We highlight novel drug repurposing opportunities, clinical trial candidates which are supported by genetic evidence, and unexplored therapeutic opportunities in the lesser-known regions of the schizophrenia genome. By identifying translational gaps and opportunities across the schizophrenia disease space, we discuss a framework for translating increasingly well-powered genetic association studies into personalized treatments for schizophrenia and initiating the vital task of characterizing clinically relevant drug targets in underexplored regions of the human genome.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Puebla M, Tapia PJ, Espinoza H. Key Role of Astrocytes in Postnatal Brain and Retinal Angiogenesis. Int J Mol Sci 2022; 23:ijms23052646. [PMID: 35269788 PMCID: PMC8910249 DOI: 10.3390/ijms23052646] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Angiogenesis is a key process in various physiological and pathological conditions in the nervous system and in the retina during postnatal life. Although an increasing number of studies have addressed the role of endothelial cells in this event, the astrocytes contribution in angiogenesis has received less attention. This review is focused on the role of astrocytes as a scaffold and in the stabilization of the new blood vessels, through different molecules release, which can modulate the angiogenesis process in the brain and in the retina. Further, differences in the astrocytes phenotype are addressed in glioblastoma, one of the most devastating types of brain cancer, in order to provide potential targets involved in the cross signaling between endothelial cells, astrocytes and glioma cells, that mediate tumor progression and pathological angiogenesis. Given the relevance of astrocytes in angiogenesis in physiological and pathological conditions, future studies are required to better understand the interrelation between endothelial and astrocyte signaling pathways during this process.
Collapse
Affiliation(s)
- Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Av. Plaza 680, Las Condes, Santiago 7550000, Chile;
| | - Pablo J. Tapia
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Av. Lota 2465, Providencia, Santiago 7500000, Chile;
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Av. República 71, Santiago 8320000, Chile
| | - Hilda Espinoza
- Facultad de Ciencias de la Salud, Universidad del Alba, Av. Ejército Libertador 171, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
10
|
Li XL, Yu Y, Hu Y, Wu HT, Li XS, Chen GY, Cheng Y. Fibroblast Growth Factor 9 as a Potential Biomarker for Schizophrenia. Front Psychiatry 2022; 13:788677. [PMID: 35546939 PMCID: PMC9082542 DOI: 10.3389/fpsyt.2022.788677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/14/2022] [Indexed: 11/26/2022] Open
Abstract
Preclinical and clinical studies have suggested that fibroblast growth factor (FGF) system contributed to the onset and development of schizophrenia (SCZ). However, there was no strong clinical evidence to link an individual FGF with SCZ. In this study, we aim to measure blood FGF9 levels in the patients with SCZ with and/or without medication, and test whether FGF9 has a potential to be a biomarker for SCZ. We recruited 130 patients with SCZ and 111 healthy individuals, and the ELISA and qRT-PCR assays were used to measure serum FGF9 levels in the participants. ELISA assay demonstrated that serum FGF9 protein levels were dramatically reduced in first-episode, drug-free patients, but not in chronically medicated patients when compared to healthy control subjects. Further analysis showed that treatment of the first-episode, drug-free SCZ patients with antipsychotics for 8 weeks significantly increased the serum FGF9 levels. In addition, we found that blood FGF9 mRNA levels were significantly lower in first-onset SCZ patients than controls. Under the receiver operating characteristic curve, the optimal cutoff values for FGF9 protein level as an indicator for diagnosis of drug-free SCZ patients was projected to be 166.4 pg/ml, which yielded a sensitivity of 0.955 and specificity of 0.86, and the area under the curve was 0.973 (95% CI, 0.954-0.993). Furthermore, FGF9 had good performance to discriminate between drug-free SCZ patients and chronically medicated patients, the optimal cutoff value for FGF9 concentration was projected to be 165.035 pg/ml with a sensitivity of 0.86 and specificity of 0.919, and the AUC was 0.968 (95% CI, 0.944, 0.991). Taken together, our results for the first time demonstrated the dysregulation of FGF9 in SCZ, and FGF9 has the potential to be served as a biomarker for SCZ.
Collapse
Affiliation(s)
- Xiao-Ling Li
- The Third People's Hospital of Foshan, Foshan, China.,Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yun Yu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Hu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Huan-Tong Wu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xue-Song Li
- The Third People's Hospital of Foshan, Foshan, China
| | - Guang-Yang Chen
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| |
Collapse
|
11
|
Klimaschewski L, Claus P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol Neurobiol 2021; 58:3884-3902. [PMID: 33860438 PMCID: PMC8280051 DOI: 10.1007/s12035-021-02367-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
12
|
Sasabayashi D, Takahashi T, Takayanagi Y, Suzuki M. Anomalous brain gyrification patterns in major psychiatric disorders: a systematic review and transdiagnostic integration. Transl Psychiatry 2021; 11:176. [PMID: 33731700 PMCID: PMC7969935 DOI: 10.1038/s41398-021-01297-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Anomalous patterns of brain gyrification have been reported in major psychiatric disorders, presumably reflecting their neurodevelopmental pathology. However, previous reports presented conflicting results of patients having hyper-, hypo-, or normal gyrification patterns and lacking in transdiagnostic consideration. In this article, we systematically review previous magnetic resonance imaging studies of brain gyrification in schizophrenia, bipolar disorder, major depressive disorder, and autism spectrum disorder at varying illness stages, highlighting the gyral pattern trajectory for each disorder. Patients with each psychiatric disorder may exhibit deviated primary gyri formation under neurodevelopmental genetic control in their fetal life and infancy, and then exhibit higher-order gyral changes due to mechanical stress from active brain changes (e.g., progressive reduction of gray matter volume and white matter integrity) thereafter, representing diversely altered pattern trajectories from those of healthy controls. Based on the patterns of local connectivity and changes in neurodevelopmental gene expression in major psychiatric disorders, we propose an overarching model that spans the diagnoses to explain how deviated gyral pattern trajectories map onto clinical manifestations (e.g., psychosis, mood dysregulation, and cognitive impairments), focusing on the common and distinct gyral pattern changes across the disorders in addition to their correlations with specific clinical features. This comprehensive understanding of the role of brain gyrification pattern on the pathophysiology may help to optimize the prediction and diagnosis of psychiatric disorders using objective biomarkers, as well as provide a novel nosology informed by neural circuits beyond the current descriptive diagnostics.
Collapse
Affiliation(s)
- Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan. .,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.
| | - Tsutomu Takahashi
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,grid.267346.20000 0001 2171 836XResearch Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Yoichiro Takayanagi
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,Arisawabashi Hospital, Toyama, Japan
| | - Michio Suzuki
- grid.267346.20000 0001 2171 836XDepartment of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan ,grid.267346.20000 0001 2171 836XResearch Center for Idling Brain Science, University of Toyama, Toyama, Japan
| |
Collapse
|
13
|
Sarigecili E, Makharoblidze K, Çobanogullari MD, Yildirim DD, Komur M, Okuyaz C. Neurodevelopmental risk evaluation of premature closure of the anterior fontanelle. Childs Nerv Syst 2021; 37:561-566. [PMID: 32737565 DOI: 10.1007/s00381-020-04846-6] [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] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 11/24/2022]
Abstract
PURPOSE To evaluate neurological development of completely healthy children with anterior fontanelle premature closure via Denver Developmental Screening Test II and to compare the results with control group. METHOD AND RESULTS The records of 140 patients applied to Mersin University Pediatric Neurology Outpatient Clinic between 2011 and 2019 with the complaint of premature closure of the anterior fontanelle were retrospectively reviewed. Patients with microcephaly, craniosynostosis, infection, sequelae of hypoxia-ischemia, metabolic disorders, intracranial hemorrhage, epilepsy, endocrine problems, and dysmorphic features were excluded from the study. Sixty-six completely healthy children with anterior fontanelle premature closure were included in the study. Denver Developmental Screening Test II was performed by the same developmental specialist to the children with premature closure of the anterior fontanelle as well as to the healthy control group. For each child included in the case and the control group, 90% of the values for each development area were calculated and recorded. Then, the results were compared. Denver II Developmental Screening Test (p < 0.001) and gross motor subtest (p < 0.001) results showed statistically significant retardation in the case group compared with the control group. CONCLUSIONS The study was the first study in the literature on the gross motor development of children with premature closure of anterior fontanelle, and it has been found significantly undeveloped compared with the control group, and it has been concluded that similar patients should be evaluated from this view point in pediatric neurology department.
Collapse
Affiliation(s)
- Esra Sarigecili
- Faculty of Medicine, Department of Pediatric Neurology, Mersin University, Mersin, Turkey.
| | - Khatuna Makharoblidze
- Faculty of Medicine, Department of Pediatric Neurology, Mersin University, Mersin, Turkey
| | | | | | - Mustafa Komur
- Faculty of Medicine, Department of Pediatric Neurology, Mersin University, Mersin, Turkey
| | - Cetin Okuyaz
- Faculty of Medicine, Department of Pediatric Neurology, Mersin University, Mersin, Turkey
| |
Collapse
|
14
|
Expression of fibroblast growth factor 9 and its receptors in the dentate gyrus of hippocampus in poststroke depression rats. Neuroreport 2021; 32:321-325. [PMID: 33470768 DOI: 10.1097/wnr.0000000000001591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Studies have found that fibroblast growth factor 9 (FGF9) might have a negative effect in the psychiatric diseases, such as depression or anxiety, but its potential role in the pathophysiology of poststroke depression (PSD) remains uncertain. Here, we set out to investigate the expression changes of FGF9 and its receptors in PSD rats. Middle cerebral artery occlusion (MCAO) combined with chronic unpredictable mild stress was used to establish the PSD rat model. Then, the rats were randomly divided into four groups: control (sham-operation), MCAO, PSD and treated (fluoxetine injection by intraperitoneal). Weight measurement, sucrose preference test, open-field test and forced swim test were performed to evaluate the behavioral changes, and then Western blot and real-time quantitative PCR were used to detect the expression level of FGF9, fibroblast growth factor receptor 1 (FGFR1) and receptor 3 (FGFR3) in the dentate gyrus of rat hippocampus. We found that FGF9 protein and mRNA expression increased significantly in the MCAO and PSD groups; FGFR3 protein and mRNA expression decreased significantly in the MCAO and PSD groups; FGFR1 protein and mRNA expression decreased significantly in the PSD group, but increased in the treated group. Furthermore, the changes mentioned above were reversed obviously by fluoxetine. These results indicated that upregulated FGF9 expression and downregulated FGFR1 and FGFR3 expression may be involved in the pathogenesis of PSD, and the FGF9/FGFR signaling pathway may be considered as an attractive target for further study.
Collapse
|
15
|
Kon E, Calvo-Jiménez E, Cossard A, Na Y, Cooper JA, Jossin Y. N-cadherin-regulated FGFR ubiquitination and degradation control mammalian neocortical projection neuron migration. eLife 2019; 8:47673. [PMID: 31577229 PMCID: PMC6786859 DOI: 10.7554/elife.47673] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022] Open
Abstract
The functions of FGF receptors (FGFRs) in early development of the cerebral cortex are well established. Their functions in the migration of neocortical projection neurons, however, are unclear. We have found that FGFRs regulate multipolar neuron orientation and the morphological change into bipolar cells necessary to enter the cortical plate. Mechanistically, our results suggest that FGFRs are activated by N-Cadherin. N-Cadherin cell-autonomously binds FGFRs and inhibits FGFR K27- and K29-linked polyubiquitination and lysosomal degradation. Accordingly, FGFRs accumulate and stimulate prolonged Erk1/2 phosphorylation. Neurons inhibited for Erk1/2 are stalled in the multipolar zone. Moreover, Reelin, a secreted protein regulating neuronal positioning, prevents FGFR degradation through N-Cadherin, causing Erk1/2 phosphorylation. These findings reveal novel functions for FGFRs in cortical projection neuron migration, suggest a physiological role for FGFR and N-Cadherin interaction in vivo and identify Reelin as an extracellular upstream regulator and Erk1/2 as downstream effectors of FGFRs during neuron migration.
Collapse
Affiliation(s)
- Elif Kon
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Elisa Calvo-Jiménez
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alexia Cossard
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Youn Na
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
16
|
Yuan H, Ni X, Zheng M, Han X, Song Y, Yu M. Effect of catalpol on behavior and neurodevelopment in an ADHD rat model. Biomed Pharmacother 2019; 118:109033. [PMID: 31545235 DOI: 10.1016/j.biopha.2019.109033] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 10/26/2022] Open
Abstract
Studies suggest that abnormal neurodevelopment of prefrontal striatal circuits is implicated in the pathogenesis of attention deficit hyperactivity disorder (ADHD). In the present study, we investigated the effect of catalpol, an active ingredient of Rehmanniae radix preparata, which is the most frequently used Chinese medicinal herb for the treatment of ADHD, on behavior and neurodevelopment in spontaneously hypertensive rats (SHR). SHR were divided into SHR group (vehicle, i.g.), methylphenidate (MPH) group (2 mg/kg/day, i.g.), and catalpol group (50 mg/kg/day i.g.), and Wistar-Kyoto (WKY) rats were used as control group (vehicle, i.g.). Open Field Test (OFT) and Morris water maze (MWM) test were performed to assess the effect of catalpol on behavior. Results revealed that both catalpol and MPH treatment decreased average speed, time spent in the central area, rearing times, and central area visits, increased the immobility time of SHR in OFT, and increased number of visits to the annulus, and time spent in target quadrant in the MWM test. Hematoxylin and eosin (H&E) staining showed that catalpol reduced irregular neuronal arrangement, ruptured nuclear membranes, and resulted in disappearance of the nucleolus in the prefrontal cortex (PFC) and striatum of SHR. Moreover, immuno-fluorescent staining of NeuN and myelin basic protein (MBP) indicated that catalpol ameliorated neuronal loss and contributed to myelination. Finally, western blot and immunostaining analysis suggested that several regulatory proteins involved in PFC development were up-regulated by catalpol treatment, such as brain-derived neurotrophic factor (BDNF), cyclin-dependent kinase 5 (Cdk5), p35, fibroblast growth factor (FGF) 21 and its receptor (FGFR)1. Taken together, catalpol can effectively ameliorate hyperactive and impulsive behavior, improve spatial learning and memory in SHR, likely through the neurodevelopmental pathways. Nonetheless, whether catalpol could attenuate inattention in SHR and the pathway by which catalpol reduces neuronal loss remain to be further studied.
Collapse
Affiliation(s)
- Haixia Yuan
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Xinqiang Ni
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| | - Min Zheng
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| | - Xinmin Han
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Yuchen Song
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Minfeng Yu
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| |
Collapse
|
17
|
Hövel FFV, Leiter I, Rumpel R, Langenhagen A, Wedekind D, Häger C, Bleich A, Palme R, Grothe C. FGF-2 isoforms influence the development of dopaminergic neurons in the murine substantia nigra, but not anxiety-like behavior, stress susceptibility, or locomotor behavior. Behav Brain Res 2019; 374:112113. [PMID: 31381976 DOI: 10.1016/j.bbr.2019.112113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Loss of fibroblast growth factor 2 (FGF-2) is responsible for the development of an increased number of dopaminergic (DA) neurons in the murine substantia nigra pars compacta (SNpc). Furthermore, dysregulation of its expression patterns within the central nervous system (CNS) is associated with behavioral abnormalities in mice. Until now, the contributions of the individual FGF-2 isoforms (one low (LMW) and two high molecular weight (HMW) isoforms) in the CNS are elusive. METHODS To unravel the specific effects of FGF-2 isoforms, we compared three knockout mouse lines, one only deficient for LMW, one deficient for HMW and another lacking both isoforms, regarding DA neuronal development. With this regard, three time points of ontogenic development of the SNpc were stereologically investigated. Furthermore, behavioral aspects were analyzed in young adult mice, supplemented by corticosterone measurements. RESULTS Juvenile mice lacking either LMW or HMW develop equal supernumerary DA neuron numbers in the SNpc. Compensatory increased LMW expression is observed in animals lacking HMW. Meanwhile, no knockout mouse line demonstrated changes in anxiety-like behavior, stress susceptibility, or locomotor behavior. CONCLUSIONS Both FGF-2 isoforms crucially influence DA neuronal development in the murine SNpc. However, absence of LMW or HMW alone alters neither anxiety-like nor locomotor behavior, or stress susceptibility. Therefore, FGF-2 is not a determinant and causative factor for behavioral alterations alone, but probably in combination with appropriate conditions, like environmental or genetic factors.
Collapse
Affiliation(s)
- Friederike Freiin von Hövel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany; Center for Systems Neuroscience (ZSN), Hanover, Germany
| | - Ina Leiter
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany; Center for Systems Neuroscience (ZSN), Hanover, Germany
| | - Regina Rumpel
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany
| | - Alina Langenhagen
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany; Center for Systems Neuroscience (ZSN), Hanover, Germany
| | - Dirk Wedekind
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany
| | - Christine Häger
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany
| | - Rupert Palme
- Unit of Physiology, Pathophysiology and Experimental Endocrinology, Department of Biomedical Sciences, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hanover, Germany; Center for Systems Neuroscience (ZSN), Hanover, Germany.
| |
Collapse
|
18
|
Dysregulation of Fibroblast Growth Factor 10 in the Peripheral Blood of Patients with Schizophrenia. J Mol Neurosci 2019; 69:69-74. [PMID: 31256336 DOI: 10.1007/s12031-019-01331-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023]
Abstract
The fibroblast growth factor (FGF) system has been suggested to be involved in the development of schizophrenia (SCZ). However, the potential roles of all FGFs have not been well studied in the literature. Here, we investigated the concentration of peripheral blood fibroblast 10 (FGF10) in patients with SCZ to determine whether FGF10 could serve as a biomarker for SCZ. We recruited 130 SCZ patients (57 first-episode, drug-free patients and 73 chronically medicated patients) and 111 healthy controls. Our results showed that serum FGF10 levels were significantly decreased in SCZ patients when compared with controls. Sub-group analyses revealed that both first-episode, drug-free patients and chronically medicated patients had lower levels of FGF10 than controls. Moreover, both male and female SCZ patients had significantly decreased blood FGF10 levels relative to control subjects. Using a receiver operating characteristic curve, the optimal cutoff value of FGF10 level as an indicator for diagnosis of first-onset SCZ patients was projected to be 152.3 pg/ml, which yielded a sensitivity of 0.658 and specificity of 0.649, with an area under the curve of 0.665 (95% confidence interval, 0.577-0.754). Taken together, our results are the first to demonstrate an association between FGF10 and SCZ, providing further evidence for the neurotrophic factor hypothesis of SCZ.
Collapse
|
19
|
Kroeze Y, Oti M, van Beusekom E, Cooijmans RHM, van Bokhoven H, Kolk SM, Homberg JR, Zhou H. Transcriptome Analysis Identifies Multifaceted Regulatory Mechanisms Dictating a Genetic Switch from Neuronal Network Establishment to Maintenance During Postnatal Prefrontal Cortex Development. Cereb Cortex 2019; 28:833-851. [PMID: 28108491 DOI: 10.1093/cercor/bhw407] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Indexed: 12/20/2022] Open
Abstract
The prefrontal cortex (PFC) is one of the latest brain regions to mature, which allows the acquisition of complex cognitive abilities through experience. To unravel the underlying gene expression changes during postnatal development, we performed RNA-sequencing (RNA-seq) in the rat medial PFC (mPFC) at five developmental time points from infancy to adulthood, and analyzed the differential expression of protein-coding genes, long intergenic noncoding RNAs (lincRNAs), and alternative exons. We showed that most expression changes occur in infancy, and that the number of differentially expressed genes reduces toward adulthood. We observed 137 differentially expressed lincRNAs and 796 genes showing alternative exon usage during postnatal development. Importantly, we detected a genetic switch from neuronal network establishment in infancy to maintenance of neural networks in adulthood based on gene expression dynamics, involving changes in protein-coding and lincRNA gene expression as well as alternative exon usage. Our gene expression datasets provide insights into the multifaceted transcriptional regulation of the developing PFC. They can be used to study the basic developmental processes of the mPFC and to understand the mechanisms of neurodevelopmental and neuropsychiatric disorders. Our study provides an important contribution to the ongoing efforts to complete the "brain map", and to the understanding of PFC development.
Collapse
Affiliation(s)
- Yvet Kroeze
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EZ Nijmegen, The Netherlands.,Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Martin Oti
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands.,Carlos Chagas Filho Biophysics Institute (IBCCF), Federal University of Rio de Janeiro (UFRJ), 21941-902 Rio de Janeiro, Brazil
| | - Ellen van Beusekom
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Roel H M Cooijmans
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Sharon M Kolk
- Department of Molecular Animal Physiology, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University, 6525 GA Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EZ Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.,Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
20
|
Zhang R, Ma Z, Liu K, Li Y, Liu D, Xu L, Deng X, Qu R, Ma Z, Ma S. Baicalin exerts antidepressant effects through Akt/FOXG1 pathway promoting neuronal differentiation and survival. Life Sci 2019; 221:241-248. [DOI: 10.1016/j.lfs.2019.02.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
|
21
|
Deng Z, Deng S, Zhang MR, Tang MM. Fibroblast Growth Factors in Depression. Front Pharmacol 2019; 10:60. [PMID: 30804785 PMCID: PMC6370647 DOI: 10.3389/fphar.2019.00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Major depressive disorder (MDD) is one of the most serious diseases and now becomes a major public health problem in the world. The pathogenesis of depression remains poorly understood. Fibroblast growth factors (FGFs) belong to a large family of growth factors that are involved in brain development during early periods as well as maintenance and repair throughout adulthood. In recent years, studies have found a correlation between the members of the FGF system and depression. These signaling molecules may be expected to be biomarkers for the diagnosis and prognosis of MDD, and may provide new drug targets for the treatment of depression. Here, we reviewed the correlation between some members of the FGF system and depression.
Collapse
Affiliation(s)
- Zheng Deng
- Hospital Evaluation Office, Xiangya Hospital, Central South University, Changsha, China
| | - Sheng Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute of Hospital Pharmacy, Central South University, Changsha, China
| | - Mu-Rong Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute of Hospital Pharmacy, Central South University, Changsha, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mi-Mi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute of Hospital Pharmacy, Central South University, Changsha, China
| |
Collapse
|
22
|
FGFR2 gene polymorphism rs2981582 is associated with non-functioning pituitary adenomas in Chinese Han population: a case-control study. Biosci Rep 2018; 38:BSR20181081. [PMID: 30206133 PMCID: PMC6239272 DOI: 10.1042/bsr20181081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/23/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022] Open
Abstract
The association of the fibroblast growth factor receptor 2 gene (FGFR2) polymorphism rs2981582 with breast cancer has been extensively studied, whereas the role of this polymorphism in non-functioning pituitary adenoma (NFPA) has not been elucidated. We thus investigated a potential association of rs2981582 with NFPA. A total of 79 patients and 142 healthy control participants were enrolled in our study. DNA of the participants was extracted from peripheral blood samples and genotyped by using the MassARRAY method. We found that the AA genotype was associated with a higher risk of developing NFPA (OR = 1.743, 95%CI: 1.151–2.64, P=0.008). After adjusting for risk factors, significant difference was still observed between the two groups (OR = 1.862, 95%CI: 1.172–2.957, P=0.008). Moreover, under the assumptions of the recessive model (OR = 3.051, 95%CI: 1.403–6.635, P=0.005) and the additive model (AG: OR = 0.329, 95%CI: 0.144–0.755, P=0.009; AA: OR = 0.326, 95%CI: 0.141–0.757, P=0.009), rs2981582 was associated with an increased risk of NFPA. Our results proved that FGFR2 rs2981582 AA genotype was associated with a higher risk of NFPA. The recessive model and additive model also showed increased the risk of NFPA.
Collapse
|
23
|
Yuan H, Yang M, Han X, Ni X. The Therapeutic Effect of the Chinese Herbal Medicine, Rehmanniae Radix Preparata, in Attention Deficit Hyperactivity Disorder via Reversal of Structural Abnormalities in the Cortex. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:3052058. [PMID: 30405737 PMCID: PMC6204205 DOI: 10.1155/2018/3052058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/02/2018] [Accepted: 09/10/2018] [Indexed: 12/27/2022]
Abstract
Rehmanniae radix preparata is extracted from wine-steaming the Rehmannia root, a scrophulariaceae plant. It has been used for thousands of years with effects of nourishing kidney-yin, benefiting essence and filling marrow based on traditional Chinese medicine (TCM) theory. Rehmanniae radix preparata has antioxidant, antisenescence, anti-inflammatory, and neuroprotective properties. It is the most popular Traditional Chinese medicinal compound (TCMC) used in attention deficit hyperactivity disorder (ADHD) therapy. However, few studies have been conducted exploring the effects and potential mechanisms of Rehmanniae radix preparata alone on ADHD. Recent studies have shown that Rehmanniae radix preparata inhibits spontaneous activity in mice, improves learning and memory in rats following thalamic arcuate nucleus injury, and exhibits antidepressant effects. Catalpol, an active component of Rehmanniae radix preparata, elevates brain-derived neurotrophic factor (BDNF), and attenuates neuronal apoptosis and energy metabolism failure. ADHD is characterized by hyperactivity-impulsivity and impairments in learning and memory. Its pathomechanism is closely related to structural abnormalities in the cortex that is mediated by dysfunction in neuronal development, apoptosis, and energy metabolism. We hypothesize that Rehmanniae radix preparata may be effective at treating ADHD by alleviating neurodevelopmental abnormalities, neuronal apoptosis, and energy metabolism failure.
Collapse
Affiliation(s)
- Haixia Yuan
- Nanjing University of Chinese Medicine, First Clinical Medical College, Institute of Pediatrics of traditional Chinese Medicine, Qixia District, Nanjing, 210029, Jiangsu Province, China
| | - Meng Yang
- Nanjing University of Chinese Medicine, Institute of Chinese medicine literature, Qixia District, Nanjing, 210029, Jiangsu Province, China
| | - Xinmin Han
- Nanjing University of Chinese Medicine, First Clinical Medical College, Institute of Pediatrics of traditional Chinese Medicine, Qixia District, Nanjing, 210029, Jiangsu Province, China
| | - Xinqiang Ni
- Shenzhen traditional Chinese Medicine Hospital, Pediatrics of traditional Chinese Medicine, Shenzhen, 518038, Guangdong Province, China
- Institute of Geriatrics, Shenzhen, 518035, Guangdong Province, China
| |
Collapse
|
24
|
Even-Chen O, Barak S. The role of fibroblast growth factor 2 in drug addiction. Eur J Neurosci 2018; 50:2552-2561. [PMID: 30144335 DOI: 10.1111/ejn.14133] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a member of the FGF-family, which consists of 22 members, with four known FGF receptors (five in humans). Over the last 30 years, FGF2 has been extensively studied for its role in cell proliferation, differentiation, growth, survival and angiogenesis during development, as well as for its role in adult neurogenesis and regenerative plasticity. Over the past decade, FGF2 has been implicated in learning and memory, as well as in several neuropsychiatric disorders, including anxiety, stress, depression and drug addiction. In this review, we present accumulating evidence indicating the involvement of FGF2 in neuroadaptations caused by drugs of abuse, namely, amphetamine, cocaine, nicotine and alcohol. Moreover, evidence suggests that FGF2 is a positive regulator of alcohol and drug-related behaviors. Thus, although additional studies are yet required, we suggest that reducing FGF2 activity may provide a novel therapeutic approach for substance use disorders.
Collapse
Affiliation(s)
- Oren Even-Chen
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
25
|
Abstract
Developmental dyslexia (DD) is a neurobiological disorder featured by reading disabilities. In recent years, genome-wide approaches provided new perspectives to discover novel candidate genes of DD. In a previous study, rs9313548 located downstream of FGF18 showed borderline genome-wide significant association with DD. Herein, we selected rs9313548 and 11 independent tag single nucleotide polymorphisms covering gene region of FGF18 to perform association analysis with DD among 978 Chinese dyslexic cases and 998 controls recruited from elementary schools. However, we did not observe any single nucleotide polymorphism exceeding significant threshold. Our preliminary results suggested that FGF18 might not be a susceptibility gene for DD in Chinese population.
Collapse
|
26
|
Numakawa T, Odaka H, Adachi N, Chiba S, Ooshima Y, Matsuno H, Nakajima S, Yoshimura A, Fumimoto K, Hirai Y, Kunugi H. Basic fibroblast growth factor increased glucocorticoid receptors in cortical neurons through MAP kinase pathway. Neurochem Int 2018; 118:217-224. [PMID: 29958871 DOI: 10.1016/j.neuint.2018.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 01/09/2023]
Abstract
Prolonged and intense stress chronically increases blood concentration of glucocorticoids, which in turn causes downregulation of glucocorticoid receptor (GR) in the central nervous system (CNS). This process has been suggested to be involved in the pathogenesis of major depressive disorder (MDD). Here, we found that basic fibroblast growth factor (bFGF) increased the expression of GR in the rat cerebral cortex and cultured cortical neurons and restored the reduced GR expression caused by glucocorticoid exposure. Among intracellular signaling pathways stimulated by bFGF, extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathway was responsible for the upregulation of GR. The bFGF-induced GR was functional as a transcription factor to enhance transcription of a target gene. Because high stress augments bFGF levels in the brain, it is likely that bFGF plays a compensating role for reduced GR expression after stress and thus should be studied as a therapeutic target for the treatment of MDD.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Haruki Odaka
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan.
| | - Shuichi Chiba
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Yoshiko Ooshima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Administrative Section of Radiation Protection, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Hitomi Matsuno
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Shingo Nakajima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Endowed Research Division of Human Welfare Sciences, Ochanomizu University, Tokyo, Japan
| | - Aya Yoshimura
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Aichi, Japan
| | - Kazuhiro Fumimoto
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Yohei Hirai
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
27
|
Haljas K, Amare AT, Alizadeh BZ, Hsu YH, Mosley T, Newman A, Murabito J, Tiemeier H, Tanaka T, van Duijn C, Ding J, Llewellyn DJ, Bennett DA, Terracciano A, Launer L, Ladwig KH, Cornelis MC, Teumer A, Grabe H, Kardia SLR, Ware EB, Smith JA, Snieder H, Eriksson JG, Groop L, Räikkönen K, Lahti J. Bivariate Genome-Wide Association Study of Depressive Symptoms With Type 2 Diabetes and Quantitative Glycemic Traits. Psychosom Med 2018; 80:242-251. [PMID: 29280852 PMCID: PMC6051528 DOI: 10.1097/psy.0000000000000555] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Shared genetic background may explain phenotypic associations between depression and Type 2 diabetes (T2D). We aimed to study, on a genome-wide level, if genetic correlation and pleiotropic loci exist between depressive symptoms and T2D or glycemic traits. METHODS We estimated single-nucleotide polymorphism (SNP)-based heritability and analyzed genetic correlation between depressive symptoms and T2D and glycemic traits with the linkage disequilibrium score regression by combining summary statistics of previously conducted meta-analyses for depressive symptoms by CHARGE consortium (N = 51,258), T2D by DIAGRAM consortium (N = 34,840 patients and 114,981 controls), fasting glucose, fasting insulin, and homeostatic model assessment of β-cell function and insulin resistance by MAGIC consortium (N = 58,074). Finally, we investigated pleiotropic loci using a bivariate genome-wide association study approach with summary statistics from genome-wide association study meta-analyses and reported loci with genome-wide significant bivariate association p value (p < 5 × 10). Biological annotation and function of significant pleiotropic SNPs were assessed in several databases. RESULTS The SNP-based heritability ranged from 0.04 to 0.10 in each individual trait. In the linkage disequilibrium score regression analyses, depressive symptoms showed no significant genetic correlation with T2D or glycemic traits (p > 0.37). However, we identified pleiotropic genetic variations for depressive symptoms and T2D (in the IGF2BP2, CDKAL1, CDKN2B-AS, and PLEKHA1 genes), and fasting glucose (in the MADD, CDKN2B-AS, PEX16, and MTNR1B genes). CONCLUSIONS We found no significant overall genetic correlations between depressive symptoms, T2D, or glycemic traits suggesting major differences in underlying biology of these traits. However, several potential pleiotropic loci were identified between depressive symptoms, T2D, and fasting glucose, suggesting that previously established phenotypic associations may be partly explained by genetic variation in these specific loci.
Collapse
Affiliation(s)
- Kadri Haljas
- From the Departments of Psychology and Logopedics (Haljas, Räikkönen) and Psychology and Logopedics, Faculty of Medicine (Lahti), and Helsinki Collegium for Advanced Studies (Lahti), University of Helsinki, Helsinki, Finland; Department of Epidemiology (Amare, Alizadeh, Snieder), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Harvard Medical School (Hsu), Boston, Massachusetts; Institute for Molecular Medicine Finland (FIMM) (Groop), Helsinki, Finland; Lund University Diabetes Centre (Groop), Lund University, Lund, Sweden; Department of General Practice and Primary Health Care (Eriksson), University of Helsinki and Helsinki University Hospital; Folkhälsan Research Center (Eriksson), Helsinki, Finland; Department of Medicine (Mosley), University of Mississippi Medical Center, Jackson, Mississippi; Department of Epidemiology, School of Public Health (Newman), University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Medicine, Section of General Internal Medicine (Murabito), Boston University School of Medicine, Boston; Boston University and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts (Murabito); Departments of Epidemiology and Psychiatry (Tiemeier), Erasmus University Medical Center, Rotterdam, the Netherlands; Translational Gerontology Branch (Tanaka), National Institute on Aging, Baltimore, Maryland; Genetic Epidemiology Unit, Department of Epidemiology (van Duijn), Erasmus University Medical Center, Rotterdam; Centre for Medical Systems Biology (van Duijn), Leiden, the Netherlands; Department of Internal Medicine, Division of Geriatrics (Ding), Wake Forest University, Winston-Salem, North Carolina; University of Exeter Medical School (Llewellyn), Exeter, UK; Rush Alzheimer's Disease Center (Bennett), Chicago, Illinois; Florida State University, College of Medicine (Terracciano), Tallahassee, Florida; Laboratory of Epidemiology and Population Sciences (Launer), National Institute on Aging, Bethesda, Maryland; Department of Psychiatry and Psychotherapy (Grabe), Helios Hospital Stralsund; Department of Psychiatry and Psychotherapy (Grabe) and Institute for Community Medicine (Teumer), University Medicine Greifswald; German Center for Neurodegenerative Diseases (Grabe), Site Rostock/Greifswald, Greifswald, Germany; Institute of Epidemiology II, Mental Health Research Unit, Helmholtz Zentrum München (Ladwig), German Research Center for Environmental Health, Neuherberg, Germany; Psychosomatic Medicine and Psychotherapy (Ladwig), Universitäts-Klinikum Rechts der Isar, Technische Universität München, Munich, Germany & German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Preventive Medicine (Cornelis), Northwestern University Feinberg School of Medicine, Chicago, Illinois; and Department of Epidemiology, School of Public Health (Kardia, Ware, Smith), and Survey Research Center, Institute for Social Research (Ware, Smith), University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Neurotrophins and neuroinflammation in fetuses exposed to maternal depression and anxiety disorders during pregnancy: a comparative study on cord blood. Arch Womens Ment Health 2018; 21:105-111. [PMID: 28884439 DOI: 10.1007/s00737-017-0774-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022]
Abstract
In recent years, there have been changes in the approach to maternal psychiatric disorders and their effects on the fetus, with the focus redirected to the search for biological markers. Neurotrophic factors and inflammatory processes have received particular attention in the past few years. According to the Structured Clinical Interview for Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), the study sample (n = 136) consisted of three groups: mothers with major depressive disorder (MDD group, n = 25), mothers with anxiety disorder (AD group, n = 18), and mothers without any psychiatric disorders (not diagnosed (ND) group, n = 93). During the delivery/cesarean section, a blood sample was obtained from the umbilical cord. Serum concentrations of BDNF, NT-3, FGF2, TNF-α, and neopterin were determined by enzyme-linked immunosorbent assay (ELISA), according to the manufacturer's procedure. Clinical and biochemical characteristics were assessed. We did not find a significant difference among the three study groups with regard to BDNF, NT-3, and TNF-α levels. The ANOVA test indicated statistically significant differences in FGF2 levels and neopterin between the study groups. The newborns of mothers with AD had significantly higher FGF2 levels and significantly higher neopterin levels when compared with those of mothers with MDD and healthy mothers. The present study sheds light on the effects of higher FGF2 and neopterin levels in fetuses exposed to AD. Our results should be replicated through further prospective studies with a larger sample size.
Collapse
|
29
|
Fgf9 Y162C Mutation Alters Information Processing and Social Memory in Mice. Mol Neurobiol 2017; 55:4580-4595. [DOI: 10.1007/s12035-017-0659-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/14/2017] [Indexed: 12/13/2022]
|
30
|
Mazgaj R, Tal A, Goetz R, Lazar M, Rothman K, Messinger JW, Malaspina D, Gonen O. Hypo-metabolism of the rostral anterior cingulate cortex associated with working memory impairment in 18 cases of schizophrenia. Brain Imaging Behav 2016; 10:115-23. [PMID: 25804309 DOI: 10.1007/s11682-015-9372-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Working memory (Work-Mem), the capacity to hold and manipulate information, activates the anterior cingulate cortex (ACC), especially its caudal subregion. Impaired Work-Mem and structural and functional abnormalities of the ACC are reported in schizophrenia. This study aims to elucidate the pathogenesis of Work-Mem dysfunction in schizophrenia by comparing metabolite concentrations across ACC subregions. This retrospective study of 18 schizophrenia cases and 10 matched controls used proton magnetic resonance spectroscopic imaging ((1)H-MRSI, TR/TE = 1800/35 ms, 0.5 cm(3) spatial resolution) to test whether the Work-Mem Index of the Wechsler Adult Intelligence Scale, third edition is associated with differences in the rostral to caudal ACC ratios of N-acetylaspartate (NAA) and creatine (Cr). Higher caudal:rostral ACC Cr (but not NAA) concentrations were associated with decreased Work-Mem Index in cases (r = -0.6, p = 0.02), with a similar trend in controls (r = -0.56, p = 0.10), although caudal:rostral ACC Cr correlated with NAA in cases and controls (r = 0.67 and 0.62, p < 0.05 for both). NAA and Cr ratios did not correlate with myo-inositol, excluding gliosis as the underlying process. Subjects' sex and age had no effects on these relationships. The findings suggest that rostral ACC energy hypo-metabolism, possibly arising from neurodevelopmental processes, is associated with working memory impairment in schizophrenia. Changes in the rostral (not the expected caudal) subregion underscore the interconnections between the ACC subregions and may offer laboratory markers for treatment trials, etiology studies, and perhaps even enhanced identification of prodromal "at risk" subjects.
Collapse
Affiliation(s)
- Robert Mazgaj
- Department of Psychiatry, New York University School of Medicine, 1 Park Avenue, 8th Floor, Rm 222, New York, NY, 10016, USA
- Creedmoor State Psychiatric Center, Queens Village, New York, NY, 11427, USA
| | - Assaf Tal
- Department of Radiology, New York University School of Medicine, 660 First Avenue, New York, NY, 10016, USA
| | - Raymond Goetz
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Mariana Lazar
- Department of Radiology, New York University School of Medicine, 660 First Avenue, New York, NY, 10016, USA
| | - Karen Rothman
- Department of Psychiatry, New York University School of Medicine, 1 Park Avenue, 8th Floor, Rm 222, New York, NY, 10016, USA
- Creedmoor State Psychiatric Center, Queens Village, New York, NY, 11427, USA
| | - Julie Walsh Messinger
- Department of Psychiatry, New York University School of Medicine, 1 Park Avenue, 8th Floor, Rm 222, New York, NY, 10016, USA
- Creedmoor State Psychiatric Center, Queens Village, New York, NY, 11427, USA
- Department of Psychiatry, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Dolores Malaspina
- Department of Psychiatry, New York University School of Medicine, 1 Park Avenue, 8th Floor, Rm 222, New York, NY, 10016, USA.
- Creedmoor State Psychiatric Center, Queens Village, New York, NY, 11427, USA.
| | - Oded Gonen
- Department of Radiology, New York University School of Medicine, 660 First Avenue, New York, NY, 10016, USA
| |
Collapse
|
31
|
Wu CK, Tseng PT, Chen YW, Tu KY, Lin PY. Significantly higher peripheral fibroblast growth factor-2 levels in patients with major depressive disorder: A preliminary meta-analysis under MOOSE guidelines. Medicine (Baltimore) 2016; 95:e4563. [PMID: 27537581 PMCID: PMC5370807 DOI: 10.1097/md.0000000000004563] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In vivo and in vitro studies demonstrate the important roles of fibroblast growth factor (FGF) and FGF receptors (FGFRs) in neural survival, neurogenesis, oxidative stress, and emotional behavior. However, evidence on the role of FGF and FGFR in the pathophysiology of major depressive disorder (MDD) remains limited and inconclusive. OBJECTIVES This preliminary meta-analysis aimed to examine changes in peripheral or central FGF and FGFR levels in patients with MDD. DATA SOURCES Electronic research through platform of PubMed and ClinicalTrials.gov. STUDY ELIGIBILITY CRITERIA, PARTICIPANTS, AND INTERVENTIONS We used the inclusion criteria: articles discussing the comparisons of FGF levels, either in peripheral or central environment, in patients with MDD and in healthy controls (HC); articles on clinical trials in humans; and case-control trials. Case reports or series and nonclinical trials were excluded. STUDY APPRAISAL AND SYNTHESIS METHODS Using a thorough literature search, the FGF/FGFR levels in patients with MDD and HC were compared. Four studies on peripheral FGF-2 and 3 on central FGF-2 and FGFR1 levels were included. RESULTS The findings reveal significantly higher peripheral FGF-2 protein and central FGFR1 RNA levels in patients with MDD than in HC (P = 0.005 and 0.006, separately), but no significant association with clinical variables. There was also no significant difference in the central FGF-2 levels in patients with MDD and in HC (P = 0.180). LIMITATION The study has limitations of a small number of included studies, lack of meta-analysis of the FGF changes along with treatment, and lack of direct evidence on correlation of peripheral FGF-2 with central FGF-2 levels. CONCLUSIONS AND IMPLICATIONS OF KEY FINDINGS This preliminary meta-analysis points out a new direction for future studies investigating the relationship among MDD, oxidative stress, and the FGF family.
Collapse
Affiliation(s)
- Ching-Kuan Wu
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | - Ping-Tao Tseng
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | | | - Kun-Yu Tu
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
- Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Correspondence: Pao-Yen Lin, Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, 123, Dapi Road, Niaosong, Kaohsiung 833, Taiwan (e-mail: )
| |
Collapse
|
32
|
Corley SM, Canales CP, Carmona-Mora P, Mendoza-Reinosa V, Beverdam A, Hardeman EC, Wilkins MR, Palmer SJ. RNA-Seq analysis of Gtf2ird1 knockout epidermal tissue provides potential insights into molecular mechanisms underpinning Williams-Beuren syndrome. BMC Genomics 2016; 17:450. [PMID: 27295951 PMCID: PMC4907016 DOI: 10.1186/s12864-016-2801-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/26/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Williams-Beuren Syndrome (WBS) is a genetic disorder associated with multisystemic abnormalities, including craniofacial dysmorphology and cognitive defects. It is caused by a hemizygous microdeletion involving up to 28 genes in chromosome 7q11.23. Genotype/phenotype analysis of atypical microdeletions implicates two evolutionary-related transcription factors, GTF2I and GTF2IRD1, as prime candidates for the cause of the facial dysmorphology. RESULTS Using a targeted Gtf2ird1 knockout mouse, we employed massively-parallel sequencing of mRNA (RNA-Seq) to understand changes in the transcriptional landscape associated with inactivation of Gtf2ird1 in lip tissue. We found widespread dysregulation of genes including differential expression of 78 transcription factors or coactivators, several involved in organ development including Hey1, Myf6, Myog, Dlx2, Gli1, Gli2, Lhx2, Pou3f3, Sox2, Foxp3. We also found that the absence of GTF2IRD1 is associated with increased expression of genes involved in cellular proliferation, including growth factors consistent with the observed phenotype of extreme thickening of the epidermis. At the same time, there was a decrease in the expression of genes involved in other signalling mechanisms, including the Wnt pathway, indicating dysregulation in the complex networks necessary for epidermal differentiation and facial skin patterning. Several of the differentially expressed genes have known roles in both tissue development and neurological function, such as the transcription factor Lhx2 which regulates several genes involved in both skin and brain development. CONCLUSIONS Gtf2ird1 inactivation results in widespread gene dysregulation, some of which may be due to the secondary consequences of gene regulatory network disruptions involving several transcription factors and signalling molecules. Genes involved in growth factor signalling and cell cycle progression were identified as particularly important for explaining the skin dysmorphology observed in this mouse model. We have noted that a number of the dysregulated genes have known roles in brain development as well as epidermal differentiation and maintenance. Therefore, this study provides clues as to the underlying mechanisms that may be involved in the broader profile of WBS.
Collapse
Affiliation(s)
- Susan M Corley
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW Australia, Sydney, NSW, Australia.
| | - Cesar P Canales
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Paulina Carmona-Mora
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | | | | | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Stephen J Palmer
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| |
Collapse
|
33
|
Turner CA, Eren-Koçak E, Inui EG, Watson SJ, Akil H. Dysregulated fibroblast growth factor (FGF) signaling in neurological and psychiatric disorders. Semin Cell Dev Biol 2016; 53:136-43. [PMID: 26454097 PMCID: PMC4833700 DOI: 10.1016/j.semcdb.2015.10.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/05/2015] [Indexed: 12/27/2022]
Abstract
The role of the fibroblast growth factor (FGF) system in brain-related disorders has received considerable attention in recent years. To understand the role of this system in neurological and psychiatric disorders, it is important to identify the specific members of the FGF family that are implicated, their location and the various mechanisms they can be modulated. Each disorder appears to impact specific molecular players in unique anatomical locations, and all of these could conceivably become targets for treatment. In the last several years, the issue of how to target this system directly has become an area of increasing interest. To date, the most promising therapeutics are small molecule inhibitors and antibodies that modulate FGF receptor (FGFR) function. Beyond attempting to modify the primary players affected by a given brain disorder, it may prove useful to target molecules, such as membrane-bound or extracellular proteins that interact with FGF ligands or FGFRs to modulate signaling.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | | | - Stanley J Watson
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Huda Akil
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Quach TT, Lerch JK, Honnorat J, Khanna R, Duchemin AM. Neuronal networks in mental diseases and neuropathic pain: Beyond brain derived neurotrophic factor and collapsin response mediator proteins. World J Psychiatry 2016; 6:18-30. [PMID: 27014595 PMCID: PMC4804265 DOI: 10.5498/wjp.v6.i1.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/24/2015] [Accepted: 01/07/2016] [Indexed: 02/05/2023] Open
Abstract
The brain is a complex network system that has the capacity to support emotion, thought, action, learning and memory, and is characterized by constant activity, constant structural remodeling, and constant attempt to compensate for this remodeling. The basic insight that emerges from complex network organization is that substantively different networks can share common key organizational principles. Moreover, the interdependence of network organization and behavior has been successfully demonstrated for several specific tasks. From this viewpoint, increasing experimental/clinical observations suggest that mental disorders are neural network disorders. On one hand, single psychiatric disorders arise from multiple, multifactorial molecular and cellular structural/functional alterations spreading throughout local/global circuits leading to multifaceted and heterogeneous clinical symptoms. On the other hand, various mental diseases may share functional deficits across the same neural circuit as reflected in the overlap of symptoms throughout clinical diagnoses. An integrated framework including experimental measures and clinical observations will be necessary to formulate a coherent and comprehensive understanding of how neural connectivity mediates and constraints the phenotypic expression of psychiatric disorders.
Collapse
|
35
|
Kálmán S, Garbett KA, Janka Z, Mirnics K. Human dermal fibroblasts in psychiatry research. Neuroscience 2016; 320:105-21. [PMID: 26855193 DOI: 10.1016/j.neuroscience.2016.01.067] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
In order to decipher the disease etiology, progression and treatment of multifactorial human brain diseases we utilize a host of different experimental models. Recently, patient-derived human dermal fibroblast (HDF) cultures have re-emerged as promising in vitro functional system for examining various cellular, molecular, metabolic and (patho)physiological states and traits of psychiatric disorders. HDF studies serve as a powerful complement to postmortem and animal studies, and often appear to be informative about the altered homeostasis in neural tissue. Studies of HDFs from patients with schizophrenia (SZ), depression, bipolar disorder (BD), autism, attention deficit and hyperactivity disorder and other psychiatric disorders have significantly advanced our understanding of these devastating diseases. These reports unequivocally prove that signal transduction, redox homeostasis, circadian rhythms and gene*environment (G*E) interactions are all amenable for assessment by the HDF model. Furthermore, the reported findings suggest that this underutilized patient biomaterial, combined with modern molecular biology techniques, may have both diagnostic and prognostic value, including prediction of response to therapeutic agents.
Collapse
Affiliation(s)
- S Kálmán
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary.
| | - K A Garbett
- Department of Psychiatry, Vanderbilt University, 8128 MRB III, 465 21st Avenue, Nashville, TN 37232, USA.
| | - Z Janka
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary.
| | - K Mirnics
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary; Department of Psychiatry, Vanderbilt University, 8128 MRB III, 465 21st Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
36
|
Increased innervation of forebrain targets by midbrain dopaminergic neurons in the absence of FGF-2. Neuroscience 2016; 314:134-44. [DOI: 10.1016/j.neuroscience.2015.11.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/21/2022]
|
37
|
Alshammari MA, Alshammari TK, Nenov MN, Scala F, Laezza F. Fibroblast Growth Factor 14 Modulates the Neurogenesis of Granule Neurons in the Adult Dentate Gyrus. Mol Neurobiol 2015; 53:7254-7270. [PMID: 26687232 DOI: 10.1007/s12035-015-9568-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/29/2015] [Indexed: 11/25/2022]
Abstract
Adult neurogenesis, the production of mature neurons from progenitor cells in the adult mammalian brain, is linked to the etiology of neurodegenerative and psychiatric disorders. However, a thorough understanding of the molecular elements at the base of adult neurogenesis remains elusive. Here, we provide evidence for a previously undescribed function of fibroblast growth factor 14 (FGF14), a brain disease-associated factor that controls neuronal excitability and synaptic plasticity, in regulating adult neurogenesis in the dentate gyrus (DG). We found that FGF14 is dynamically expressed in restricted subtypes of sex determining region Y-box 2 (Sox2)-positive and doublecortin (DCX)-positive neural progenitors in the DG. Bromodeoxyuridine (BrdU) incorporation studies and confocal imaging revealed that genetic deletion of Fgf14 in Fgf14 -/- mice leads to a significant change in the proportion of proliferating and immature and mature newly born adult granule cells. This results in an increase in the late immature and early mature population of DCX and calretinin (CR)-positive neurons. Electrophysiological extracellular field recordings showed reduced minimal threshold response and impaired paired-pulse facilitation at the perforant path to DG inputs in Fgf14 -/- compared to Fgf14 +/+ mice, supporting disrupted synaptic connectivity as a correlative read-out to impaired neurogenesis. These new insights into the biology of FGF14 in neurogenesis shed light into the signaling pathways associated with disrupted functions in complex brain diseases.
Collapse
Affiliation(s)
- Musaad A Alshammari
- Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX, USA
- Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Tahani K Alshammari
- Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX, USA
- Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Federico Scala
- Biophysics Graduate Program, Institute of Human Physiology, Università Cattolica, Rome, Italy
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Fernanda Laezza
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA.
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA.
- Center for Biomedical Engineering, The University of Texas Medical Branch, Galveston, TX, USA.
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA.
| |
Collapse
|
38
|
Molecular underpinnings of prefrontal cortex development in rodents provide insights into the etiology of neurodevelopmental disorders. Mol Psychiatry 2015; 20:795-809. [PMID: 25450230 PMCID: PMC4486649 DOI: 10.1038/mp.2014.147] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/12/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022]
Abstract
The prefrontal cortex (PFC), seat of the highest-order cognitive functions, constitutes a conglomerate of highly specialized brain areas and has been implicated to have a role in the onset and installation of various neurodevelopmental disorders. The development of a properly functioning PFC is directed by transcription factors, guidance cues and other regulatory molecules and requires the intricate and temporal orchestration of a number of developmental processes. Disturbance or failure of any of these processes causing neurodevelopmental abnormalities within the PFC may contribute to several of the cognitive deficits seen in patients with neurodevelopmental disorders. In this review, we elaborate on the specific processes underlying prefrontal development, such as induction and patterning of the prefrontal area, proliferation, migration and axonal guidance of medial prefrontal progenitors, and their eventual efferent and afferent connections. We furthermore integrate for the first time the available knowledge from genome-wide studies that have revealed genes linked to neurodevelopmental disorders with experimental molecular evidence in rodents. The integrated data suggest that the pathogenic variants in the neurodevelopmental disorder-associated genes induce prefrontal cytoarchitectonical impairments. This enhances our understanding of the molecular mechanisms of prefrontal (mis)development underlying the four major neurodevelopmental disorders in humans, that is, intellectual disability, autism spectrum disorders, attention deficit hyperactivity disorder and schizophrenia, and may thus provide clues for the development of novel therapies.
Collapse
|
39
|
Veniaminova EA, Zubareva OE. The changes in exploratory behavior and Fgf2 gene expression in cells of the rat brain after the early postnatal administration of bacterial lipopolysaccharide. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415020105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Lee J, Jo DG, Park D, Chung HY, Mattson MP. Adaptive cellular stress pathways as therapeutic targets of dietary phytochemicals: focus on the nervous system. Pharmacol Rev 2015; 66:815-68. [PMID: 24958636 DOI: 10.1124/pr.113.007757] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During the past 5 decades, it has been widely promulgated that the chemicals in plants that are good for health act as direct scavengers of free radicals. Here we review evidence that favors a different hypothesis for the health benefits of plant consumption, namely, that some phytochemicals exert disease-preventive and therapeutic actions by engaging one or more adaptive cellular response pathways in cells. The evolutionary basis for the latter mechanism is grounded in the fact that plants produce natural antifeedant/noxious chemicals that discourage insects and other organisms from eating them. However, in the amounts typically consumed by humans, the phytochemicals activate one or more conserved adaptive cellular stress response pathways and thereby enhance the ability of cells to resist injury and disease. Examplesof such pathways include those involving the transcription factors nuclear factor erythroid 2-related factor 2, nuclear factor-κB, hypoxia-inducible factor 1α, peroxisome proliferator-activated receptor γ, and forkhead box subgroup O, as well as the production and action of trophic factors and hormones. Translational research to develop interventions that target these pathways may lead to new classes of therapeutic agents that act by stimulating adaptive stress response pathways to bolster endogenous defenses against tissue injury and disease. Because neurons are particularly sensitive to potentially noxious phytochemicals, we focus on the nervous system but also include findings from other cell types in which actions of phytochemicals on specific signal transduction pathways have been more thoroughly studied.
Collapse
Affiliation(s)
- Jaewon Lee
- Department of Pharmacy, College of Pharmacy, and Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Geumjeong-gu, Busan, Republic of Korea (J.L., D.P., H.Y.C.); School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea (D.-G.J.); Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland (M.P.M.); and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (M.P.M.)
| | - Dong-Gyu Jo
- Department of Pharmacy, College of Pharmacy, and Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Geumjeong-gu, Busan, Republic of Korea (J.L., D.P., H.Y.C.); School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea (D.-G.J.); Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland (M.P.M.); and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (M.P.M.)
| | - Daeui Park
- Department of Pharmacy, College of Pharmacy, and Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Geumjeong-gu, Busan, Republic of Korea (J.L., D.P., H.Y.C.); School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea (D.-G.J.); Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland (M.P.M.); and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (M.P.M.)
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, and Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Geumjeong-gu, Busan, Republic of Korea (J.L., D.P., H.Y.C.); School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea (D.-G.J.); Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland (M.P.M.); and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (M.P.M.)
| | - Mark P Mattson
- Department of Pharmacy, College of Pharmacy, and Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Geumjeong-gu, Busan, Republic of Korea (J.L., D.P., H.Y.C.); School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea (D.-G.J.); Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland (M.P.M.); and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (M.P.M.)
| |
Collapse
|
41
|
Roalf DR, Vandekar SN, Almasy L, Ruparel K, Satterthwaite TD, Elliott MA, Podell J, Gallagher S, Jackson CT, Prasad K, Wood J, Pogue-Geile MF, Nimgaonkar VL, Gur RC, Gur RE. Heritability of subcortical and limbic brain volume and shape in multiplex-multigenerational families with schizophrenia. Biol Psychiatry 2015; 77:137-46. [PMID: 24976379 PMCID: PMC4247350 DOI: 10.1016/j.biopsych.2014.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 04/25/2014] [Accepted: 05/21/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Brain abnormalities of subcortical and limbic nuclei are common in patients with schizophrenia, and variation in these structures is considered a putative endophenotype for the disorder. Multiplex-multigenerational families with schizophrenia provide an opportunity to investigate the impact of shared genetic ancestry, but these families have not been previously examined to study structural brain abnormalities. We estimate the heritability of subcortical and hippocampal brain volumes in multiplex-multigenerational families and the heritability of subregions using advanced shape analysis. METHODS The study comprised 439 participants from two sites who underwent 3T structural magnetic resonance imaging. The participants included 190 European-Americans from 32 multiplex-multigenerational families with schizophrenia and 249 healthy comparison subjects. Subcortical and hippocampal volume and shape were measured in 14 brain structures. Heritability was estimated for volume and shape. RESULTS Volume and shape were heritable in families. Estimates of heritability in subcortical and limbic volumes ranged from .45 in the right hippocampus to .84 in the left putamen. The shape of these structures was heritable (range, .40-.49), and specific subregional shape estimates of heritability tended to exceed heritability estimates of volume alone. CONCLUSIONS These results demonstrate that volume and shape of subcortical and limbic brain structures are potential endophenotypic markers in schizophrenia. The specificity obtained using shape analysis may improve selection of imaging phenotypes that better reflect the underlying neurobiology. Our findings can aid in the identification of specific genetic targets that affect brain structure and function in schizophrenia.
Collapse
|
42
|
Merico D, Costain G, Butcher NJ, Warnica W, Ogura L, Alfred SE, Brzustowicz LM, Bassett AS. MicroRNA Dysregulation, Gene Networks, and Risk for Schizophrenia in 22q11.2 Deletion Syndrome. Front Neurol 2014; 5:238. [PMID: 25484875 PMCID: PMC4240070 DOI: 10.3389/fneur.2014.00238] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/02/2014] [Indexed: 01/20/2023] Open
Abstract
The role of microRNAs (miRNAs) in the etiology of schizophrenia is increasingly recognized. Microdeletions at chromosome 22q11.2 are recurrent structural variants that impart a high risk for schizophrenia and are found in up to 1% of all patients with schizophrenia. The 22q11.2 deletion region overlaps gene DGCR8, encoding a subunit of the miRNA microprocessor complex. We identified miRNAs overlapped by the 22q11.2 microdeletion and for the first time investigated their predicted target genes, and those implicated by DGCR8, to identify targets that may be involved in the risk for schizophrenia. The 22q11.2 region encompasses seven validated or putative miRNA genes. Employing two standard prediction tools, we generated sets of predicted target genes. Functional enrichment profiles of the 22q11.2 region miRNA target genes suggested a role in neuronal processes and broader developmental pathways. We then constructed a protein interaction network of schizophrenia candidate genes and interaction partners relevant to brain function, independent of the 22q11.2 region miRNA mechanisms. We found that the predicted gene targets of the 22q11.2 deletion miRNAs, and targets of the genome-wide miRNAs predicted to be dysregulated by DGCR8 hemizygosity, were significantly represented in this schizophrenia network. The findings provide new insights into the pathway from 22q11.2 deletion to expression of schizophrenia, and suggest that hemizygosity of the 22q11.2 region may have downstream effects implicating genes elsewhere in the genome that are relevant to the general schizophrenia population. These data also provide further support for the notion that robust genetic findings in schizophrenia may converge on a reasonable number of final pathways.
Collapse
Affiliation(s)
- Daniele Merico
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children , Toronto, ON , Canada
| | - Gregory Costain
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Nancy J Butcher
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada ; Institute of Medical Science, University of Toronto , Toronto, ON , Canada
| | - William Warnica
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Lucas Ogura
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Simon E Alfred
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Linda M Brzustowicz
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University , Piscataway, NJ , USA
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health , Toronto, ON , Canada ; Institute of Medical Science, University of Toronto , Toronto, ON , Canada ; The Dalglish Family Hearts and Minds Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network , Toronto, ON , Canada ; Department of Psychiatry, Toronto General Research Institute, University Health Network , Toronto, ON , Canada ; Department of Psychiatry, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
43
|
Scherf JM, Hu XS, Tepp WH, Ichtchenko K, Johnson EA, Pellett S. Analysis of gene expression in induced pluripotent stem cell-derived human neurons exposed to botulinum neurotoxin A subtype 1 and a type A atoxic derivative. PLoS One 2014; 9:e111238. [PMID: 25337697 PMCID: PMC4206481 DOI: 10.1371/journal.pone.0111238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/19/2014] [Indexed: 11/18/2022] Open
Abstract
Botulinum neurotoxin type A1 (BoNT/A1) is a potent protein toxin responsible for the potentially fatal human illness botulism. Notwithstanding, the long-lasting flaccid muscle paralysis caused by BoNT/A has led to its utility as a powerful and versatile bio-pharmaceutical. The flaccid paralysis is due to specific cleavage of neuronal SNAREs by BoNTs. However, actions of BoNTs on intoxicated neurons besides the cleavage of SNAREs have not been studied in detail. In this study we investigated by microarray analysis the effects of BoNT/A and a catalytically inactive derivative (BoNT/A ad) on the transcriptome of human induced pluripotent stem cell (hiPSC)-derived neurons at 2 days and 2 weeks after exposure. While there were only minor changes in expression levels at 2 days post exposure, at 2 weeks post exposure 492 genes were differentially expressed more than 2-fold in BoNT/A1-exposed cells when compared to non-exposed populations, and 682 genes were differentially expressed in BoNT/A ad-exposed cells. The vast majority of genes were similarly regulated in BoNT/A1 and BoNT/A ad-exposed neurons, and the few genes differentially regulated between BoNT/A1 and BoNT/A ad-exposed neurons were differentially expressed less than 3.5 fold. These data indicate a similar response of neurons to BoNT/A1 and BoNT/A ad exposure. The most highly regulated genes in cells exposed to either BoNT/A1 or BoNT/A ad are involved in neurite outgrowth and calcium channel sensitization.
Collapse
Affiliation(s)
- Jacob M. Scherf
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiaoyang Serene Hu
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - William H. Tepp
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Konstantin Ichtchenko
- Department of Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Eric A. Johnson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
44
|
α2-Adrenoceptors are targets for antipsychotic drugs. Psychopharmacology (Berl) 2014; 231:801-12. [PMID: 24488407 DOI: 10.1007/s00213-014-3459-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/11/2014] [Indexed: 01/29/2023]
Abstract
RATIONALE Almost all antipsychotic drugs (APDs), irrespective of whether they belong to the first-generation (e.g. haloperidol) or second-generation (e.g. clozapine), are dopamine D2 receptor antagonists. Second-generation APDs, which differ from first-generation APDs in possessing a lower propensity to induce extrapyramidal side effects, target a variety of monoamine receptors such as serotonin (5-hydroxytryptamine) receptors (e.g. 5-HT1A, 5-HT2A, 5-HT2C, 5-HT6, 5-HT7) and α1- and α2-adrenoceptors in addition to their antagonist effects at D2 receptors. OBJECTIVE This short review is focussed on the potential role of α2-adrenoceptors in the antipsychotic therapy. RESULTS Schizophrenia is characterised by three categories of symptoms: positive symptoms, negative symptoms and cognitive deficits. α2-Adrenoceptors are classified into three distinct subtypes in mammals, α2A, α2B and α2C. Whereas the α2B-adrenoceptor seems to play only a minor role in the brain, activation of postsynaptic α2A-adrenoceptors in the prefrontal cortex improves cognitive functions. Preclinical models such as D-amphetamine-induced locomotion, the conditioned avoidance response and the pharmacological N-methyl-D-aspartate receptor hypofunction model have shown that α2C-adrenoceptor blockade or the combination of D2 receptor antagonists with idazoxan (α2A/2C-adrenoceptor antagonist) could be useful in schizophrenia. A potential benefit of a treatment combination of first-generation APDs with the α2A/2C-adrenoceptor antagonists idazoxan or mirtazapine was also demonstrated in patients with schizophrenia. CONCLUSIONS It is concluded that α2-adrenoceptors may be promising targets in the antipsychotic therapy.
Collapse
|