1
|
Hawkey AB, Shekey N, Dean C, Asrat H, Koburov R, Holloway ZR, Kullman SW, Levin ED. Developmental exposure to pesticides that disrupt retinoic acid signaling causes persistent retinoid and behavioral dysfunction in zebrafish. Toxicol Sci 2024; 198:246-259. [PMID: 38237923 DOI: 10.1093/toxsci/kfae001] [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: 03/27/2024] Open
Abstract
Early developmental exposure to environmental toxicants may play a role in the risk for developing autism. A variety of pesticides have direct effects on retinoic acid (RA) signaling and as RA signaling has important roles in neurodevelopment, such compounds may cause developmental neurotoxicity through an overlapping adverse outcome pathway. It is hypothesized that a pesticide's embryonic effects on retinoid function may correspond with neurobehavioral disruption later in development. In the current studies, we determined the effects of RA-acting pesticides on neurobehavioral development in zebrafish. Buprofezin and imazalil caused generalized hypoactivity in the larval motility test, whereas chlorothalonil and endosulfan I led to selective hypoactivity and hyperactivity, respectively. With buprofezin, chlorothalonil, and imazalil, hypoactivity and/or novel anxiety-like behaviors persisted in adulthood and buprofezin additionally decreased social attraction responses in adulthood. Endosulfan I did not produce significant adult behavioral effects. Using qPCR analyses of adult brain tissue, we observed treatment-induced alterations in RA synthesis or catabolic genes, indicating persistent changes in RA homeostasis. These changes were compound-specific, with respect to expression directionality, and potential patterns of homeostatic disruption. Results suggest the likely persistence of disruptions in RA signaling well into adulthood and may represent compensatory mechanisms following early life stage exposures. This study demonstrates that early developmental exposure to environmental toxicants that interfere with RA signaling causes short as well as long-term behavioral disruption in a well-established zebrafish behavioral model and expand upon the meaning of the RA adverse outcome pathway, indicating that observed effects likely correspond with the nature of underlying homeostatic effects.
Collapse
Affiliation(s)
- Andrew B Hawkey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Biomedical Sciences, Midwestern University, Downers Grove, Illinois 60515, USA
| | - Nathan Shekey
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27606, USA
| | - Cassandra Dean
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Helina Asrat
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Reese Koburov
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Zade R Holloway
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Seth W Kullman
- Toxicology Program, North Carolina State University, Raleigh, North Carolina 27606, USA
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
2
|
Wang X, Ma T, Wei C, Liu J, Yu T, Zou Y, Liu S, Yang Z, Xi J. Toxic effects of exogenous retinoic acid on the neurodevelopment of zebrafish (Danio rerio) embryos. Neurotoxicol Teratol 2023; 100:107291. [PMID: 37689270 DOI: 10.1016/j.ntt.2023.107291] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Endogenous retinoic acid (RA) is essential for embryonic development and maintaining adult physiological processes. Human-caused RA residues in the environment threaten the survival of organisms in the environment. We employed zebrafish as a model to explore the developmental impacts of excess RA. We used exogenous RA to raise the amount of RA signal in the embryos and looked at the effects of excess RA on embryonic morphological development. Upregulation of the RA signal significantly reduced embryo hatching and increased embryo malformation. To further understand the neurotoxic impact of RA signaling on early neurodevelopment, we measured the expression of neurodevelopmental marker genes and cell death and proliferation markers in zebrafish embryos. Exogenous RA disrupted stem cell (SC) and neuron marker gene expression and exacerbated apoptosis in the embryos. Furthermore, we looked into the links between the transcriptional coactivator RBM14 and RA signaling to better understand the mechanism of RA neurotoxicity. There was a negative interaction between RA signaling and the transcription coactivator RBM14, and the morpholino-induced RBM14 down-regulation can partially block the effects of RAR antagonist BMS493-induced RA signaling inhibition on embryonic malformation and cell apoptosis. In conclusion, exogenous RA causes neurodevelopmental toxicity, and RBM14 may be involved in this neurotoxic process.
Collapse
Affiliation(s)
- Xiaoxuan Wang
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Ting Ma
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Cizhao Wei
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, Hubei, China
| | - Juan Liu
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Ting Yu
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Zou
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Song Liu
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zheqiong Yang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, Hubei, China.
| | - Jinlei Xi
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Bremner JD. Isotretinoin and neuropsychiatric side effects: Continued vigilance is needed. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2021; 6:100230. [PMID: 37168254 PMCID: PMC10168661 DOI: 10.1016/j.jadr.2021.100230] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Isotretinoin (13-cis-retinoic acid, marketed under the names Accutane, Roaccutane, and others) is an effective treatment for acne that has been on the market for over 30 years, although reports of neuropsychiatric side effects continue to be reported. Isotretinoin is an isomer of the active form of Vitamin A, 13-trans-retinoic acid, which has known psychiatric side effects when given in excessive doses, and is part of the family of compounds called retinoids, which have multiple functions in the central nervous system. Methods The literature was reviewed in pubmed and psychinfo for research related to isotretinoin and neuropsychiatric side effects including depression, suicidal thoughts, suicide, mania, anxiety, impulsivity, emotional lability, violence, aggression, and psychosis. Results Multiple case series have shown that successful treatment of acne with isotretinoin results in improvements in measures of quality of life and self esteem However, studies show individual cases of clinically significant depression and other neuropsychiatric events that, although not common, are persistent in the literature. Since the original cases of depression were reported to the United States Food and Drug Administration, numerous cases have been reported to regulatory agencies in the United Kingdom, France, Ireland, Denmark, Australia, Canada, and other countries, making isotretinoin one of the top five medications in the world associated with depression and other neuropsychiatric side effects. Clinicians are advised to warn patients of the risks of neuropsychiatric side effects with isotretinoin which may arise from the medication itself, and not just as a side effect of acne or youth.
Collapse
Affiliation(s)
- J Douglas Bremner
- Department of Psychiatry & Behavioral Sciences, and Department of Radiology and Imaging Sciences, Emory University School of Medicine, VA Medical Center, Decatur, GA, United States
| |
Collapse
|
4
|
Hudson KM, Shiver E, Yu J, Mehta S, Jima DD, Kane MA, Patisaul HB, Cowley M. Transcriptomic, proteomic, and metabolomic analyses identify candidate pathways linking maternal cadmium exposure to altered neurodevelopment and behavior. Sci Rep 2021; 11:16302. [PMID: 34381081 PMCID: PMC8357970 DOI: 10.1038/s41598-021-95630-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 07/28/2021] [Indexed: 02/07/2023] Open
Abstract
Cadmium (Cd) is a ubiquitous toxic heavy metal of major public concern. Despite inefficient placental transfer, maternal Cd exposure impairs fetal growth and development. Increasing evidence from animal models and humans suggests maternal Cd exposure negatively impacts neurodevelopment; however, the underlying molecular mechanisms are unclear. To address this, we utilized multiple -omics approaches in a mouse model of maternal Cd exposure to identify pathways altered in the developing brain. Offspring maternally exposed to Cd presented with enlarged brains proportional to body weights at birth and altered behavior at adulthood. RNA-seq in newborn brains identified exposure-associated increases in Hox gene and myelin marker expression and suggested perturbed retinoic acid (RA) signaling. Proteomic analysis showed altered levels of proteins involved in cellular energy pathways, hypoxic response, and RA signaling. Consistent with transcriptomic and proteomic analyses, we identified increased levels of retinoids in maternally-exposed newborn brains. Metabolomic analyses identified metabolites with significantly altered abundance, supportive of changes to cellular energy pathways and hypoxia. Finally, maternal Cd exposure reduced mitochondrial DNA levels in newborn brains. The identification of multiple pathways perturbed in the developing brain provides a basis for future studies determining the mechanistic links between maternal Cd exposure and altered neurodevelopment and behavior.
Collapse
Affiliation(s)
- Kathleen M Hudson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Emily Shiver
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Jianshi Yu
- School of Pharmacy Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Sanya Mehta
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Dereje D Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA
| | - Maureen A Kane
- School of Pharmacy Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Michael Cowley
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA.
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
5
|
Duque-Díaz E, Coveñas R. Mapping of folic acid in the children brainstem. Anat Cell Biol 2021; 54:340-349. [PMID: 33967031 PMCID: PMC8493014 DOI: 10.5115/acb.21.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 11/27/2022] Open
Abstract
Using highly specific antisera, the neuroanatomical distribution of folic acid (FA) and retinoic acid (RA) has been studied for the first time in the children brainstem. Neither immunoreactive structures containing RA nor immunoreactive fibers containing FA were found. FA-immunoreactive perikarya (fusiform, small/medium in size, one short dendrite) were only found in the pons in three regions: central gray, reticular formation, and locus coeruleus. The number of cell bodies decreased with age. In the first case studied (2 years), a moderate density of cell bodies was observed in the central gray and reticular formation, whereas a low density was found in the locus coeruleus. In the second case (6 years), a low density of these perikarya was observed in the central gray, reticular formation, and locus coeruleus. In the third case (7 years), a low density of FA-immunoreactive cell bodies was found in the central gray and reticular formation, whereas in the locus coeruleus no immunoreactive cell bodies were observed. The distribution of FA in the central nervous system of humans and monkeys is different and, in addition, in these species the vitamin was located in different parts of the nerve cells. The restricted distribution of FA suggests that the vitamin is involved in specific physiological mechanisms.
Collapse
Affiliation(s)
- Ewing Duque-Díaz
- Laboratory of Neurosciences, School of Medicine, Universidad de Santander, Bucaramanga, Colombia
| | - Rafael Coveñas
- Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, Salamanca, Spain.,Grupo GIR BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
| |
Collapse
|
6
|
Liu H, Tan M, Cheng B, Wang S, Xiao L, Zhu J, Wu Q, Lai X, Zhang Q, Chen J, Li T. Valproic Acid Induces Autism-Like Synaptic and Behavioral Deficits by Disrupting Histone Acetylation of Prefrontal Cortex ALDH1A1 in Rats. Front Neurosci 2021; 15:641284. [PMID: 33994921 PMCID: PMC8113628 DOI: 10.3389/fnins.2021.641284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/06/2021] [Indexed: 12/27/2022] Open
Abstract
Objectives This study aimed to investigate the impact of valproic acid (VPA) on the histone acetylation of acetaldehyde dehydrogenase 1A1 (ALDH1A1) and the mechanism underlying VPA-induced autism-like behavior. Methods Female Sprague-Dawley rats were intraperitoneally injected with VPA during gestation to establish an autism model in their offspring. Some offspring prenatally exposed to VPA were randomly treated with MS-275, one histone deacetylase (HDAC) inhibitor, or retinoic acid (RA) after birth. Behavioral tests were conducted on the offspring 6 weeks after birth. Electrophysiological experiments were performed to investigate long-term potentiation (LTP) in the prefrontal cortex (PFC). The expression levels of AMPA receptors (GluA1 and 2), NMDA receptors (GluN1 and 2), synapsin 1 (SYN1), HDAC, acetylated histone 3 (AcH3), RA receptor alpha (RARα), and ALDH1A1 in the PFC were measured by Western blotting and quantitative polymerase chain reaction. ALDH enzyme activity in PFC tissue was detected using a Micro ALDH Assay Kit. The RA level in the PFC was measured using ultrahigh-performance liquid chromatography/tandem mass spectrometry. A chromatin immunoprecipitation (ChIP) experiment explored the interaction between the ALDH1A1 gene and AcH3. Results Offspring prenatally exposed to VPA showed autism-like behavior, upregulated the levels of LTP and GluN2A, GluA1, and SYN1 proteins relevant to synaptic plasticity in the PFC. The expression levels of HDAC3 mRNA and protein were increased. On the other hand, there was a significant reduction in the levels of AcH3, RARα, RA, ALDH1A1 mRNA and protein, the level of ALDH activity and AcH3 enrichment in the ALDH1A1 promoter region in VPA-induced offspring. Administration of MS-275 in VPA offspring significantly elevated the levels of AcH3, ALDH1A1 mRNA and protein, ALDH activity, RA, the level of RARα protein and the binding of AcH3 to the ALDH1A1 promoter. In addition, the GluA1 protein level and LTP were reduced, and most behavioral deficits were reversed. After RA supplementation in the VPA-treated offspring, the RA and RARα protein levels were significantly upregulated, GluA1 protein and LTP were downregulated, and most autism-like behavioral deficits were effectively reversed. Conclusion These findings suggest that VPA impairs histoneacetylation of ALDH1A1 and downregulates the RA-RARα pathway. Such epigenetic modification of ALDH1A1 by VPA leads to autism-like synaptic and behavioral deficits.
Collapse
Affiliation(s)
- Huan Liu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Mei Tan
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Boli Cheng
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Si Wang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Lu Xiao
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Jiang Zhu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Qionghui Wu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Xi Lai
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Qian Zhang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, Chongqing, China
| |
Collapse
|
7
|
Lenz M, Kruse P, Eichler A, Straehle J, Beck J, Deller T, Vlachos A. All-trans retinoic acid induces synaptic plasticity in human cortical neurons. eLife 2021; 10:e63026. [PMID: 33781382 PMCID: PMC8009674 DOI: 10.7554/elife.63026] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
A defining feature of the brain is the ability of its synaptic contacts to adapt structurally and functionally in an experience-dependent manner. In the human cortex, however, direct experimental evidence for coordinated structural and functional synaptic adaptation is currently lacking. Here, we probed synaptic plasticity in human cortical slices using the vitamin A derivative all-trans retinoic acid (atRA), a putative treatment for neuropsychiatric disorders such as Alzheimer's disease. Our experiments demonstrated that the excitatory synapses of superficial (layer 2/3) pyramidal neurons underwent coordinated structural and functional changes in the presence of atRA. These synaptic adaptations were accompanied by ultrastructural remodeling of the calcium-storing spine apparatus organelle and required mRNA translation. It was not observed in synaptopodin-deficient mice, which lack spine apparatus organelles. We conclude that atRA is a potent mediator of synaptic plasticity in the adult human cortex.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
| | - Jakob Straehle
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University FrankfurtFreiburg im BreisgauGermany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburg im BreisgauGermany
- Center Brain Links Brain Tools, University of FreiburgFreiburg im BreisgauGermany
| |
Collapse
|
8
|
Retinoic acid and depressive disorders: Evidence and possible neurobiological mechanisms. Neurosci Biobehav Rev 2020; 112:376-391. [DOI: 10.1016/j.neubiorev.2020.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
|
9
|
Reay WR, Cairns MJ. The role of the retinoids in schizophrenia: genomic and clinical perspectives. Mol Psychiatry 2020; 25:706-718. [PMID: 31666680 PMCID: PMC7156347 DOI: 10.1038/s41380-019-0566-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/23/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
Abstract
Signalling by retinoid compounds is vital for embryonic development, with particular importance for neurogenesis in the human brain. Retinoids, metabolites of vitamin A, exert influence over the expression of thousands of transcripts genome wide, and thus, act as master regulators of many important biological processes. A significant body of evidence in the literature now supports dysregulation of the retinoid system as being involved in the aetiology of schizophrenia. This includes mechanistic insights from large-scale genomic, transcriptomic and, proteomic studies, which implicate disruption of disparate aspects of retinoid biology such as transport, metabolism, and signalling. As a result, retinoids may present a valuable clinical opportunity in schizophrenia via novel pharmacotherapies and dietary intervention. Further work, however, is required to expand on the largely observational data collected thus far and confirm causality. This review will highlight the fundamentals of retinoid biology and examine the evidence for retinoid dysregulation in schizophrenia.
Collapse
Affiliation(s)
- William R. Reay
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| | - Murray J. Cairns
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| |
Collapse
|
10
|
Xu G, Wu F, Gu X, Zhang J, You K, Chen Y, Getachew A, Zhuang Y, Zhong X, Lin Z, Guo D, Yang F, Pan T, Wei H, Li YX. Direct Conversion of Human Urine Cells to Neurons by Small Molecules. Sci Rep 2019; 9:16707. [PMID: 31723223 PMCID: PMC6854089 DOI: 10.1038/s41598-019-53007-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/25/2019] [Indexed: 02/07/2023] Open
Abstract
Transdifferentiation of other cell type into human neuronal cells (hNCs) provides a platform for neural disease modeling, drug screening and potential cell-based therapies. Among all of the cell donor sources, human urine cells (hUCs) are convenient to obtain without invasive harvest procedure. Here, we report a novel approach for the transdifferentiation of hUCs into hNCs. Our study demonstrated that a combination of seven small molecules (CAYTFVB) cocktail induced transdifferentiation of hUCs into hNCs. These chemical-induced neuronal cells (CiNCs) exhibited typical neuron-like morphology and expressed mature neuronal markers. The neuronal-like morphology revealed in day 1, and the Tuj1-positive CiNCs reached to about 58% in day 5 and 38.36% Tuj1+/MAP2+ double positive cells in day 12. Partial electrophysiological properties of CiNCs was obtained using patch clamp. Most of the CiNCs generated using our protocol were glutamatergic neuron populations, whereas motor neurons, GABAergic or dopaminergic neurons were merely detected. hUCs derived from different donors were converted into CiNCs in this work. This method may provide a feasible and noninvasive approach for reprogramming hNCs from hUCs for disease models and drug screening.
Collapse
Affiliation(s)
- Guosheng Xu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangzhou Blood Center, Guangzhou, China
| | - Feima Wu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaotong Gu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jiaye Zhang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai You
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yan Chen
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Anteneh Getachew
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuanqi Zhuang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofen Zhong
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zuoxian Lin
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dongsheng Guo
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fan Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Tingcai Pan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongcheng Wei
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yin-Xiong Li
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| |
Collapse
|
11
|
The Thalamus Regulates Retinoic Acid Signaling and Development of Parvalbumin Interneurons in Postnatal Mouse Prefrontal Cortex. eNeuro 2019; 6:eN-NWR-0018-19. [PMID: 30868103 PMCID: PMC6385081 DOI: 10.1523/eneuro.0018-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/01/2022] Open
Abstract
GABAergic inhibitory neurons in the prefrontal cortex (PFC) play crucial roles in higher cognitive functions. Despite the link between aberrant development of PFC interneurons and a number of psychiatric disorders, mechanisms underlying the development of these neurons are poorly understood. Here we show that the retinoic acid (RA)-degrading enzyme CYP26B1 (cytochrome P450 family 26, subfamily B, member 1) is transiently expressed in the mouse frontal cortex during postnatal development, and that medial ganglionic eminence (MGE)-derived interneurons, particularly in parvalbumin (PV)-expressing neurons, are the main cell type that has active RA signaling during this period. We found that frontal cortex-specific Cyp26b1 knock-out mice had an increased density of PV-expressing, but not somatostatin-expressing, interneurons in medial PFC, indicating a novel role of RA signaling in controlling PV neuron development. The initiation of Cyp26b1 expression in neonatal PFC coincides with the establishment of connections between the thalamus and the PFC. We found that these connections are required for the postnatal expression of Cyp26b1 in medial PFC. In addition to this region-specific role in postnatal PFC that regulates RA signaling and PV neuron development, the thalamocortical connectivity had an earlier role in controlling radial dispersion of MGE-derived interneurons throughout embryonic neocortex. In summary, our results suggest that the thalamus plays multiple, temporally separate roles in interneuron development in the PFC.
Collapse
|
12
|
Dong W, Zhang Y. Retinoic acid receptor α expression exerts an anti-apoptosis effect on PC12 cells following oxygen-glucose deprivation. Exp Ther Med 2018; 16:3525-3533. [PMID: 30233705 PMCID: PMC6143853 DOI: 10.3892/etm.2018.6639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/20/2017] [Indexed: 12/12/2022] Open
Abstract
It has been established that the primary form of neuron death following hypoxic ischemic brain damage is apoptosis. Imbalances in the expression of genes in the B-cell lymphoma 2 (Bcl-2) family located in the mitochondrion, and in the expression of their encoded proteins, are key events in the mitochondrial apoptotic pathway, which lead to damage of cellular structure and function. The present study aimed to explore the regulatory effect of retinoic acid receptor α (RAR-α) on the apoptosis of PC12 cells induced by oxygen-glucose deprivation (OGD) in the retinoic acid signaling pathway. Recombinant adenovirus RAR-α small interfering RNA (Ad-siRAR-α) was used to transduce PC12 cells, and the efficiency of RAR-α expression inhibition was detected by semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). An empty adenovirus vector was transfected in PC12 cells, which were used as the control. Flow cytometry with Annexin V-propidium iodide (PI) and fluorescence probe JC-1 staining was used to detect the apoptosis rate and mitochondrial transmembrane potential (MMP), respectively, of PC12 cells after transduction with Ad-siRAR-α. Furthermore, the expression levels of key genes in the RAR-α and mitochondrial apoptotic pathway, Bcl-2 and Bcl-2-associated protein (Bax) were analyzed by RT-quantitative (q)PCR and western blot analysis. RAR-α mRNA expression was observed to be decreased in PC12 cells following OGD-induced injury, and this decrease can be reversed by 4 µmol/l ATRA treatment. After 36 h transfection with Ad-siRAR-α, RAR-α gene expression was significantly inhibited compared with the control (P<0.05). The results of Annexin V-PI, fluorescence probe JC-1 staining and flow cytometry demonstrated that the apoptosis rate significantly increased and MMP significantly decreased in OGD-induced PC12 cells following transduction with Ad-siRAR-α compared with the control (both P<0.05). RT-qPCR and western blot analysis indicated that Bax expression was significantly increased and Bcl-2 expression was significantly decreased in PC12 cells transduced with Ad-siRAR-α after OGD-induced injury at the mRNA and protein level (P<0.05). In conclusion, Ad-siRAR-α transduction could promote apoptosis in OGD-induced PC12 cells. This suggests that the expression of Bax and Bcl-2 in the mitochondrial apoptosis signaling pathway is, at least in part, mediated by RAR-α expression, thereby indicating that RAR-α expression exerts an anti-apoptotic effect on OGD-damaged PC12 cells.
Collapse
Affiliation(s)
- Wanliang Dong
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Yuankun Zhang
- Department of Cardiovascular Internal Medicine, People's Hospital of Zhengzhou, Zhengzhou, Henan 450053, P.R. China
| |
Collapse
|
13
|
Lai X, Wu X, Hou N, Liu S, Li Q, Yang T, Miao J, Dong Z, Chen J, Li T. Vitamin A Deficiency Induces Autistic-Like Behaviors in Rats by Regulating the RARβ-CD38-Oxytocin Axis in the Hypothalamus. Mol Nutr Food Res 2018; 62. [PMID: 29266770 DOI: 10.1002/mnfr.201700754] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/24/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Xi Lai
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Xiaofeng Wu
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Nali Hou
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Shu Liu
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Qing Li
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Ting Yang
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Jingkun Miao
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
- Certer for Clinical Molecular Medicine; Children's Hospital of Chongqing Medical University; Chongqing China
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders; Chongqing China
| | - Jie Chen
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
| | - Tingyu Li
- Children's Nutrition Research Center; Children's Hospital of Chongqing Medical University; Chongqing China
- Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child Development and Critical Disorders; Chongqing China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders; Chongqing China
| |
Collapse
|
14
|
Abstract
Smith-Magenis syndrome (SMS; OMIM #182290) is a complex genetic disorder characterized by distinctive physical features, developmental delay, cognitive impairment, and a typical behavioral phenotype. SMS is caused by interstitial 17p11.2 deletions, encompassing multiple genes and including the retinoic acid-induced 1 gene (RAI1), or by mutations in RAI1 itself. About 10% of all the SMS patients, in fact, carry an RAI1 mutation responsible for the phenotype. RAI1 (OMIM *607642) is a dosage-sensitive gene expressed in many tissues and highly conserved among species. Over the years, several studies have demonstrated that RAI1 (or its homologs in animal models) acts as a transcriptional factor implicated in embryonic neurodevelopment, neuronal differentiation, cell growth and cell cycle regulation, bone and skeletal development, lipid and glucose metabolisms, behavioral functions, and circadian activity. Patients with RAI1 pathogenic variants show some phenotypic differences when compared to those carrying the typical deletion. They usually have lower incidence of hypotonia and less cognitive impairment than those with 17p11.2 deletions but more frequently show the behavioral characteristics of the syndrome and overeating issues. These differences reflect the primary pathogenetic role of RAI1 without the pathogenetic contribution of the other genes included in the typical 17p11.2 deletion. The better comprehension of physiological roles of RAI1, its molecular co-workers and interactors, and its contribution in determining the typical SMS phenotype will certainly open a new path for therapeutic interventions.
Collapse
Affiliation(s)
- Mariateresa Falco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Sonia Amabile
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Fabio Acquaviva
- Department of Translational Medical Sciences (DISMET), Section of Pediatric Clinical Genetics, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
15
|
Ali H, Hamadani J, Mehra S, Tofail F, Hasan MI, Shaikh S, Shamim AA, Wu LSF, West KP, Christian P. Effect of maternal antenatal and newborn supplementation with vitamin A on cognitive development of school-aged children in rural Bangladesh: a follow-up of a placebo-controlled, randomized trial. Am J Clin Nutr 2017; 106:77-87. [PMID: 28490513 DOI: 10.3945/ajcn.116.134478] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 04/07/2017] [Indexed: 11/14/2022] Open
Abstract
Background: The impact of early vitamin A supplementation on neurodevelopmental function has not been adequately studied. In rural Bangladesh we examined cognitive and motor function and scholastic achievement in a cohort of children who were exposed to vitamin A in utero or at birth.Objective: The aim of this study was to examine independent and combined effects of antenatal and newborn supplementation with vitamin A on the cognitive function of children at 8 y of age.Design: A cohort of rural Bangladeshi children from 2 previous double-blind, placebo-controlled cluster-randomized trials were revisited at age 8 y between February 2013 and June 2014. Data on sociodemographic, social, and physical conditions; schooling; child care behavior; anthropometric measures; and cognitive function were collected with the use of various psychometric assessment tools.Results: Among 11,950 children from the parent trial who were last known to be alive, a subset of 1803 children balanced by treatment group in a selected contiguous study area were re-enrolled and 1613 (89%) provided consent for assessments. Of these, 1577 (87%) children had a complete cognitive evaluation. All groups were highly comparable on baseline variables collected in the previous trials and factors measured at re-enrollment. Overall, there was no impact of either maternal or newborn supplementation with vitamin A on intelligence, memory, and motor function. Compared with placebo, children who received both interventions had significantly better performance in reading, spelling, and math computation, with increased mean (95% CI) scores of 8.0 (2.2, 13.8), 6.8 (1.9, 11.7), and 4.8 (0.6, 9.0), respectively.Conclusions: General intelligence or memory and motor functions were not affected by antenatal or newborn supplementation with vitamin A. Scholastic performance and aspects of executive function improved when both interventions were provided. These trials were registered at clinicaltrials.gov as NCT00198822 and NCT00128557.
Collapse
Affiliation(s)
- Hasmot Ali
- The JiVitA Project, Johns Hopkins University Bangladesh, Paschim Para, Gaibandha, Bangladesh; .,Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jena Hamadani
- Child Development Unit, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sucheta Mehra
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Fahmida Tofail
- Child Development Unit, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md Imrul Hasan
- Child Development Unit, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Saijuddin Shaikh
- The JiVitA Project, Johns Hopkins University Bangladesh, Paschim Para, Gaibandha, Bangladesh.,Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | - Lee S-F Wu
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Keith P West
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Parul Christian
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.,Bill & Melinda Gates Foundation, Seattle, WA
| |
Collapse
|
16
|
Niculae AŞ, Pavăl D. From molecules to behavior: An integrative theory of autism spectrum disorder. Med Hypotheses 2016; 97:74-84. [PMID: 27876135 DOI: 10.1016/j.mehy.2016.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/02/2016] [Accepted: 10/19/2016] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) comprises a group of neurodevelopmental disorders for which various theories have been proposed. Each theory brings valuable insights and has experimental evidence backing it, yet none provides an overarching explanation for each of the pathological aspects involved in ASD. Here we present an integrative theory of ASD, centered on a sequence of events spanning from the molecular to the behavioral level. We propose that an abnormality in the interplay between retinoic acid and sex hormones predisposes an individual to specific molecular malfunctions. In turn, this molecular syndrome generates an altered brain connectivity between the cerebellum, the midbrain dopaminergic areas, and the prefrontal cortex. Lastly, this disconnection would generate specific behavioral traits traditionally involved in ASD. Therefore, this paper represents a step forward in unifying different levels of pathological features into novel integrated testable hypotheses.
Collapse
Affiliation(s)
- Alexandru-Ştefan Niculae
- The Department of Molecular Sciences, Faculty of Medicine, 'Iuliu Hațieganu' University of Medicine and Pharmacy, 6 Louis Pasteur, 400349 Cluj-Napoca, Romania
| | - Denis Pavăl
- The Department of Molecular Sciences, Faculty of Medicine, 'Iuliu Hațieganu' University of Medicine and Pharmacy, 6 Louis Pasteur, 400349 Cluj-Napoca, Romania.
| |
Collapse
|
17
|
Ababon MR, Li BI, Matteson PG, Millonig JH. Quantitative Measurement of Relative Retinoic Acid Levels in E8.5 Embryos and Neurosphere Cultures Using the F9 RARE-Lacz Cell-based Reporter Assay. J Vis Exp 2016. [PMID: 27684594 PMCID: PMC5091987 DOI: 10.3791/54443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Retinoic acid (RA) is an important developmental morphogen that coordinates anteroposterior and dorsoventral axis patterning, somitic differentiation, neurogenesis, patterning of the hindbrain and spinal cord, and the development of multiple organ systems. Due to its chemical nature as a small amphipathic lipid, direct detection and visualization of RA histologically remains technically impossible. Currently, methods used to infer the presence and localization of RA make use of reporter systems that detect the biological activity of RA. Most established reporter systems, both transgenic mice and cell lines, make use of the highly potent RA response element (RARE) upstream of the RAR-beta gene to drive RA-inducible expression of reporter genes, such as beta-galactosidase or luciferase. The transgenic RARE-LacZ mouse is useful in visualizing spatiotemporal changes in RA signaling especially during embryonic development. However, it does not directly measure overall RA levels. As a reporter system, the F9 RARE-LacZ cell line can be used in a variety of ways, from simple detection of RA to quantitative measurements of RA levels in tissue explants. Here we describe the quantitative determination of relative RA levels generated in embryos and neurosphere cultures using the F9 RARE-LacZ reporter cell line.
Collapse
Affiliation(s)
- Myka R Ababon
- Center for Advanced Biotechnology and Medicine, Rutgers University
| | - Bo I Li
- Department of Neuroscience and Cell Biology, Rutgers University
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Rutgers University
| | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Rutgers University; Department of Neuroscience and Cell Biology, Rutgers University;
| |
Collapse
|
18
|
Evidence for genetic regulation of mRNA expression of the dosage-sensitive gene retinoic acid induced-1 (RAI1) in human brain. Sci Rep 2016; 6:19010. [PMID: 26743651 PMCID: PMC4705554 DOI: 10.1038/srep19010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/02/2015] [Indexed: 12/12/2022] Open
Abstract
RAI1 (retinoic acid induced-1) is a dosage-sensitive gene that causes Smith-Magenis syndrome (SMS) when mutated or deleted and Potocki-Lupski Syndrome (PTLS) when duplicated, with psychiatric features commonly observed in both syndromes. How common genetic variants regulate this gene, however, is unknown. In this study, we found that RAI1 mRNA expression in Chinese prefrontal and temporal cortex correlate with genotypes of common single nucleotide polymorphisms (SNPs) located in the RAI1 5′-upstream region. Using genotype imputation, “R2-Δ2” analysis, and data from the RegulomeDB database, we identified SNPs rs4925102 and rs9907986 as possible regulatory variants, accounting for approximately 30–40% of the variance in RAI1 mRNA expression in both brain regions. Specifically, rs4925102 and rs9907986 are predicted to disrupt the binding of retinoic acid RXR-RAR receptors and the transcription factor DEAF1 (Deformed epidermal autoregulatory factor-1), respectively. Consistent with these predictions, we observed binding of RXRα and RARα to the predicted RAI1 target in chromatin immunoprecipitation assays. Retinoic acid is crucial for early development of the central neural system, and DEAF1 is associated with intellectual disability. The observation that a significant portion of RAI1 mRNA expression is genetically controlled raises the possibility that common RAI1 5′-region regulatory variants contribute more generally to psychiatric disorders.
Collapse
|
19
|
Bailey JM, Oliveri AN, Karbhari N, Brooks RAJ, De La Rocha AJ, Janardhan S, Levin ED. Persistent behavioral effects following early life exposure to retinoic acid or valproic acid in zebrafish. Neurotoxicology 2015; 52:23-33. [PMID: 26439099 DOI: 10.1016/j.neuro.2015.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Moderate to severe dysregulation in retinoid signaling during early development is associated with a constellation of physical malformations and/or neural tube defects, including spina bifida. It is thought that more subtle dysregulation of this system, which might be achievable via dietary (i.e. hypervitaminosis A) or pharmacological (i.e. valproic acid) exposure in humans, will manifest on behavioral domains including sociability, without overt physical abnormalities. METHODS During early life, zebrafish were exposed to low doses of two chemicals that disrupt retinoid signaling. From 0 to 5dpf, larvae were reared in aqueous solutions containing retinoic acid (0, 0.02, 0.2 or 2nM) or valproic acid (0, 0.5, 5.0 or 50μM). One cohort of zebrafish was assessed using a locomotor activity screen at 6-dpf; another was reared to adulthood and assessed using a neurobehavioral test battery (startle habituation, novel tank exploration, shoaling, and predator escape/avoidance). RESULTS There was no significant increase in the incidence of physical malformation among exposed fish compared to controls. Both retinoic acid and valproic acid exposures during development disrupted larval activity with persisting behavioral alterations later in life, primarily manifesting as decreased social affiliation. CONCLUSIONS Social behavior and some aspects of motor function were altered in exposed fish; the importance of examining emotional or psychological consequences of early life exposure to retinoid acting chemicals is discussed.
Collapse
Affiliation(s)
- Jordan M Bailey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anthony N Oliveri
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nishika Karbhari
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Roy A J Brooks
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amberlene J De La Rocha
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sheila Janardhan
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
20
|
Devanna P, Middelbeek J, Vernes SC. FOXP2 drives neuronal differentiation by interacting with retinoic acid signaling pathways. Front Cell Neurosci 2014; 8:305. [PMID: 25309332 PMCID: PMC4176457 DOI: 10.3389/fncel.2014.00305] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/10/2014] [Indexed: 11/14/2022] Open
Abstract
FOXP2 was the first gene shown to cause a Mendelian form of speech and language disorder. Although developmentally expressed in many organs, loss of a single copy of FOXP2 leads to a phenotype that is largely restricted to orofacial impairment during articulation and linguistic processing deficits. Why perturbed FOXP2 function affects specific aspects of the developing brain remains elusive. We investigated the role of FOXP2 in neuronal differentiation and found that FOXP2 drives molecular changes consistent with neuronal differentiation in a human model system. We identified a network of FOXP2 regulated genes related to retinoic acid signaling and neuronal differentiation. FOXP2 also produced phenotypic changes associated with neuronal differentiation including increased neurite outgrowth and reduced migration. Crucially, cells expressing FOXP2 displayed increased sensitivity to retinoic acid exposure. This suggests a mechanism by which FOXP2 may be able to increase the cellular differentiation response to environmental retinoic acid cues for specific subsets of neurons in the brain. These data demonstrate that FOXP2 promotes neuronal differentiation by interacting with the retinoic acid signaling pathway and regulates key processes required for normal circuit formation such as neuronal migration and neurite outgrowth. In this way, FOXP2, which is found only in specific subpopulations of neurons in the brain, may drive precise neuronal differentiation patterns and/or control localization and connectivity of these FOXP2 positive cells.
Collapse
Affiliation(s)
- Paolo Devanna
- Language and Genetics Department, Max Planck Institute for Psycholinguistics Nijmegen, Netherlands
| | - Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Netherlands
| | - Sonja C Vernes
- Language and Genetics Department, Max Planck Institute for Psycholinguistics Nijmegen, Netherlands ; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Netherlands
| |
Collapse
|
21
|
Shi Z, Shen T, Liu Y, Huang Y, Jiao J. Retinoic acid receptor γ (Rarg) and nuclear receptor subfamily 5, group A, member 2 (Nr5a2) promote conversion of fibroblasts to functional neurons. J Biol Chem 2014; 289:6415-6428. [PMID: 24459144 PMCID: PMC3945308 DOI: 10.1074/jbc.m113.515601] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/14/2014] [Indexed: 01/09/2023] Open
Abstract
Somatic cells can be reprogrammed to neurons and various other cell types with retrovirus or lentivirus. The limitation of this technology is that these genome-integration viruses may increase the risk of gene mutation and cause insertional mutagenesis. We recently found that non-integration adenovirus carrying neuronal transcription factors can induce fibroblasts to neurons. However, the conversion efficiency by the adenovirus is lower than that of the retrovirus or lentivirus. Therefore, it is crucial to identify other factors or chemical compounds to obtain neurons with high efficiency. In this study we show that the combination of Rarg (retinoic acid receptor γ) and Nr5a2 (nuclear receptor subfamily 5, group A, member 2; also known as Lrh-1 (liver receptor homologue 1)) rapidly promote the iN cell maturation within 1 week and greatly facilitate the conversion with neuronal purities of ∼50% and yields of >130%. They also improve neuronal pattern formation, electrophysiological characteristics, and functional integration in vivo. Moreover, the chemical compound agonists to Rarg and Nr5a2 function effectively as well. This approach may be used for the generation and application of iN cells in regenerative medicine.
Collapse
Affiliation(s)
- Zixiao Shi
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianjin Shen
- School of Life Sciences, Anhui University, Anhui 230601, China
| | - Yanli Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanyuan Huang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwei Jiao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
22
|
Buckley GJ, Murray-Kolb LE, Khatry SK, LeClerq SC, Wu L, West KP, Christian P. Cognitive and motor skills in school-aged children following maternal vitamin A supplementation during pregnancy in rural Nepal: a follow-up of a placebo-controlled, randomised cohort. BMJ Open 2013; 3:e002000. [PMID: 23667158 PMCID: PMC3651971 DOI: 10.1136/bmjopen-2012-002000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/22/2013] [Accepted: 04/03/2013] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE To determine the effects of maternal vitamin A supplementation from preconception through postpartum on cognitive and motor development of children at 10-13 years of age in rural Nepal. DESIGN Follow-up assessment of children born to women randomly assigned by a village to receive either supplemental vitamin A (7000 µg retinol equivalents) or placebo weekly during a continuous 3.5-year period from 1994-1997. The participants came from 12 wards, a subset of 270 wards in the original trial. Trained staff tested children for cognition by the Universal Nonverbal Intelligence Test (UNIT) and motor ability using four subtests from the Movement Assessment Battery for Children (MABC). Data on schooling, home environment and nutritional and socioeconomic status were also collected. SETTING Southern plains district of Sarlahi, Nepal. PARTICIPANTS 390 Nepalese children 10-13 years of age. MAIN OUTCOME MEASURES Raw scores on UNIT and square-root transformed scores on an abridged version of the MABC tests, expressed as cluster-summarised (mean±SD) values to account for the design of the original trial. RESULTS There were no differences in UNIT (79.61±5.99 vs 80.69±6.71) or MABC (2.64±0.07 vs 2.49±0.09) test scores in children whose mothers were exposed to vitamin A vs placebo (mean differences: -1.07, 95% CI -7.10 to 9.26, p=0.78; 0.15, 95% CI 0.43 to -0.08, p=0.15), respectively. More children in the placebo group had repeated a grade in school (28% of placebo vs 16.7% of vitamin A, p=0.01). CONCLUSIONS Preconceptional to postpartum maternal vitamin A supplementation, in an undernourished setting, does not improve cognition or motor development at ages 10-13 years.
Collapse
Affiliation(s)
- Gillian J Buckley
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Laura E Murray-Kolb
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Subarna K Khatry
- Nepal Nutrition Intervention Project, Sarlahi (NNIPS), Nepal Netra Jyoti Sangh, Tripureswor, Kathmandu, Nepal
| | - Steven C LeClerq
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Keith P West
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Parul Christian
- Center for Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Mawson AR. Toward a theory of childhood learning disorders, hyperactivity, and aggression. ISRN PSYCHIATRY 2012; 2012:589792. [PMID: 23762766 PMCID: PMC3671718 DOI: 10.5402/2012/589792] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 06/14/2012] [Indexed: 02/03/2023]
Abstract
Learning disorders are often associated with persistent hyperactivity and aggression and are part of a spectrum of neurodevelopmental disorders. A potential clue to understanding these linked phenomena is that physical exercise and passive forms of stimulation are calming, enhance cognitive functions and learning, and are recommended as complementary treatments for these problems. The theory is proposed that hyperactivity and aggression are intense stimulation-seeking behaviors (SSBs) driven by increased brain retinergic activity, and the stimulation thus obtained activates opposing nitrergic systems which inhibit retinergic activity, induce a state of calm, and enhance cognition and learning. In persons with cognitive deficits and associated behavioral disorders, the retinergic system may be chronically overactivated and the nitrergic system chronically underactivated due to environmental exposures occurring pre- and/or postnatally that affect retinoid metabolism or expression. For such individuals, the intensity of stimulation generated by SSB may be insufficient to activate the inhibitory nitrergic system. A multidisciplinary research program is needed to test the model and, in particular, to determine the extent to which applied physical treatments can activate the nitrergic system directly, providing the necessary level of intensity of sensory stimulation to substitute for that obtained in maladaptive and harmful ways by SSB, thereby reducing SSB and enhancing cognitive skills and performance.
Collapse
Affiliation(s)
- Anthony R. Mawson
- School of Health Sciences, College of Public Service, Jackson State University, 350 West Woodrow Wilson Drive, Room 229, Jackson, MS 39213, USA
| |
Collapse
|
24
|
Bao Y, Ibram G, Blaner WS, Quesenberry CP, Shen L, McKeague IW, Schaefer CA, Susser ES, Brown AS. Low maternal retinol as a risk factor for schizophrenia in adult offspring. Schizophr Res 2012; 137:159-65. [PMID: 22381190 PMCID: PMC3520602 DOI: 10.1016/j.schres.2012.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 02/01/2012] [Accepted: 02/03/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND Prenatal micronutrient deficiency has been linked to later development of schizophrenia among offspring; however, no study has specifically investigated the association between vitamin A and this disorder. Vitamin A is an essential nutrient which is required by the early embryo and fetus for gene expression and regulation, cell differentiation, proliferation and migration. Previous work suggests that vitamin A deficiency in the second trimester may be particularly relevant to the etiopathogenesis of neurobehavioral phenotypes some of which are observed in schizophrenia. METHODS We examined whether low maternal vitamin A levels in the second trimester are associated with the risk of schizophrenia and other schizophrenia spectrum disorders (SSD) in the Prenatal Determinants of Schizophrenia study; third trimester vitamin A levels were also examined in relation to SSD. The cases were derived from a population-based birth cohort; all cohort members belonged to a prepaid health plan. Archived maternal serum samples were assayed for vitamin A in cases (N=55) and up to 2 controls per case (N=106) matched on length of membership in the health plan, date of birth (±28 days), sex, and gestational timing and availability of archived maternal sera. RESULTS For the second trimester, low maternal vitamin A, defined as values in the lowest tertile of the distribution among controls, was associated with a greater than threefold increased risk of SSD, adjusting for maternal education and age (OR=3.04, 95% CI=1.06, 8.79, p=.039). No association between third trimester maternal vitamin A and SSD was observed. CONCLUSIONS Although further investigations are warranted, this is the first birth cohort study to our knowledge to report an association between low maternal vitamin A levels and SSD among offspring.
Collapse
Affiliation(s)
- YuanYuan Bao
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States
| | - Ghionul Ibram
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY 10032, United States
| | - William S. Blaner
- Institute of Human Nutrition, College of Physicians and Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, United States
| | - Charles P. Quesenberry
- Division of Research, Kaiser Permanente, 3505 Broadway, Oakland, CA 94611, United States
| | - Ling Shen
- Division of Research, Kaiser Permanente, 3505 Broadway, Oakland, CA 94611, United States
| | - Ian W. McKeague
- Department of Biostatistics, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY 10032, United States
| | - Catherine A. Schaefer
- Division of Research, Kaiser Permanente, 3505 Broadway, Oakland, CA 94611, United States
| | - Ezra S. Susser
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY 10032, United States
| | - Alan S. Brown
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY 10032, United States
- Corresponding author at: New York State Psychiatric Institute, 1051 Riverside Drive, Unit 23, New York, NY 10032, United States. Tel.: +1 212 543 5629. (A.S. Brown)
| |
Collapse
|
25
|
Goldberg JS. Monitoring maternal Beta carotene and retinol consumption may decrease the incidence of neurodevelopmental disorders in offspring. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2011; 6:1-8. [PMID: 24453512 PMCID: PMC3888066 DOI: 10.4137/cmrh.s8372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retinoic acids (13-cis and 13-trans) are known teratogens, and their precursor is retinol, a form of vitamin A. In 1995, Rothman et al demonstrated an association between excessive vitamin A, >10,000 IU/day, during the first trimester of pregnancy and teratogenic effects, particularly in the central nervous system. However, vitamin A deficiency has long been known to be deleterious to the mother and fetus. Therefore, there may be a narrow therapeutic ratio for vitamin A during pregnancy that has not previously been fully appreciated. Neurodevelopmental disorders may not be apparent by macroscopic brain examination or imaging, and proving the existence of a behavioral teratogen is not straightforward. However, an excess of retinoic acid and some neurodevelopmental disorders are both associated with abnormalities in cerebellar morphology. Physical and chemical evidence strongly supports the notion that beta carotene crosses the placenta and is metabolized to retinol. Only very limited amounts of beta carotene are stored in fetal fat cells as evidenced by the fact that maternal fat is yellow from beta carotene, whereas non-brown neonatal fat is white. Furthermore, newborns of carotenemic mothers do not share the yellow complexion of their mothers. The excess 13-trans retinoic acid derived from metabolized beta carotene in the fetus increases the concentration of the more teratogenic 13-cis retinoic acid since the isomerization equilibrium is shifted to the left. Therefore, this paper proposes that consideration be given to monitoring all potential sources of fetal 13-cis and 13-trans retinoic acid, including nutritional supplements, dietary retinol, and beta carotene, particularly in the first trimester of pregnancy.
Collapse
Affiliation(s)
- Joel S Goldberg
- Durham Veterans Affairs Medical Center and Duke University, 508 Fulton Street, Durham, NC, 27705
| |
Collapse
|
26
|
Are retinoids potential therapeutic agents in disorders of social cognition including autism? FEBS Lett 2011; 585:1529-36. [PMID: 21557943 DOI: 10.1016/j.febslet.2011.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 05/03/2011] [Accepted: 05/03/2011] [Indexed: 11/20/2022]
Abstract
Increasing evidence suggests that the nonapeptide, oxytocin (OT), helps shape social and affiliative behaviors not only in lower mammals but also in humans. Recently, an essential mediator of brain OT release has been discovered, ADP-ribosyl cyclase and/or CD38. We have subsequently shown that polymorphisms across the CD38 gene are associated with autism spectrum disorders (ASD). Notably, CD38 expression in lymphoblastoid cells (LBC) is reduced in cell lines derived from ASD subjects compared to parental cell lines. Intriguingly, a correlation was observed between CD38 expression and measures of social function in ASD. Finally, we have shown that all-trans retinoic acid (ATRA), a known inducer of CD38 transcription, can rescue low CD38 expressing LBC lines derived from ASD subjects and restore normal levels of transcription of this ectoenzyme providing 'proof of principle' in a peripheral model that retinoids are potential therapeutic agents in ASD.
Collapse
|
27
|
Jiang W, Wen EY, Gong M, Shi Y, Chen L, Bi Y, Zhang Y, Liu YF, Chen J, Qu P, Liu YX, Wei XP, Li TY. The pattern of retinoic acid receptor expression and subcellular, anatomic and functional area translocation during the postnatal development of the rat cerebral cortex and white matter. Brain Res 2011; 1382:77-87. [PMID: 21241670 DOI: 10.1016/j.brainres.2011.01.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/16/2010] [Accepted: 01/10/2011] [Indexed: 11/16/2022]
Abstract
Retinoic acid (RA), which is an important modulator of brain development, neural cell proliferation, neurite outgrowth, and synaptic plasticity, is regulated via changes in RA receptors. The pattern of RA receptor changes in the rat cerebral cortex and white matter during postnatal development has not been extensively studied. Therefore, we studied the mRNA expression patterns of 6 RA receptors in the postnatal rat cerebral cortex and white matter at 1, 3, 7, 10, 14, 21, 28, and 35days. We found that RARβ, RXRα and RXRβ mRNA levels gradually increased during postnatal development. RARα presented a nearly unimodal trend, but RARγ and RXRγ were generally bimodal. RARα, RARγ, and RXRγ mRNA levels peaked at postnatal day 21 (P21). The pattern of RARα expression was consistent with that of its mRNA levels. RARα and RXRγ mRNA levels were the highest among those of all RA receptors during postnatal development. Interestingly, RARα expression was mainly located in the cytoplasm in the postnatal development apart from P3d. We further showed that RARα is expressed mainly in layers 2 and 3, partly in layers 1 and 4, and in a limited manner in layers 5 and 6 in the parietal cortex. Most RARα expression occurs in layers 2, 3, and 4 in the temporal lobe cortex. We realized that the M1 S2 region of RARα is highly expressed and that the position of RARα changes dynamically to meet the needs of different regions during development. These results support the idea that the RA receptor plays an important role in the cerebrum during postnatal development and implementation of these functions may be mainly dependent on the non-transcriptional or post- transcriptional regulation.
Collapse
Affiliation(s)
- Wei Jiang
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sakai Y, Dräger UC. Detection of retinoic acid catabolism with reporter systems and by in situ hybridization for CYP26 enzymes. Methods Mol Biol 2010; 652:277-94. [PMID: 20552435 DOI: 10.1007/978-1-60327-325-1_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Retinoic acid (RA), an active form of vitamin A, is essential for life in vertebrates, owing to its capacity of influencing expression of a sizable fraction of all genes and proteins. It functions via two modes: (1) as controlling ligand for specific transcription factors in the nucleus it stimulates or inhibits gene expression from RA response elements in gene promoters; (2) in non-genomic pathways it activates kinase-signaling cascades that converge with additional influences to regulate gene expression and mRNA translation. RA performs a critical role in morphogenesis of the developing embryo, which is reflected in spatio-temporally changing expression patterns of RA-synthesizing and RA-degrading enzymes and in its biophysical characteristics as a small diffusible lipid. Because its histological localization cannot be directly visualized for technical reasons, its sites of action in vivo are inferred from the locations of the metabolic enzymes and through use of two kinds of RA reporter systems. Here we explain techniques for use of RA reporter cells and RA reporter mice, and we describe in situ hybridization methods for the three major RA-degrading enzymes: CYP26A1, CYP26B1, and CYP26C1. Comparisons of the different indicators for sites of RA signaling demonstrate that local RA peaks and troughs are important for inferring some but not all locations of RA actions. When integrated within cells of living mice, expression of the RA reporter construct is rarely a simple measure of local RA levels, especially in the developing brain, but it appears to provide cues to an RA involvement in site-specific regulatory networks in combination with other spatial determinants.
Collapse
Affiliation(s)
- Yasuo Sakai
- Department of Plastic Surgery, Osaka University School of Medicine, Osaka, Japan
| | | |
Collapse
|
29
|
Mawson AR. On the association between low resting heart rate and chronic aggression: retinoid toxicity hypothesis. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:205-13. [PMID: 19063930 DOI: 10.1016/j.pnpbp.2008.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 10/03/2008] [Accepted: 10/24/2008] [Indexed: 11/19/2022]
Abstract
Low resting heart rate is a strong and consistent predictor of conduct disorder and chronic aggression. Explanations such as fearlessness and low arousal-induced stimulus-seeking have been offered, assuming a causal association between the phenomena, but the origin of low heart rate and its significance for understanding aggression and violence remain obscure. Retinoids (vitamin A and its congeners) play important roles in embryogenesis and neural development. Several lines of evidence also suggest a causal role of retinoids in aggression as well as cognitive and mood disorders. The hypothesis is proposed that retinoid overexpression in utero induces, via a noradrenergic-to-cholinergic switch, alterations in cardiac functioning and hemodynamics resulting in low resting heart rate, brain structural and functional changes, minor physical anomalies, and persistent aggression. Retinoid toxicity occurring early in pregnancy could represent a final common pathway by which various prenatal challenges result in conduct disorder and chronic aggression (e.g., maternal cigarette smoking, alcohol consumption, drug use, exposure to environmental chemicals, stress, trauma or infection). Implications of the model for understanding related aspects of chronic aggression are discussed, as well as strategies for prevention and treatment.
Collapse
Affiliation(s)
- Anthony R Mawson
- Department of Pediatrics, Division of Genetics and Epidemiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi 39216, USA.
| |
Collapse
|
30
|
Bremner JD, McCaffery P. The neurobiology of retinoic acid in affective disorders. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32:315-31. [PMID: 17707566 PMCID: PMC2704911 DOI: 10.1016/j.pnpbp.2007.07.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 07/02/2007] [Accepted: 07/02/2007] [Indexed: 01/29/2023]
Abstract
Current models of affective disorders implicate alterations in norepinephrine, serotonin, dopamine, and CRF/cortisol; however treatments targeted at these neurotransmitters or hormones have led to imperfect resolution of symptoms, suggesting that the neurobiology of affective disorders is incompletely understood. Until now retinoids have not been considered as possible contributors to affective disorders. Retinoids represent a family of compounds derived from vitamin A that perform a large number of functions, many via the vitamin A product, retinoic acid. This signaling molecule binds to specific retinoic acid receptors in the brain which, like the glucocorticoid and thyroid hormone receptors, are part of the nuclear receptor superfamily and regulate gene transcription. Research in the field of retinoic acid in the CNS has focused on the developing brain, in part stimulated by the observation that isotretinoin (13-cis retinoic acid), an isomer of retinoic acid used in the treatment of acne, is highly teratogenic for the CNS. More recent work has suggested that retinoic acid may influence the adult brain; animal studies indicated that the administration of isotretinoin is associated with alterations in behavior as well as inhibition of neurogenesis in the hippocampus. Clinical evidence for an association between retinoids and depression includes case reports in the literature, studies of health care databases, and other sources. A preliminary PET study in human subjects showed that isotretinoin was associated with a decrease in orbitofrontal metabolism. Several studies have shown that the molecular components required for retinoic acid signaling are expressed in the adult brain; the overlap of brain areas implicated in retinoic acid function and stress and depression suggest that retinoids could play a role in affective disorders. This report reviews the evidence in this area and describes several systems that may be targets of retinoic acid and which contribute to the pathophysiology of depression.
Collapse
Affiliation(s)
- J Douglas Bremner
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA 30306, USA.
| | | |
Collapse
|
31
|
Ruano D, Macedo A, Soares MJ, Valente J, Azevedo MH, Hutz MH, Gama CS, Lobato MI, Belmonte-de-Abreu P, Goodman AB, Pato C, Saraiva MJ, Heutink P, Palha JA. Transthyretin: no association between serum levels or gene variants and schizophrenia. J Psychiatr Res 2007; 41:667-72. [PMID: 16716350 DOI: 10.1016/j.jpsychires.2006.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Revised: 03/13/2006] [Accepted: 04/04/2006] [Indexed: 11/18/2022]
Abstract
It has been proposed that schizophrenia results from an environmental insult in genetically predisposed individuals. Environmental factors capable of modulating transcriptional activity and their carriers could link the genetic and environmental components of schizophrenia. Among these is transthyretin (TTR), a major carrier of thyroid hormones and retinol-binding protein (RBP). Retinoids and thyroid hormones regulate the expression of several genes, both during development and in the adult brain. Decreased TTR levels have been reported in the cerebrospinal fluid of patients with depression and Alzheimer's disease, and the absence of TTR influences behavior in mice. DNA variants capable of altering TTR ability to carry its ligands, either due to reduced transcription of the gene or to structural modifications of the protein, may influence development of the central nervous system and behavior. In the present study we searched for variants in the regulatory and coding regions of the TTR gene, and measured circulating levels of TTR and RBP. We found a novel single nucleotide polymorphism (SNP), ss46566417, 18 bp upstream of exon 4. Neither this SNP nor the previously described rs1800458 were found associated with schizophrenia. In addition, serum TTR and RBP levels did not differ between mentally healthy and schizophrenic individuals. In conclusion, our data does not support an involvement of the TTR gene in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Dina Ruano
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wagner E, Luo T, Sakai Y, Parada LF, Dräger UC. Retinoic acid delineates the topography of neuronal plasticity in postnatal cerebral cortex. Eur J Neurosci 2006; 24:329-40. [PMID: 16836633 DOI: 10.1111/j.1460-9568.2006.04934.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Retinoic acid is well recognized to promote neuronal differentiation in the embryonic nervous system, but how it influences the postnatal cerebral cortex remains largely unknown. The domain of highest retinoic acid actions in the cortex of the mouse constricts postnatally to a narrow band that includes the dorsal visual stream and the attentional and executive networks. This band of cortex, which is distinguished by the retinoic acid-synthesizing enzyme RALDH3, exhibits signs of delayed maturation and enhanced plasticity compared to the surrounding cortex, as indicated by suppression of parvalbumin, neurofilament, cytochrome oxidase and perineuronal net maturation, and persistence of the embryonic, polysialated form of the neural cell-adhesion molecule PSA-NCAM. During the first postnatal week, the RALDH3-expressing territory translocates in the caudal cortex from the medial limbic lobe to the adjacent neocortex. This topographical shift requires the neurotrophin NT-3 because in mice lacking neuronal NT-3 the RALDH3 enzyme maintains its early postnatal pattern up to adulthood. In the NT-3-null mutants, expression of the markers, whose topography colocalizes with RALDH3 in the normal cortex, matches the abnormal RALDH3 pattern. This indicates that the uneven retinoic acid distribution serves a role in patterning the maturation and to some extent function of the normal postnatal cerebral cortex.
Collapse
Affiliation(s)
- Elisabeth Wagner
- Eunice Kennedy Shriver Center for Mental Retardation, 200 Trapelo Road., Waltham, MA 02452, USA
| | | | | | | | | |
Collapse
|
33
|
Palha JA, Goodman AB. Thyroid hormones and retinoids: a possible link between genes and environment in schizophrenia. BRAIN RESEARCH REVIEWS 2006; 51:61-71. [PMID: 16325258 DOI: 10.1016/j.brainresrev.2005.10.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 08/28/2005] [Accepted: 10/03/2005] [Indexed: 12/14/2022]
Abstract
Phenotypic discordance for schizophrenia in monozygotic twins clearly indicates involvement of environmental factors as key determinants in disease development. Positive findings from genome scans, linkage and association studies apply in only a minority of those affected, while post-mortem brain investigations reveal altered expression of genes and proteins involved in numerous neurodevelopmental, metabolic and neurotransmitter pathways. Such altered expressions could result, on the one hand, from mutations in coding regions or polymorphisms in the promoter and regulatory regions in genes within those areas identified by gene searches or, on the other hand, from inadequate amounts of modulators, transporters and synthesizers of transcription factors necessary for regulation of the putative genes. Hormones and vitamins are such modulators. They could serve as bridges between genes and environment in schizophrenia. Multiple evidence supports the suggestion of retinoids and thyroid hormones as plausible actors in these roles. Both are not only essential for normal development of the central nervous system but also regulate the expression of many neurotransmitters, their synthesizing enzymes and receptors, and other genes in broader signaling transduction cascades affecting pathways that are altered in response to treatment. Functional and positional candidate genes include retinoic acid and thyroid hormone receptors, retinaldehyde dehydrogenases and deiodinases, which synthesize the powerful morphogens, retinoic acid and triiodothyronine, and the enzymes involved in their inactivation. This review highlights selective evidence supporting the retinoid and thyroid hormone hypotheses of schizophrenia.
Collapse
Affiliation(s)
- Joana Almeida Palha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal.
| | | |
Collapse
|
34
|
Abstract
Retinoic acid, an active form of vitamin A, regulates gene expression throughout the body, and many components of the signaling system through which it acts are present in the brain. Very little is known, however, about how retinoic acid functions in neurobiological systems. Several studies have provided evidence that retinoic acid plays a role in sleep, learning, and memory, but the precise mechanisms through which it influences these processes remain unclear. All of these processes involve local or long-range inhibition and synchronized neuronal activity between separate locations in the brain. A critical component in the generation of the synchronized firing of cortical neurons (cortical synchrony) is a network of inhibitory interneurons containing parvalbumin, a cell population affected by retinoid perturbations, such as exposure to a vitamin A overdose. An understanding of the role of retinoids in normal brain function would provide clues to the long-standing question of whether abnormalities in retinoic acid signaling contribute to the pathogenesis of some brain diseases with uncertain etiologies that involve both genetic and environmental factors.
Collapse
Affiliation(s)
- Ursula C Dräger
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, Waltham, MA 02452, USA.
| |
Collapse
|
35
|
McCaffery P, Zhang J, Crandall JE. Retinoic acid signaling and function in the adult hippocampus. ACTA ACUST UNITED AC 2006; 66:780-91. [PMID: 16688774 DOI: 10.1002/neu.20237] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Retinoic acid (RA) is an essential growth factor, derived from vitamin A, which controls growth by activating specific receptors that are members of the nuclear receptor family of transcriptional regulators. Its function in control of growth and differentiation in the embryonic CNS has been extensively investigated, but a role for RA in the mature brain has only recently become apparent. Although the adult CNS has much less capacity for change compared to the embryonic CNS, a limited amount of flexibility, referred to as neural plasticity, still exists. It is these processes that RA influences in the adult brain, including long-term potentiation and neurogenesis. The hippocampus is a brain region dependent upon neural plasticity for its function in learning and memory, and this review focuses on the roles that RA may play in regulating these processes in the adult.
Collapse
Affiliation(s)
- Peter McCaffery
- UMMS/E.K. Shriver Center, Waltham, Massachusetts 02452, USA.
| | | | | |
Collapse
|