1
|
Peixoto-Rodrigues MC, Monteiro-Neto JR, Teglas T, Toborek M, Soares Quinete N, Hauser-Davis RA, Adesse D. Early-life exposure to PCBs and PFAS exerts negative effects on the developing central nervous system. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136832. [PMID: 39689563 DOI: 10.1016/j.jhazmat.2024.136832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/18/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
Persistent organic pollutants (POPs) are ubiquitous in the environment and display the capacity to bioaccumulate in living organisms, constituting a hazard to both wildlife and humans. Although restrictions have been applied to prohibit the production of several POPs since the 1960s, high levels of these compounds can still be detected in many environmental and biological matrices, due to their chemical properties and significantly long half-lives. Some POPs can be passed from mother to the fetus and can gain entry to the central nervous system (CNS), by crossing the blood-brain barrier (BBB), resulting in significant deleterious effects, including neurocognitive and psychiatric abnormalities, which may lead to long-term socio-economic burdens. A growing body of evidence obtained from clinical and experimental studies has increasingly indicated that these POPs may influence neurodevelopment through several cellular and molecular mechanisms. However, studies assessing their mechanisms of action are still incipient, requiring further research. Polychlorinated biphenyls (PCBs) and per- and polyfluoroalkyl substances (PFAS) are two of the main classes of POPs associated with disturbances in different human systems, mainly the nervous and endocrine systems. This narrative review discusses the main PCB and PFAS effects on the CNS, focusing on neuroinflammation and oxidative stress and their consequences for neural development and BBB integrity. Moreover, we propose which mechanisms could be involved in POP-induced neurodevelopmental defects. In this sense, we highlight potential cellular and molecular pathways by which these POPs can affect neurodevelopment and could be further explored to propose preventive therapies and formulate public health policies.
Collapse
Affiliation(s)
- Maria Carolina Peixoto-Rodrigues
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fiocruz, Brazil; Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Brazil
| | | | - Timea Teglas
- Research Institute of Sport Science, Hungarian University of Sports Science, Budapest, Hungary; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Hungarian University of Sports Science, Budapest, Hungary
| | - Michal Toborek
- Institute of Physiotherapy and Health Sciences, Blood-Brain Barrier Research Center, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Natalia Soares Quinete
- Departament of Chemistry and Biochemistry & Institute of Environment, Florida International University, Miami, Florida, United States
| | - Rachel Ann Hauser-Davis
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fiocruz, Brazil
| | - Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Brazil; Laboratory of Ocular Immunology and Transplantation, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
2
|
López-Ramos JC, Martínez-Lara E, Serrano J, Fernández P, Parras GG, Ruiz-Marcos A, Rodrigo J. Nitric oxide synthase system in the brain development of neonatal hypothyroid rats. Neuroscience 2025; 565:155-171. [PMID: 39461663 DOI: 10.1016/j.neuroscience.2024.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Thyroid hormones play an important morphogenetic role during the fetal and neonatal periods and regulate numerous metabolic processes. In the central nervous system, they control myelination and overall brain development, regional gene expression, and regulation of oxygen consumption. Their deficiency in the fetal and neonatal periods causes severe mental retardation, due to lack of thyroid function, or to iodine deficiency. At the same time, nitric oxide is an atypical neurotransmitter that also has special relevance in neuronal development and plasticity and functions as a vasodilator, regulating cerebral blood flow. Although under physiological conditions it functions as a neuroprotector, in excess it can be neurotoxic. We have studied, by immunocytochemical and Western blot techniques, the evolution of the expression of neuronal and inducible isoforms of the enzyme nitric oxide synthase, and of nitrotyrosine as a marker of protein nitration produced by the presence of nitric oxide, during the early stages of postnatal brain development. We induced hypothyroidism by administering mercaptomethylimidazole to pregnant mothers, from the seventh day of gestation until the sacrifice of the offspring. The results show a delay in the evolution of the expression of the two isoforms of the enzyme nitric oxide synthase in hypothyroid animals, followed by an anomalous overexpression in later stages. Finally, the expression of nitrotyrosine follows an evolution that is synchronized with that shown by both isoenzymes in control and hypothyroid animals.
Collapse
Affiliation(s)
- Juan Carlos López-Ramos
- División de Neurociencias, Universidad Pablo de Olavide, ES-41013 Sevilla, Spain; Instituto Cajal, Avda. Doctor Arce, 24, 28002 Madrid, Spain.
| | - Esther Martínez-Lara
- Departmento de Biología Experimental, Campus de Las Lagunillas s/n, Universidad de Jaén, 23071 Jaén, Spain
| | - Julia Serrano
- Instituto Cajal, Avda. Doctor Arce, 24, 28002 Madrid, Spain
| | | | - Gloria G Parras
- División de Neurociencias, Universidad Pablo de Olavide, ES-41013 Sevilla, Spain
| | | | - José Rodrigo
- Instituto Cajal, Avda. Doctor Arce, 24, 28002 Madrid, Spain
| |
Collapse
|
3
|
Vujovic F, Farahani RM. Thyroid Hormones and Brain Development: A Focus on the Role of Mitochondria as Regulators of Developmental Time. Cells 2025; 14:150. [PMID: 39936942 PMCID: PMC11816491 DOI: 10.3390/cells14030150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/13/2025] Open
Abstract
Thyroid hormones (THs) regulate metabolism in a homeostatic state in an adult organism. During the prenatal period, prior to the establishment of homeostatic mechanisms, THs assume additional functions as key regulators of brain development. Here, we focus on reviewing the role of THs in orchestrating cellular dynamics in a developing brain. The evidence from the reviewed scientific literature suggests that the developmental roles of the hormones are predominantly mediated by non-genomic mitochondrial effects of THs due to attenuation of genomic effects of THs that antagonise non-genomic impacts. We argue that the key function of TH signalling during brain development is to orchestrate the tempo of self-organisation of neural progenitor cells. Further, evidence is provided that major neurodevelopmental consequences of hypothyroidism stem from an altered tempo of cellular self-organisation.
Collapse
Affiliation(s)
- Filip Vujovic
- IDR/WSLHD Research and Education Network, Sydney, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Ramin M Farahani
- IDR/WSLHD Research and Education Network, Sydney, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
4
|
Cainelli E, Vedovelli L, Bisiacchi P. The mother-child interface: A neurobiological metamorphosis. Neuroscience 2024; 561:92-106. [PMID: 39427701 DOI: 10.1016/j.neuroscience.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
From the start of pregnancy, mother and child induce reciprocal neurobiological changes in the brain that will prove critical for neurodevelopment and survival of both. Molecular communication between mother and fetus is constantly active and persists even after the fetus starts to synthesize its hormones in late gestation. Intriguingly, some mother and fetus exchange cells remain in the other's brain and body with long-lasting effects and memories that do not follow the laws of classical genetics but involve complex epigenetic mechanisms. After childbirth, mother and child go through a transitional phase, a sort of limbo in which both will have a peculiar functioning profile, which is adaptive for contingencies but also renders them vulnerable. The interplay between these two "limbo" states allows for an easier transition to the subsequent phases of development. In this review, we will trace mother's and child's path from pregnancy to the months following birth and, in particular, unravel i) the key features of pregnancy and brain development and the reciprocal influences; ii) how a transitory pattern of functioning characterize mother and child, moving them toward more flexible and evolved forms; and iii) how mother and fetus act during childbirth to promote neuroprotection, pain reduction, and neurophysiological changes. Therefore, this review covers a wide range of topics, integrating neuroanatomical, neurological, biochemical, neurophysiological, and psychological studies in a meaningful way, trying to integrate them in a holistic view of the mother-child interface that is usually neglected.
Collapse
Affiliation(s)
- Elisa Cainelli
- Department of General Psychology, University of Padova, 35131 Padova, Italy.
| | - Luca Vedovelli
- Unit of Biostatistics, Epidemiology, and Public Health, Department of Cardiac, Thoracic, Vascular and Public Health Sciences, University of Padova, 35131 Padova, Italy.
| | - Patrizia Bisiacchi
- Department of General Psychology, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, PNC, 35131 Padova, Italy.
| |
Collapse
|
5
|
O’Shaughnessy KL, Sasser AL, Bell KS, Riutta C, Ford JL, Grindstaff R, Gilbert ME. Bypassing the brain barriers: upregulation of serum miR-495 and miR-543-3p reflects thyroid-mediated developmental neurotoxicity in the rat. Toxicol Sci 2024; 198:128-140. [PMID: 38070162 PMCID: PMC11697567 DOI: 10.1093/toxsci/kfad125] [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: 02/29/2024] Open
Abstract
Evaluating the neurodevelopmental effects of thyroid-disrupting chemicals is challenging. Although some standardized developmental and reproductive toxicity studies recommend serum thyroxine (T4) measures in developing rats, extrapolating between a serum T4 reduction and neurodevelopmental outcomes is not straightforward. Previously, we showed that the blood-brain and blood-cerebrospinal fluid barriers may be affected by developmental hypothyroidism in newborn rats. Here, we hypothesized that if the brain barriers were functionally disturbed by abnormal thyroid action, then small molecules may escape from the brain tissue and into general circulation. These small molecules could then be identified in blood samples, serving as a direct readout of thyroid-mediated developmental neurotoxicity. To address these hypotheses, pregnant rats were exposed to propylthiouracil (PTU, 0 or 3 ppm) to induce thyroid hormone insufficiency, and dams were permitted to give birth. PTU significantly reduced serum T4 in postnatal offspring. Consistent with our hypothesis, we show that tight junctions of the brain barriers were abnormal in PTU-exposed pups, and the blood-brain barrier exhibited increased permeability. Next, we performed serum microRNA Sequencing (miRNA-Seq) to identify noncoding RNAs that may reflect these neurodevelopmental disturbances. Of the differentially expressed miRNAs identified, 7 were upregulated in PTU-exposed pups. Validation by qRT-PCR shows that miR-495 and miR-543-3p were similarly upregulated in males and females. Interestingly, these miRNAs have been linked to cell junction dysfunction in other models, paralleling the identified abnormalities in the rat brain. Taken together, these data show that miR-495 and miR-543-3p may be novel in vivo biomarkers of thyroid-mediated developmental neurotoxicity.
Collapse
Affiliation(s)
- Katherine L. O’Shaughnessy
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
| | - Aubrey L. Sasser
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA 37831
| | - Kiersten S. Bell
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA 37831
| | - Cal Riutta
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA 37831
| | - Jermaine L. Ford
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27709
| | - Rachel Grindstaff
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
| | - Mary E. Gilbert
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC, USA 27709
| |
Collapse
|
6
|
Bernal J, Morte B, Diez D. Thyroid hormone regulators in human cerebral cortex development. J Endocrinol 2022; 255:R27-R36. [PMID: 36219489 DOI: 10.1530/joe-22-0189] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022]
Abstract
Brain development is critically dependent on the timely supply of thyroid hormones. The thyroid hormone transporters are central to the action of thyroid hormones in the brain, facilitating their passage through the blood-brain barrier. Mutations of the monocarboxylate transporter 8 (MCT8) cause the Allan-Herndon-Dudley syndrome, with altered thyroid hormone concentrations in the blood and profound neurological impairment and intellectual deficit. Mouse disease models have revealed interplay between transport, deiodination, and availability of T3 to receptors in specific cells. However, the mouse models are not satisfactory, given the fundamental differences between the mouse and human brains. The goal of the present work is to review human neocortex development in the context of thyroid pathophysiology. Recent developments in single-cell transcriptomic approaches aimed at the human brain make it possible to profile the expression of thyroid hormone regulators in single-cell RNA-Seq datasets of the developing human neocortex. The data provide novel insights into the specific cellular expression of thyroid hormone transporters, deiodinases, and receptors.
Collapse
Affiliation(s)
- Juan Bernal
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Morte
- Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Diez
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
7
|
Sahani SK, Pathak A, Nepali B, Rai N. Lissencephaly with Congenital Hypothyroidism: A Case Report. JNMA J Nepal Med Assoc 2022; 60:978-981. [PMID: 36705174 PMCID: PMC9795095 DOI: 10.31729/jnma.7893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Lissencephaly is a malformation of cortical development associated with deficient neuronal migration and abnormal formation of cerebral convolutions or gyri. The lissencephaly spectrum consists of agyria, pachygyria, and subcortical band heterotopia. At least 19 genes have been identified in the causation of lissencephaly and related syndrome. Lissencephaly is associated with many other congenital disorders but the association of lissencephaly with congenital hypothyroidism is rarely reported. We report a case of a 10-year-old girl having lissencephaly with congenital hypothyroidism. Early diagnosis of lissencephaly and genetic counselling can be made in suspected cases and further possible interventions can be taken. Also, regular follow-up, monitoring, and better conservative management lead to a better prognosis. Keywords congenital abnormalities; hypothyroidism; lissencephaly; neuronal migration disorders.
Collapse
Affiliation(s)
| | - Anil Pathak
- KIST Medical College and Teaching Hospital, Imadol, Lalitpur, Nepal,Correspondence: Mr Anil Pathak, KIST Medical College and Teaching Hospital, Imadol, Lalitpur, Nepal. , Phone: +977-9867225086
| | - Bishal Nepali
- KIST Medical College and Teaching Hospital, Imadol, Lalitpur, Nepal
| | - Nilshan Rai
- KIST Medical College and Teaching Hospital, Imadol, Lalitpur, Nepal
| |
Collapse
|
8
|
Su Y, Yang X, Yang L, Liu X, She Z, Zhang Y, Dong Z. Thyroid hormones regulate reelin expression in neuropsychiatric disorders. Can J Physiol Pharmacol 2022; 100:1033-1044. [PMID: 36166833 DOI: 10.1139/cjpp-2022-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The incidence and prevalence of hypothyroidism in pregnancy have increased over the past two decades, leading to the occurrence of neuropsychiatric disorders. However, the underlying mechanisms of thyroid hormone (TH)-regulated gene expression and neuropsychiatric development during the postnatal period remain unknown. Recent achievements have shown that reelin, a large extracellular glycoprotein, plays a crucial role in neuronal migration and localization during the development of neocortex and cerebellar cortex, thereby participating in the development of neuropsychiatric diseases. Reelin-induced neuronal migration requires triiodothyronine (T3) from the deiodination of thyroxine (T4) by fetal brain deiodinases. Previous studies have reported decreased reelin levels and abnormal gene expression, which are the same as the pathological alternations in reelin-induced neuropsychiatric disorders including schizophrenia and autism. Low T3 in the fetal brain due to hypothyroxinemia during pregnancy may be detrimental to neuronal migration, leading to neuropsychiatric disorders. In this review, we focus on the reelin expression between hypothyroidism and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yadi Su
- College of Stomatology, Chongqing Medical University, Chongqing, 401334, PR China
| | - Xiaoyu Yang
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Lu Yang
- College of Stomatology, Chongqing Medical University, Chongqing, 401334, PR China
| | - Xinjing Liu
- College of Public Health and Management, Chongqing Medical University, Chongqing, 401334, PR China
| | - Zhenghang She
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Youwen Zhang
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| |
Collapse
|
9
|
Kim HK, Song J. Hypothyroidism and Diabetes-Related Dementia: Focused on Neuronal Dysfunction, Insulin Resistance, and Dyslipidemia. Int J Mol Sci 2022; 23:ijms23062982. [PMID: 35328405 PMCID: PMC8952212 DOI: 10.3390/ijms23062982] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 01/27/2023] Open
Abstract
The incidence of dementia is steadily increasing worldwide. The risk factors for dementia are diverse, and include genetic background, environmental factors, sex differences, and vascular abnormalities. Among the subtypes of dementia, diabetes-related dementia is emerging as a complex type of dementia related to metabolic imbalance, due to the increase in the number of patients with metabolic syndrome and dementia worldwide. Thyroid hormones are considered metabolic regulatory hormones and affect various diseases, such as liver failure, obesity, and dementia. Thyroid dysregulation affects various cellular mechanisms and is linked to multiple disease pathologies. In particular, hypothyroidism is considered a critical cause for various neurological problems-such as metabolic disease, depressive symptoms, and dementia-in the central nervous system. Recent studies have demonstrated the relationship between hypothyroidism and brain insulin resistance and dyslipidemia, leading to diabetes-related dementia. Therefore, we reviewed the relationship between hypothyroidism and diabetes-related dementia, with a focus on major features of diabetes-related dementia such as insulin resistance, neuronal dysfunction, and dyslipidemia.
Collapse
Affiliation(s)
- Hee Kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, 264 Seoyangro, Hwasun 58128, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun 58128, Korea
- Correspondence: ; Tel.: +82-61-379-2706; Fax: +82-61-375-5834
| |
Collapse
|
10
|
Uchida K, Suzuki M. Congenital Hypothyroidism and Brain Development: Association With Other Psychiatric Disorders. Front Neurosci 2021; 15:772382. [PMID: 34955723 PMCID: PMC8695682 DOI: 10.3389/fnins.2021.772382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022] Open
Abstract
Thyroid hormones play an important role in brain development, and thyroid hormone insufficiency during the perinatal period results in severe developmental delays. Perinatal thyroid hormone deficiency is clinically known as congenital hypothyroidism, which is caused by dysgenesis of the thyroid gland or low iodine intake. If the disorder is not diagnosed or not treated early, the neuronal architecture is perturbed by thyroid hormone insufficiency, and neuropathological findings, such as abnormal synapse formation, defects in neuronal migration, and impairment of myelination, are observed in the brains of such patients. Furthermore, the expression of psychiatric disorder-related molecules, especially parvalbumin, is significantly decreased by thyroid hormone insufficiency during the perinatal period. Animal experiments using hypothyroidism models display decreased parvalbumin expression and abnormal brain architecture, and these experimental results show reproducibility and stability. These basic studies reinforce the results of epidemiological studies, suggesting the relevance of thyroid dysfunction in psychiatric disorders. In this review, we discuss the disruption of brain function associated with congenital hypothyroidism from the perspective of basic and clinical research.
Collapse
Affiliation(s)
- Katsuya Uchida
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Mao Suzuki
- Laboratory of Biomodeling, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
11
|
Timmerman BM, Mooney-Leber SM, Brummelte S. The effects of neonatal procedural pain and maternal isolation on hippocampal cell proliferation and reelin concentration in neonatal and adult male and female rats. Dev Psychobiol 2021; 63:e22212. [PMID: 34813104 DOI: 10.1002/dev.22212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Preterm births accounted for over 10% of all U.S. live births in 2019 and the rate is rising. Neonatal stressors, especially procedural pain, experienced by preterm infants in the neonatal intensive care unit (NICU) have been associated with neurodevelopmental impairments. Parental care can alleviate stress during stressful or painful procedures; however, infants in the NICU often receive reduced parental care compared with their peers. Animal studies suggest that decreased maternal care similarly impairs neurodevelopment but also influences the effects of neonatal pain. It is important to mimic both stressors in animal models of neonatal stress exposure. In this study, researchers investigated the individual and combined impact of neonatal pain and maternal isolation on reelin protein levels and cellular proliferation in the hippocampal dentate gyrus of 8 days old and adult rats. Exposure to either stressor individually, but not both, increased reelin levels in the dentate gyrus of adult females without significantly altering reelin levels in adult males. However, cell proliferation levels at either age were unaffected by the early-life stressors. These results suggest that each early-life stressor has a unique effect on markers of brain development and more research is needed to further investigate their distinct influences.
Collapse
Affiliation(s)
- Brian M Timmerman
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Sean M Mooney-Leber
- Department of Psychology, University of Wisconsin-Stevens Points, Stevens Point, Wisconsin, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA.,Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
12
|
Influence of Cerebral Vasodilation on Blood Reelin Levels in Growth Restricted Fetuses. Diagnostics (Basel) 2021; 11:diagnostics11061036. [PMID: 34199942 PMCID: PMC8228107 DOI: 10.3390/diagnostics11061036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 11/23/2022] Open
Abstract
Fetal growth restriction (FGR) is one of the most important obstetric pathologies. It is frequently caused by placental insufficiency. Previous studies have shown a relationship between FGR and impaired new-born neurodevelopment, although the molecular mechanisms involved in this association have not yet been completely clarified. Reelin is an extracellular matrix glycoprotein involved in development of neocortex, hippocampus, cerebellum and spinal cord. Reelin has been demonstrated to play a key role in regulating perinatal neurodevelopment and to contribute to the emergence and development of various psychiatric pathologies, and its levels are highly influenced by pathological conditions of hypoxia. The purpose of this article is to study whether reelin levels in new-borns vary as a function of severity of fetal growth restriction by gestational age and sex. We sub-grouped fetuses in: normal weight group (Group 1, n = 17), FGR group with normal umbilical artery Doppler and cerebral redistribution at middle cerebral artery Doppler (Group 2, n = 9), and FGR with abnormal umbilical artery Doppler (Group 3, n = 8). Our results show a significant association of elevated Reelin levels in FGR fetuses with cerebral blood redistribution compared to the normal weight group and the FGR with abnormal umbilical artery group. Future research should focus on further expanding the knowledge of the relationship of reelin and its regulated products with neurodevelopment impairment in new-borns with FGR and should include larger and more homogeneous samples and the combined use of different in vivo techniques in neonates with impaired growth during their different adaptive phases.
Collapse
|
13
|
Thyroid hormone insufficiency alters the expression of psychiatric disorder-related molecules in the hypothyroid mouse brain during the early postnatal period. Sci Rep 2021; 11:6723. [PMID: 33762687 PMCID: PMC7990947 DOI: 10.1038/s41598-021-86237-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/10/2021] [Indexed: 11/24/2022] Open
Abstract
The functional role of thyroid hormone (TH) in the cortex and hippocampus of mouse during neuronal development was investigated in this study. TH insufficiency showed a decrease in the expression of parvalbumin (PV) in the cortex and hippocampus of pups at postnatal day (PD) 14, while treatment with thyroxine from PD 0 to PD 14 ameliorated the PV loss. On the other hand, treatment with antithyroid agents in adulthood did not result in a decrease in the expression of PV in these areas. These results indicate the existence of a critical period of TH action during the early postnatal period. A decrease in MeCP2-positive neuronal nuclei was also observed in the cortical layers II–IV of the cerebral cortex. The brains were then stained with CUX1, a marker for cortical layers II–IV. In comparison with normal mice, CUX1 signals were decreased in the somatosensory cortex of the hypothyroid mice, and the total thickness of cortical layers II–IV of the mice was lower than that of normal mice. These results suggest that TH insufficiency during the perinatal period strongly and broadly affects neuronal development.
Collapse
|
14
|
Luongo C, Butruille L, Sébillot A, Le Blay K, Schwaninger M, Heuer H, Demeneix BA, Remaud S. Absence of Both Thyroid Hormone Transporters MCT8 and OATP1C1 Impairs Neural Stem Cell Fate in the Adult Mouse Subventricular Zone. Stem Cell Reports 2021; 16:337-353. [PMID: 33450189 PMCID: PMC7878696 DOI: 10.1016/j.stemcr.2020.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023] Open
Abstract
Adult neural stem cell (NSC) generation in vertebrate brains requires thyroid hormones (THs). How THs enter the NSC population is unknown, although TH availability determines proliferation and neuronal versus glial progenitor determination in murine subventricular zone (SVZ) NSCs. Mice display neurological signs of the severely disabling human disease, Allan-Herndon-Dudley syndrome, if they lack both MCT8 and OATP1C1 transporters, or MCT8 and deiodinase type 2. We analyzed the distribution of MCT8 and OATP1C1 in adult mouse SVZ. Both are strongly expressed in NSCs and at a lower level in neuronal cell precursors but not in oligodendrocyte progenitors. Next, we analyzed Mct8/Oatp1c1 double-knockout mice, where brain uptake of THs is strongly reduced. NSC proliferation and determination to neuronal fates were severely affected, but not SVZ-oligodendroglial progenitor generation. This work highlights how tight control of TH availability determines NSC function and glial-neuron cell-fate choice in adult brains.
Collapse
Affiliation(s)
- Cristina Luongo
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France
| | - Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France
| | - Anthony Sébillot
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France
| | - Karine Le Blay
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562 Lübeck, Germany
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, 45122 Essen, Germany
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, 75005 Paris, France.
| |
Collapse
|
15
|
O'Shaughnessy KL, Gilbert ME. Thyroid disrupting chemicals and developmental neurotoxicity - New tools and approaches to evaluate hormone action. Mol Cell Endocrinol 2020; 518:110663. [PMID: 31760043 PMCID: PMC8270644 DOI: 10.1016/j.mce.2019.110663] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 12/25/2022]
Abstract
It is well documented that thyroid hormone (TH) action is critical for normal brain development and is mediated by both nuclear and extranuclear pathways. Given this dependence, the impact of environmental endocrine disrupting chemicals that interfere with thyroid signaling is a major concern with direct implications for children's health. However, identifying thyroid disrupting chemicals in vivo is primarily reliant on serum thyroxine (T4) measurements within greater developmental and reproductive toxicity assessments. These studies do not examine known TH-dependent phenotypes in parallel, which complicates chemical evaluation. Additionally, there exist no recommendations regarding what degree of serum T4 dysfunction is adverse, and little consideration is given to quantifying TH action within the developing brain. This review summarizes current testing strategies in rodent models and discusses new approaches for evaluating the developmental neurotoxicity of thyroid disrupting chemicals. This includes assays to identify adverse cellular effects of the brain by both immunohistochemistry and gene expression, which would compliment serum T4 measures. While additional experiments are needed to test the full utility of these approaches, incorporation of these cellular and molecular assays could enhance chemical evaluation in the regulatory arena.
Collapse
Affiliation(s)
- Katherine L O'Shaughnessy
- United States Environmental Protection Agency, National Health and Environmental Effects Research Laboratory, Toxicity Assessment Division, Endocrine Toxicology Branch, Research Triangle Park, NC, 27711, USA.
| | - Mary E Gilbert
- United States Environmental Protection Agency, National Health and Environmental Effects Research Laboratory, Toxicity Assessment Division, Endocrine Toxicology Branch, Research Triangle Park, NC, 27711, USA.
| |
Collapse
|
16
|
Gilbert ME, O'Shaughnessy KL, Axelstad M. Regulation of Thyroid-disrupting Chemicals to Protect the Developing Brain. Endocrinology 2020; 161:bqaa106. [PMID: 32615585 PMCID: PMC8650774 DOI: 10.1210/endocr/bqaa106] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Synthetic chemicals with endocrine disrupting properties are pervasive in the environment and are present in the bodies of humans and wildlife. As thyroid hormones (THs) control normal brain development, and maternal hypothyroxinemia is associated with neurological impairments in children, chemicals that interfere with TH signaling are of considerable concern for children's health. However, identifying thyroid-disrupting chemicals (TDCs) in vivo is largely based on measuring serum tetraiodothyronine in rats, which may be inadequate to assess TDCs with disparate mechanisms of action and insufficient to evaluate the potential neurotoxicity of TDCs. In this review 2 neurodevelopmental processes that are dependent on TH action are highlighted, neuronal migration and maturation of gamma amino butyric acid-ergic interneurons. We discuss how interruption of these processes by TDCs may contribute to abnormal brain circuitry following developmental TH insufficiency. Finally, we identify issues in evaluating the developmental neurotoxicity of TDCs and the strengths and limitations of current approaches designed to regulate them. It is clear that an enhanced understanding of how THs affect brain development will lead to refined toxicity testing, reducing uncertainty and improving our ability to protect children's health.
Collapse
Affiliation(s)
- Mary E Gilbert
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Katherine L O'Shaughnessy
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Marta Axelstad
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
17
|
Spann MN, Cheslack-Postava K, Brown AS. The association of serologically documented maternal thyroid conditions during pregnancy with bipolar disorder in offspring. Bipolar Disord 2020; 22:621-628. [PMID: 31758834 DOI: 10.1111/bdi.12879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Higher rates of thyroid conditions are reported in individuals with bipolar disorder. However, no study to date has considered whether maternal thyroid conditions during pregnancy are associated with offspring risk of bipolar disorder, even though the fetus exclusively relies on maternal thyroid hormones through the early second trimester. We therefore examined the association between offspring bipolar disorder and serologically documented maternal thyroid conditions. METHODS The study was based on a nested case-control design that utilized data from the Child Health and Development Study, a birth cohort that enrolled pregnant women from 1959 to 1966. Eighty-five cases with DSM-IV-TR were ascertained and matched to controls (1:2) by date of birth, sex, gestational timing of the serum draws, and residence in Alameda County the first year receiving treatment. Archived prenatal maternal serum drawn during early to mid-gestation was used to measure two thyroid hormones, free thyroxine (fT4) and thyroid stimulating hormone (TSH). Subclinical and clinical hypothyroxinemia, hypothyroidism, and hyperthyroidism were determined based on standard methods. RESULTS Exposure to maternal hypothyroxinemia was associated with a five-fold increased risk of offspring bipolar disorder with psychotic features, but not without psychotic features. In stratified analysis, female offspring demonstrated increased risk for bipolar disorder with exposure to maternal hypothyroxinemia. No significant association was found between maternal hypothyroidism and offspring bipolar disorder. CONCLUSIONS These findings suggest that prenatal thyroid hormone deficiency, particularly a thyroid condition marked by low levels of thyroxine, may be an important developmental mechanism related to the risk of bipolar disorder with psychotic features.
Collapse
Affiliation(s)
- Marisa N Spann
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Keely Cheslack-Postava
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.,Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA
| | - Alan S Brown
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.,Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA.,Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
18
|
Abstract
Astrocytes, initially described as merely support cells, are now known as a heterogeneous population of cells actively involved in a variety of biological functions such as: neuronal migration and differentiation; regulation of cerebral blood flow; metabolic control of extracellular potassium concentration; and modulation of synapse formation and elimination; among others. Cerebellar glial cells have been shown to play a significant role in proliferation, differentiation, migration, and synaptogenesis. However, less evidence is available about the role of neuron-astrocyte interactions during cerebellar development and their impact on diseases of the cerebellum. In this review, we will focus on the mechanisms underlying cellular interactions, specifically neuron-astrocyte interactions, during cerebellar development, function, and disease. We will discuss how cerebellar glia, astrocytes, and Bergmann glia play a fundamental role in several steps of cerebellar development, such as granule cell migration, axonal growth, neuronal differentiation, and synapse formation, and in diseases associated with the cerebellum. We will focus on how astrocytes and thyroid hormones impact cerebellar development. Furthermore, we will provide evidence of how growth factors secreted by glial cells, such as epidermal growth factor and transforming growth factors, control cerebellar organogenesis. Finally, we will argue that glia are a key mediator of cerebellar development and that identification of molecules and pathways involved in neuron-glia interactions may contribute to a better understanding of cerebellar development and associated disorders.
Collapse
|
19
|
Ochsner SA, McKenna NJ. No Dataset Left Behind: Mechanistic Insights into Thyroid Receptor Signaling Through Transcriptomic Consensome Meta-Analysis. Thyroid 2020; 30:621-639. [PMID: 31910096 PMCID: PMC7187985 DOI: 10.1089/thy.2019.0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Discovery-scale omics datasets relevant to thyroid receptors (TRs) and their physiological and synthetic bioactive small-molecule ligands allow for genome-wide interrogation of TR-regulated genes. These datasets have considerable collective value as a reference resource to allow researchers to routinely generate hypotheses addressing the mechanisms underlying the cell biology and physiology of TR signaling in normal and disease states. Methods: Here, we searched the Gene Expression Omnibus database to identify a population of publicly archived transcriptomic datasets involving genetic or pharmacological manipulation of either TR isoform in a mouse tissue or cell line. After initial quality control, samples were organized into contrasts (experiments), and transcript differential expression values and associated measures of significance were generated and committed to a consensome (for consensus omics) meta-analysis pipeline. To gain insight into tissue-selective functions of TRs, we generated liver- and central nervous system (CNS)-specific consensomes and identified evidence for genes that were selectively responsive to TR signaling in each organ. Results: The TR transcriptomic consensome ranks genes based on the frequency of their significant differential expression over the entire group of experiments. The TR consensome assigns elevated rankings both to known TR-regulated genes and to genes previously uncharacterized as TR-regulated, which shed mechanistic light on known cellular and physiological roles of TR signaling in different organs. We identify evidence for unreported genomic targets of TR signaling for which it exhibits strikingly distinct regulatory preferences in the liver and CNS. Moreover, the intersection of the TR consensome with consensomes for other cellular receptors sheds light on transcripts potentially mediating crosstalk between TRs and these other signaling paradigms. Conclusions: The mouse TR datasets and consensomes are freely available in the Signaling Pathways Project website for hypothesis generation, data validation, and modeling of novel mechanisms of TR regulation of gene expression. Our results demonstrate the insights into the mechanistic basis of thyroid hormone action that can arise from an ongoing commitment on the part of the research community to the deposition of discovery-scale datasets.
Collapse
Affiliation(s)
- Scott A. Ochsner
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Neil J. McKenna
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Address correspondence to: Neil J. McKenna, PhD, The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
20
|
Talhada D, Santos CRA, Gonçalves I, Ruscher K. Thyroid Hormones in the Brain and Their Impact in Recovery Mechanisms After Stroke. Front Neurol 2019; 10:1103. [PMID: 31681160 PMCID: PMC6814074 DOI: 10.3389/fneur.2019.01103] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/02/2019] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormones are of fundamental importance for brain development and essential factors to warrant brain functions throughout life. Their actions are mediated by binding to specific intracellular and membranous receptors regulating genomic and non-genomic mechanisms in neurons and populations of glial cells, respectively. Among others, mechanisms include the regulation of neuronal plasticity processes, stimulation of angiogenesis and neurogenesis as well modulating the dynamics of cytoskeletal elements and intracellular transport processes. These mechanisms overlap with those that have been identified to enhance recovery of lost neurological functions during the first weeks and months after ischemic stroke. Stimulation of thyroid hormone signaling in the postischemic brain might be a promising therapeutic strategy to foster endogenous mechanisms of repair. Several studies have pointed to a significant association between thyroid hormones and outcome after stroke. With this review, we will provide an overview on functions of thyroid hormones in the healthy brain and summarize their mechanisms of action in the developing and adult brain. Also, we compile the major thyroid-modulated molecular pathways in the pathophysiology of ischemic stroke that can enhance recovery, highlighting thyroid hormones as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Daniela Talhada
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
- LUBIN Lab-Lunds Laboratorium för Neurokirurgisk Hjärnskadeforskning, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Cecília Reis Alves Santos
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
| | - Isabel Gonçalves
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- LUBIN Lab-Lunds Laboratorium för Neurokirurgisk Hjärnskadeforskning, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
21
|
López-Espíndola D, García-Aldea Á, Gómez de la Riva I, Rodríguez-García AM, Salvatore D, Visser TJ, Bernal J, Guadaño-Ferraz A. Thyroid hormone availability in the human fetal brain: novel entry pathways and role of radial glia. Brain Struct Funct 2019; 224:2103-2119. [PMID: 31165302 DOI: 10.1007/s00429-019-01896-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/27/2019] [Indexed: 01/13/2023]
Abstract
Thyroid hormones (TH) are crucial for brain development; their deficiency during neurodevelopment impairs neural cell differentiation and causes irreversible neurological alterations. Understanding TH action, and in particular the mechanisms regulating TH availability in the prenatal human brain is essential to design therapeutic strategies for neurological diseases due to impaired TH signaling during neurodevelopment. We aimed at the identification of cells involved in the regulation of TH availability in the human brain at fetal stages. To this end, we studied the distribution of the TH transporters monocarboxylate transporter 8 (MCT8) and organic anion-transporting polypeptide 1C1 (OATP1C1), as well as the TH-metabolizing enzymes types 2 and 3 deiodinases (DIO2 and DIO3). Paraffin-embedded human brain sections obtained from necropsies of thirteen fetuses from 14 to 38 gestational weeks were analyzed by immunohistochemistry and in situ hybridization. We found these proteins localized along radial glial cells, in brain barriers, in Cajal-Retzius cells, in migrating fibers of the brainstem and in some neurons and glial cells with particular and complex spatiotemporal patterns. Our findings point to an important role of radial glia in controlling TH delivery and metabolism and suggest two additional novel pathways for TH availability in the prenatal human brain: the outer, and the inner cerebrospinal fluid-brain barriers. Based on our data we propose a model of TH availability for neural cells in the human prenatal brain in which several cell types have the ability to autonomously control the required TH content.
Collapse
Affiliation(s)
- Daniela López-Espíndola
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain
- Escuela de Tecnología Médica and Centro de Investigaciones Biomédicas (CIB), Universidad de Valparaíso, Angamos 655, Reñaca, Viña del Mar, Chile
| | - Ángel García-Aldea
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain
| | | | | | - Domenico Salvatore
- Department of Public Health, University of Naples "Federico II", 80133, Naples, Italy
- CEINGE-Biotecnologie Avanzate s.c.a.r.l, 80145, Naples, Italy
| | - Theo J Visser
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Juan Bernal
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain.
- Center for Biomedical Research on Rare Diseases (CIBERER), U708, Madrid, Spain.
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain.
- Center for Biomedical Research on Rare Diseases (CIBERER), U708, Madrid, Spain.
| |
Collapse
|
22
|
A transient window of hypothyroidism alters neural progenitor cells and results in abnormal brain development. Sci Rep 2019; 9:4662. [PMID: 30874585 PMCID: PMC6420655 DOI: 10.1038/s41598-019-40249-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
Cortical heterotopias are clusters of ectopic neurons in the brain and are associated with neurodevelopmental disorders like epilepsy and learning disabilities. We have previously characterized the robust penetrance of a heterotopia in a rat model, induced by thyroid hormone (TH) disruption during gestation. However, the specific mechanism by which maternal TH insufficiency results in this birth defect remains unknown. Here we first determined the developmental window susceptible to endocrine disruption and describe a cellular mechanism responsible for heterotopia formation. We show that five days of maternal goitrogen treatment (10 ppm propylthiouracil) during the perinatal period (GD19-PN2) induces a periventricular heterotopia in 100% of the offspring. Beginning in the early postnatal brain, neurons begin to aggregate near the ventricles of treated animals. In parallel, transcriptional and architectural changes of this region were observed including decreased Sonic hedgehog (Shh) expression, abnormal cell adhesion, and altered radial glia morphology. As the ventricular epithelium is juxtaposed to two sources of brain THs, the cerebrospinal fluid and vasculature, this progenitor niche may be especially susceptible to TH disruption. This work highlights the spatiotemporal vulnerabilities of the developing brain and demonstrates that a transient period of TH perturbation is sufficient to induce a congenital abnormality.
Collapse
|
23
|
Stepien BK, Huttner WB. Transport, Metabolism, and Function of Thyroid Hormones in the Developing Mammalian Brain. Front Endocrinol (Lausanne) 2019; 10:209. [PMID: 31001205 PMCID: PMC6456649 DOI: 10.3389/fendo.2019.00209] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Ever since the discovery of thyroid hormone deficiency as the primary cause of cretinism in the second half of the 19th century, the crucial role of thyroid hormone (TH) signaling in embryonic brain development has been established. However, the biological understanding of TH function in brain formation is far from complete, despite advances in treating thyroid function deficiency disorders. The pleiotropic nature of TH action makes it difficult to identify and study discrete roles of TH in various aspect of embryogenesis, including neurogenesis and brain maturation. These challenges notwithstanding, enormous progress has been achieved in understanding TH production and its regulation, their conversions and routes of entry into the developing mammalian brain. The endocrine environment has to adjust when an embryo ceases to rely solely on maternal source of hormones as its own thyroid gland develops and starts to produce endogenous TH. A number of mechanisms are in place to secure the proper delivery and action of TH with placenta, blood-brain interface, and choroid plexus as barriers of entry that need to selectively transport and modify these hormones thus controlling their active levels. Additionally, target cells also possess mechanisms to import, modify and bind TH to further fine-tune their action. A complex picture of a tightly regulated network of transport proteins, modifying enzymes, and receptors has emerged from the past studies. TH have been implicated in multiple processes related to brain formation in mammals-neuronal progenitor proliferation, neuronal migration, functional maturation, and survival-with their exact roles changing over developmental time. Given the plethora of effects thyroid hormones exert on various cell types at different developmental periods, the precise spatiotemporal regulation of their action is of crucial importance. In this review we summarize the current knowledge about TH delivery, conversions, and function in the developing mammalian brain. We also discuss their potential role in vertebrate brain evolution and offer future directions for research aimed at elucidating TH signaling in nervous system development.
Collapse
|
24
|
O’Shaughnessy KL, Wood CR, Ford RL, Kosian PA, Hotchkiss MG, Degitz SJ, Gilbert ME. Thyroid Hormone Disruption in the Fetal and Neonatal Rat: Predictive Hormone Measures and Bioindicators of Hormone Action in the Developing Cortex. Toxicol Sci 2018; 166:163-179. [PMID: 30085217 PMCID: PMC6727986 DOI: 10.1093/toxsci/kfy190] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adverse neurodevelopmental consequences remain a primary concern when evaluating the effects of thyroid hormone (TH) disrupting chemicals. Though the developing brain is a known target of TH insufficiency, the relationship between THs in the serum and the central nervous system is not well characterized. To address this issue, dose response experiments were performed in pregnant rats using the goitrogen propylthiouracil (PTU) (dose range 0.1-10 ppm). THs were quantified in the serum and brain of offspring at gestational day 20 (GD20) and postnatal day 14 (PN14), two developmental stages included in OECD and EPA regulatory guideline/guidance studies. From the dose response data, the quantitative relationships between THs in the serum and brain were determined. Next, targeted gene expression analyses were performed in the fetal and neonatal cortex to test the hypothesis that TH action in the developing brain is linked to changes in TH concentrations within the tissue. Results show a significant reduction of T4/T3 in the serum and brain of the GD20 fetus in response to low doses of PTU; interestingly, very few genes were significantly different at any dose tested. In the PN14 pup significant reductions of T4/T3 in the serum and brain were also detected; however, twelve transcriptional targets were identified in the neonatal cortex that correlated well with reduced brain THs. These results show that serum T4 is a good predictor of brain THs, and offer several target genes that could serve as pragmatic readouts of T4/T3 dysfunction within the PN14 cortex.
Collapse
Affiliation(s)
- Katherine L. O’Shaughnessy
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830
| | - Carmen R. Wood
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Richard L. Ford
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830
| | - Patricia A. Kosian
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Duluth, Minnesota 55804
| | - Michelle G. Hotchkiss
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Sigmund J. Degitz
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Duluth, Minnesota 55804
| | - Mary E. Gilbert
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
25
|
Salazar P, Cisternas P, Martinez M, Inestrosa NC. Hypothyroidism and Cognitive Disorders during Development and Adulthood: Implications in the Central Nervous System. Mol Neurobiol 2018; 56:2952-2963. [PMID: 30073507 DOI: 10.1007/s12035-018-1270-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/19/2018] [Indexed: 02/06/2023]
Abstract
Thyroid hormones (THs) play a critical function in fundamental signaling of the body regulating process such as metabolism of glucose and lipids, cell maturation and proliferation, and neurogenesis, to name just a few. THs trigger biological effects both by directly affecting gene expression through the interaction with nuclear receptors (genomic effects) and by activating protein kinases and/or ion channels (short-term effects). For years, a close relationship between the THs hormones and the central nervous system (CNS) has been described, not only for neuronal cells but also for glial development and differentiation. A deficit in thyroid hormones triiodothyronine (T3) and thyroxine (T4) is observed in the hypothyroid condition, generated by a iodine deficiency or an autoimmune response of the body. In the hypothyroid condition, several cellular deregulation and alterations have been described in dendrite spine morphology, cell migration and proliferation, and impaired synaptic transmission in the hippocampus, among others. The aim of this review is to describe the role of the thyroid hormones with focus in brain function and neurodegenerative disorders.
Collapse
Affiliation(s)
- Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Milka Martinez
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,CARE UC Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Av. Bernardo O'Higgins 340, P. O. Box 114, -D, Santiago, Chile.
| |
Collapse
|
26
|
Gil-Ibañez P, García-García F, Dopazo J, Bernal J, Morte B. Global Transcriptome Analysis of Primary Cerebrocortical Cells: Identification of Genes Regulated by Triiodothyronine in Specific Cell Types. Cereb Cortex 2018; 27:706-717. [PMID: 26534908 DOI: 10.1093/cercor/bhv273] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Thyroid hormones, thyroxine, and triiodothyronine (T3) are crucial for cerebral cortex development acting through regulation of gene expression. To define the transcriptional program under T3 regulation, we have performed RNA-Seq of T3-treated and untreated primary mouse cerebrocortical cells. The expression of 1145 genes or 7.7% of expressed genes was changed upon T3 addition, of which 371 responded to T3 in the presence of cycloheximide indicating direct transcriptional regulation. The results were compared with available transcriptomic datasets of defined cellular types. In this way, we could identify targets of T3 within genes enriched in astrocytes and neurons, in specific layers including the subplate, and in specific neurons such as prepronociceptin, cholecystokinin, or cortistatin neurons. The subplate and the prepronociceptin neurons appear as potentially major targets of T3 action. T3 upregulates mostly genes related to cell membrane events, such as G-protein signaling, neurotransmission, and ion transport and downregulates genes involved in nuclear events associated with the M phase of cell cycle, such as chromosome organization and segregation. Remarkably, the transcriptomic changes induced by T3 sustain the transition from fetal to adult patterns of gene expression. The results allow defining in molecular terms the elusive role of thyroid hormones on neocortical development.
Collapse
Affiliation(s)
- Pilar Gil-Ibañez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Center for Biomedical Research on Rare Diseases, Madrid, Spain
| | - Francisco García-García
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Joaquín Dopazo
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.,Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.,Functional Genomics Node, INB at CIPF, Valencia, Spain
| | - Juan Bernal
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Center for Biomedical Research on Rare Diseases, Madrid, Spain
| | - Beatriz Morte
- Center for Biomedical Research on Rare Diseases, Madrid, Spain
| |
Collapse
|
27
|
Fanibunda SE, Desouza LA, Kapoor R, Vaidya RA, Vaidya VA. Thyroid Hormone Regulation of Adult Neurogenesis. VITAMINS AND HORMONES 2018; 106:211-251. [DOI: 10.1016/bs.vh.2017.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Raymaekers SR, Darras VM. Thyroid hormones and learning-associated neuroplasticity. Gen Comp Endocrinol 2017; 247:26-33. [PMID: 28390960 DOI: 10.1016/j.ygcen.2017.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 12/11/2022]
Abstract
Thyroid hormones (THs) are crucial for brain development and maturation in all vertebrates. Especially during pre- and perinatal development, disruption of TH signaling leads to a multitude of neurological deficits. Many animal models provided insight in the role of THs in brain development, but specific data on how they affect the brain's ability to learn and adapt depending on environmental stimuli are rather limited. In this review, we focus on a number of learning processes like spatial learning, fear conditioning, vocal learning and imprinting behavior and on how abnormal TH signaling during development shapes subsequent performance. It is clear from multiple studies that TH deprivation leads to defects in learning on all fronts, and interestingly, changes in local expression of the TH activator deiodinase type 2 seem to have an important role. Taking into account that THs are regulated in a very space-specific manner, there is thus increasing pressure to investigate more local TH regulators as potential factors involved in neuroplasticity. As these learning processes are also important for proper adult human functioning, further elucidating the role of THs in developmental neuroplasticity in various animal models is an important field for advancing both fundamental and applied knowledge on human brain function.
Collapse
Affiliation(s)
- Sander R Raymaekers
- Laboratory of Comparative Endocrinology, Biology Department, KU Leuven, Naamsestraat 61, 3000 Leuven, Belgium
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Biology Department, KU Leuven, Naamsestraat 61, 3000 Leuven, Belgium.
| |
Collapse
|
29
|
Genetic and Molecular Approaches to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050053. [PMID: 28475113 PMCID: PMC5447935 DOI: 10.3390/brainsci7050053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 11/17/2022] Open
Abstract
The migration of neuronal cells in the developing cerebral cortex is essential for proper development of the brain and brain networks. Disturbances in this process, due to genetic abnormalities or exogenous factors, leads to aberrant brain formation, brain network formation, and brain function. In the last decade, there has been extensive research in the field of neuronal migration. In this review, we describe different methods and approaches to assess and study neuronal migration in the developing cerebral cortex. First, we discuss several genetic methods, techniques and genetic models that have been used to study neuronal migration in the developing cortex. Second, we describe several molecular approaches to study aberrant neuronal migration in the cortex which can be used to elucidate the underlying mechanisms of neuronal migration. Finally, we describe model systems to investigate and assess the potential toxicity effect of prenatal exposure to environmental chemicals on proper brain formation and neuronal migration.
Collapse
|
30
|
Redman K, Ruffman T, Fitzgerald P, Skeaff S. Iodine Deficiency and the Brain: Effects and Mechanisms. Crit Rev Food Sci Nutr 2017; 56:2695-713. [PMID: 25880137 DOI: 10.1080/10408398.2014.922042] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Iodine is an essential micronutrient needed in human diets. As iodine is an integral component of thyroid hormone, it mediates the effects of thyroid hormone on brain development. Iodine deficiency is the most prevalent and preventable cause of mental impairment in the world. The exact mechanism through which iodine influences the brain is unclear, but is generally thought to begin with genetic expression. Many brain structures and systems appear to be affected with iodine deficiency, including areas such as the hippocampus, microstructures such as myelin, and neurotransmitters. The clearest evidence comes from the studies examining cognition in the cases of iodine deprivation or interventions involving iodine supplementation. Nevertheless, there are many inconsistencies and gaps in the literature of iodine deficiency, especially over the lifespan. This paper summarizes the literature on this topic, suggests a causal mechanism for iodine's effect on the brain, and indicates areas for the future research (e.g., using magnetic resonance imaging (MRI) and functional MRI to examine how iodine supplementation facilitates cognitive functioning).
Collapse
Affiliation(s)
- Kahla Redman
- a Department of Psychology , University of Otago , Dunedin , New Zealand
| | - Ted Ruffman
- a Department of Psychology , University of Otago , Dunedin , New Zealand
| | - Penelope Fitzgerald
- b Department of Human Nutrition , University of Otago , Dunedin , New Zealand
| | - Sheila Skeaff
- b Department of Human Nutrition , University of Otago , Dunedin , New Zealand
| |
Collapse
|
31
|
Moog NK, Entringer S, Heim C, Wadhwa PD, Kathmann N, Buss C. Influence of maternal thyroid hormones during gestation on fetal brain development. Neuroscience 2017; 342:68-100. [PMID: 26434624 PMCID: PMC4819012 DOI: 10.1016/j.neuroscience.2015.09.070] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/22/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Thyroid hormones (THs) play an obligatory role in many fundamental processes underlying brain development and maturation. The developing embryo/fetus is dependent on maternal supply of TH. The fetal thyroid gland does not commence TH synthesis until mid gestation, and the adverse consequences of severe maternal TH deficiency on offspring neurodevelopment are well established. Recent evidence suggests that even more moderate forms of maternal thyroid dysfunction, particularly during early gestation, may have a long-lasting influence on child cognitive development and risk of neurodevelopmental disorders. Moreover, these observed alterations appear to be largely irreversible after birth. It is, therefore, important to gain a better understanding of the role of maternal thyroid dysfunction on offspring neurodevelopment in terms of the nature, magnitude, time-specificity, and context-specificity of its effects. With respect to the issue of context specificity, it is possible that maternal stress and stress-related biological processes during pregnancy may modulate maternal thyroid function. The possibility of an interaction between the thyroid and stress systems in the context of fetal brain development has, however, not been addressed to date. We begin this review with a brief overview of TH biology during pregnancy and a summary of the literature on its effect on the developing brain. Next, we consider and discuss whether and how processes related to maternal stress and stress biology may interact with and modify the effects of maternal thyroid function on offspring brain development. We synthesize several research areas and identify important knowledge gaps that may warrant further study. The scientific and public health relevance of this review relates to achieving a better understanding of the timing, mechanisms and contexts of thyroid programing of brain development, with implications for early identification of risk, primary prevention and intervention.
Collapse
Affiliation(s)
- N K Moog
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany
| | - S Entringer
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA
| | - C Heim
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; Department of Biobehavioral Health, Pennsylvania State University, College of Health and Human Development, 219 Biobehavioral Health Building, University Park, PA 16802, USA
| | - P D Wadhwa
- University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA; Department of Psychiatry and Human Behavior, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Obstetrics and Gynecology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Epidemiology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA
| | - N Kathmann
- Department of Clinical Psychology, Humboldt-Universität zu Berlin, Rudower Chaussee 18, 12489 Berlin, Germany
| | - C Buss
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA.
| |
Collapse
|
32
|
Bernal J. Thyroid hormone regulated genes in cerebral cortex development. J Endocrinol 2017; 232:R83-R97. [PMID: 27852726 DOI: 10.1530/joe-16-0424] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
Abstract
The physiological and developmental effects of thyroid hormones are mainly due to the control of gene expression after interaction of T3 with the nuclear receptors. To understand the role of thyroid hormones on cerebral cortex development, knowledge of the genes regulated by T3 during specific stages of development is required. In our laboratory, we previously identified genes regulated by T3 in primary cerebrocortical cells in culture. By comparing these data with transcriptomics of purified cell types from the developing cortex, the cellular targets of T3 can be identified. In addition, many of the genes regulated transcriptionally by T3 have defined roles in cortex development, from which the role of T3 can be derived. This review analyzes the specific roles of T3-regulated genes in the different stages of cortex development within the physiological frame of the developmental changes of thyroid hormones and receptor concentrations in the human cerebral cortex during fetal development. These data indicate an increase in the sensitivity to T3 during the second trimester of fetal development. The main cellular targets of T3 appear to be the Cajal-Retzius and the subplate neurons. On the other hand, T3 regulates transcriptionally genes encoding extracellular matrix proteins, involved in cell migration and the control of diverse signaling pathways.
Collapse
Affiliation(s)
- Juan Bernal
- Instituto de Investigaciones BiomédicasConsejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid, and Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
33
|
Raymaekers SR, Verbeure W, Ter Haar SM, Cornil CA, Balthazart J, Darras VM. A dynamic, sex-specific expression pattern of genes regulating thyroid hormone action in the developing zebra finch song control system. Gen Comp Endocrinol 2017; 240:91-102. [PMID: 27693816 DOI: 10.1016/j.ygcen.2016.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 01/19/2023]
Abstract
The zebra finch (Taeniopygia guttata) song control system consists of several series of interconnected brain nuclei that undergo marked changes during ontogeny and sexual development, making it an excellent model to study developmental neuroplasticity. Despite the demonstrated influence of hormones such as sex steroids on this phenomenon, thyroid hormones (THs) - an important factor in neural development and maturation - have not been studied in this regard. We used in situ hybridization to compare the expression of TH transporters, deiodinases and receptors between both sexes during all phases of song development in male zebra finch. Comparisons were made in four song control nuclei: Area X, the lateral magnocellular nucleus of the anterior nidopallium (LMAN), HVC (used as proper name) and the robust nucleus of the arcopallium (RA). Most genes regulating TH action are expressed in these four nuclei at early stages of development. However, while general expression levels decrease with age, the activating enzyme deiodinase type 2 remains highly expressed in Area X, HVC and RA in males, but not in females, until 90days post-hatch (dph), which marks the end of sensorimotor learning. Furthermore, the L-type amino acid transporter 1 and TH receptor beta show elevated expression in male HVC and RA respectively compared to surrounding tissue until adulthood. Differences compared to surrounding tissue and between sexes for the other TH regulators were minor. These developmental changes are accompanied by a strong local increase in vascularization in the male RA between 20 and 30dph but not in Area X or HVC. Our results suggest that local regulation of TH signaling is an important factor in the development of the song control nuclei during the song learning phase and that TH activation by DIO2 is a key player in this process.
Collapse
Affiliation(s)
- Sander R Raymaekers
- Laboratory of Comparative Endocrinology, Biology Department, KU Leuven, Naamestraat 61, 3000 Leuven, Belgium
| | - Wout Verbeure
- Laboratory of Comparative Endocrinology, Biology Department, KU Leuven, Naamestraat 61, 3000 Leuven, Belgium
| | - Sita M Ter Haar
- Research Group in Behavioral Neuroendocrinology, GIGA Neurosciences, ULg, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Charlotte A Cornil
- Research Group in Behavioral Neuroendocrinology, GIGA Neurosciences, ULg, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Jacques Balthazart
- Research Group in Behavioral Neuroendocrinology, GIGA Neurosciences, ULg, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Biology Department, KU Leuven, Naamestraat 61, 3000 Leuven, Belgium.
| |
Collapse
|
34
|
Mata A, Urrea L, Vilches S, Llorens F, Thüne K, Espinosa JC, Andréoletti O, Sevillano AM, Torres JM, Requena JR, Zerr I, Ferrer I, Gavín R, Del Río JA. Reelin Expression in Creutzfeldt-Jakob Disease and Experimental Models of Transmissible Spongiform Encephalopathies. Mol Neurobiol 2016; 54:6412-6425. [PMID: 27726110 DOI: 10.1007/s12035-016-0177-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/28/2016] [Indexed: 12/22/2022]
Abstract
Reelin is an extracellular glycoprotein involved in key cellular processes in developing and adult nervous system, including regulation of neuronal migration, synapse formation, and plasticity. Most of these roles are mediated by the intracellular phosphorylation of disabled-1 (Dab1), an intracellular adaptor molecule, in turn mediated by binding Reelin to its receptors. Altered expression and glycosylation patterns of Reelin in cerebrospinal and cortical extracts have been reported in Alzheimer's disease. However, putative changes in Reelin are not described in natural prionopathies or experimental models of prion infection or toxicity. With this is mind, in the present study, we determined that Reelin protein and mRNA levels increased in CJD human samples and in mouse models of human prion disease in contrast to murine models of prion infection. However, changes in Reelin expression appeared only at late terminal stages of the disease, which prevent their use as an efficient diagnostic biomarker. In addition, increased Reelin in CJD and in in vitro models does not correlate with Dab1 phosphorylation, indicating failure in its intracellular signaling. Overall, these findings widen our understanding of the putative changes of Reelin in neurodegeneration.
Collapse
Affiliation(s)
- Agata Mata
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Laura Urrea
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Silvia Vilches
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Franc Llorens
- Department of Neurology, German Center for Neurodegenerative Diseases - DZNE, Universitätsmedizin Göttingen, Bonn, Germany
| | - Katrin Thüne
- Department of Neurology, German Center for Neurodegenerative Diseases - DZNE, Universitätsmedizin Göttingen, Bonn, Germany
| | - Juan-Carlos Espinosa
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Valdeolmos, Spain
| | - Olivier Andréoletti
- UMR INRA ENVT 1225, Interactions Hôtes Agents Pathogènes, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, 31076, Toulouse, France
| | - Alejandro M Sevillano
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782, Santiago de Compostela, Spain
- Department of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Valdeolmos, Spain
| | - Jesús Rodríguez Requena
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782, Santiago de Compostela, Spain
- Department of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Inga Zerr
- Department of Neurology, German Center for Neurodegenerative Diseases - DZNE, Universitätsmedizin Göttingen, Bonn, Germany
| | - Isidro Ferrer
- Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, 08028, Barcelona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
35
|
Huang CJ, Tseng CL, Chen HS, Garabwan C, Korovo S, Tang KT, Won JGS, Hsieh CH, Wang FF. Iodine Nutritional Status of School Children in Nauru 2015. Nutrients 2016; 8:nu8090520. [PMID: 27563920 PMCID: PMC5037507 DOI: 10.3390/nu8090520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 11/16/2022] Open
Abstract
Little is known about iodine nutritional status in island countries in the Pacific Ocean. The primary objective of this study was to report for the first time the iodine nutritional status of people in Nauru. In addition, sources of iodine nutrition (i.e., water and salt) were investigated. A school-based cross-sectional survey of children aged 6–12 years was conducted in three primary schools of Nauru. Urinary iodine concentration (UIC) was determined by spot urine samples. Available water and salt samples in Nauru were collected for the measurement of iodine content. A food frequency questionnaire was conducted. The median UIC was 142 μg/L, and 25.2% and 7.4% of the population had median UIC below 100 μg/L and 50 μg/L, respectively. Natural iodine-containing foods such as seaweeds and agar were rare. Iodine was undetectable in Nauruan tank water, filtered tap water, and raindrops. Of the analyzed salt products, five kinds were non-iodized, and three were iodized (iodine content: 15 ppm, 65 ppm, and 68 ppm, respectively). The results indicate that the iodine status in Nauruan school children is adequate. Iodized salt may serve as an important source of iodine nutrition in Nauru.
Collapse
Affiliation(s)
- Chun-Jui Huang
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Institute of Public Health, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Chi-Lung Tseng
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Division of Gastroenterology, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Harn-Shen Chen
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | | | | | - Kam-Tsun Tang
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Justin Ging-Shing Won
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Chang-Hsun Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan.
| | - Fan-Fen Wang
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Medicine, Yangming Branch, Taipei City Hospital, Taipei 11146, Taiwan.
| |
Collapse
|
36
|
Gyllenberg D, Sourander A, Surcel HM, Hinkka-Yli-Salomäki S, McKeague IW, Brown AS. Hypothyroxinemia During Gestation and Offspring Schizophrenia in a National Birth Cohort. Biol Psychiatry 2016; 79:962-70. [PMID: 26194598 PMCID: PMC4684794 DOI: 10.1016/j.biopsych.2015.06.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 05/07/2015] [Accepted: 06/11/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Evidence from animal and human studies indicates that thyroid hormone deficiency during early gestation alters brain development. As schizophrenia is associated with prenatal brain insults and premorbid cognitive deficits, we tested the a priori hypothesis that serologically defined maternal thyroid deficiency during early gestation to mid-gestation is associated with schizophrenia in offspring. METHODS The investigation is based on the Finnish Prenatal Study of Schizophrenia, a nested case-control study that included archived maternal sera from virtually all pregnancies since 1983 (N = >1 million). We identified all offspring in the cohort with a diagnosis of schizophrenia based on the national inpatient and outpatient register and matched them on sex, date of birth, and residence in Finland at time of onset of the case to comparison subjects (1:1) from the cohort. Maternal sera of 1010 case-control pairs were assessed for free thyroxine, and sera of 948 case-control pairs were assessed for thyroid-stimulating hormone. RESULTS Maternal hypothyroxinemia (free thyroxine ≤10th percentile, normal thyroid-stimulating hormone) was associated with an increased odds of schizophrenia (odds ratio = 1.75, 95% confidence interval = 1.22-2.50, p = .002). When adjusted for maternal psychiatric history, province of birth, and maternal smoking during pregnancy, the association remained significant (odds ratio = 1.70, 95% confidence interval = 1.13-2.55, p = .010). CONCLUSIONS In a large, national birth cohort, prospectively documented hypothyroxinemia during early gestation to mid-gestation was associated with increased odds of schizophrenia in offspring. This information can inform translational studies of maternal hypothyroxinemia examining molecular and cellular deviations relevant to schizophrenia.
Collapse
Affiliation(s)
- David Gyllenberg
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York State Psychiatric Institute, New York, New York; Department of Child Psychiatry, Faculty of Medicine, University of Turku.
| | - Andre Sourander
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York State Psychiatric Institute, New York, NY, United States, Department of Child Psychiatry, Faculty of Medicine, University of Turku, Turku, Finland, Department of Child Psychiatry, Turku University Hospital, Turku, Finland
| | | | | | - Ian W. McKeague
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Alan S. Brown
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York State Psychiatric Institute, New York, NY, United States, Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, United States
| |
Collapse
|
37
|
Günbey E, Karlı R, Gökosmanoğlu F, Düzgün B, Ayhan E, Atmaca H, Ünal R. Evaluation of olfactory function in adults with primary hypothyroidism. Int Forum Allergy Rhinol 2015; 5:919-22. [DOI: 10.1002/alr.21565] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/26/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Emre Günbey
- Department of Otorhinolaryngology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Rıfat Karlı
- Department of Otorhinolaryngology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Feyzi Gökosmanoğlu
- Department of Endocrinology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Berkan Düzgün
- Department of Otorhinolaryngology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Emre Ayhan
- Department of Otorhinolaryngology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Hulusi Atmaca
- Department of Endocrinology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| | - Recep Ünal
- Department of Otorhinolaryngology; Ondokuz Mayıs University School of Medicine; Samsun Turkey
| |
Collapse
|
38
|
Dezonne RS, Lima FRS, Trentin AG, Gomes FC. Thyroid hormone and astroglia: endocrine control of the neural environment. J Neuroendocrinol 2015; 27:435-45. [PMID: 25855519 DOI: 10.1111/jne.12283] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 02/03/2023]
Abstract
Thyroid hormones (THs) play key roles in brain development and function. The lack of THs during childhood is associated with the impairment of several neuronal connections, cognitive deficits and mental disorders. Several lines of evidence point to astrocytes as TH targets and as mediators of TH action in the central nervous system; however, the mechanisms underlying these events are still not completely known. In this review, we focus on advances in our understanding of the effects of THs on astroglial cells and the impact of these effects on neurone-astrocyte interactions. First, we discuss the signalling pathways involved in TH metabolism and the molecular mechanisms underlying TH receptor function. Then, we discuss data related to the effects of THs on astroglial cells, as well as studies regarding the generation of mutant TH receptor transgenic mice that have contributed to our understanding of TH function in brain development. We argue that astrocytes are key mediators of hormone actions on development of the cerebral cortex and cerebellum and that the identification of the molecules and pathways involved in these events might be important for determining the molecular-level basis of the neural deficits associated with endocrine diseases.
Collapse
Affiliation(s)
- R S Dezonne
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - F R S Lima
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - A G Trentin
- Departamento de Biologia Celular, Centro de Ciências Biológicas, Embriologia e Genética, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - F C Gomes
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
39
|
Treatment with Iodine in Pregnant Rats with Marginal Iodine Deficiency Improves Cell Migration in the Developing Brain of the Progeny. Mol Neurobiol 2015; 53:2212-21. [PMID: 25963726 DOI: 10.1007/s12035-015-9155-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/19/2015] [Indexed: 02/01/2023]
Abstract
Marginal iodine deficiency is a common health problem in pregnant women. Epidemiological and animal studies had shown that marginally maternal iodine deficiency could cause the mild changes of maternal thyroid function, eventually lead to a negative effect on neurodevelopment. But the underlying mechanisms responsible for the neurological impairment remain unclear. The aim of this study is to explore whether marginally maternal iodine deficiency could produce subtle changes in cell migration and cognitive function of offspring, and the optimal time of giving intervention to minimize the adverse effects. In the present study, we established a marginal iodine deficiency model, and iodine supplement was performed on pre-pregnancy (PP), G13 (gestation day 13), and postnatal day 0 (P0). Our data showed that there were changes in the cytoarchitecture and the percentage of bromodeoxyuridine (BrdU)-labeled cells in the cerebral cortex in marginal iodine deficiency rats. The Reelin expression was significantly lower, but Tenascin-C was higher in the cerebral cortex of marginal iodine deficiency group on P7 than the normal group, respectively. When iodine supplement, especially before G13 could reverse the abnormal expression of the two proteins involved in cell migration, which was consistent with the results of Morris Water Maze test. The three intervention groups had shorter escape latencies than the marginal iodine deficiency rats. The earlier that iodine is supplied, the better behavior performance would reach. Our findings suggested that iodine supplement in early stage of pregnancy could improve the cell migration of cerebral cortex and neurodevelopment of offspring.
Collapse
|
40
|
Moncayo R, Ortner K. Multifactorial determinants of cognition - Thyroid function is not the only one. BBA CLINICAL 2015; 3:289-98. [PMID: 26672993 PMCID: PMC4661586 DOI: 10.1016/j.bbacli.2015.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 12/31/2022]
Abstract
Background Since the 1960s hypothyroidism together with iodine deficiency have been considered to be a principal determinant of cognition development. Following iodine supplementation programs and improved treatment options for hypothyroidism this relation might not be valid in 2015. On the other hand neurosciences have added different inputs also related to cognition. Scope of review We will examine the characteristics of the original and current publications on thyroid function and cognition and also add some general determinants of intelligence and cognition. One central issue for us is the relation of stress to cognition knowing that both physical and psychological stress, are frequent elements in subjects with thyroid dysfunction. We have considered a special type of stress called pre-natal stress which can influence cognitive functions. Fear and anxiety can be intermingled requiring mechanisms of fear extinction. Major conclusions Recent studies have failed to show an influence of thyroid medication during pregnancy on intellectual development. Neuroscience offers a better explanation of cognition than hypothyroidism and iodine deficiency. Additional factors relevant to cognition are nutrition, infection, prenatal stress, and early life stress. In turn stress is related to low magnesium levels. Magnesium supplementation can correct both latent hypothyroidism and acquired mild cognitive deficits. General significance Cognition is a complex process that depends on many determinants and not only on thyroid function. Magnesium deficiency appears to be a basic mechanism for changes in thyroid function as well as of cognition. Untreated hypothyroidism, i.e. hypothyroxinemia, can influence IQ. Thyroxine administration to euthyroid pregnant women has no effect on cognition. The hippocampus and NMDA receptors play a central role in cognitive processes. Antenatal and early life stressors can influence cognition later in life. Stressors can lead to decreased levels of magnesium and demands supplementation.
Collapse
Affiliation(s)
- Roy Moncayo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Karina Ortner
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Kapoor R, Fanibunda SE, Desouza LA, Guha SK, Vaidya VA. Perspectives on thyroid hormone action in adult neurogenesis. J Neurochem 2015; 133:599-616. [DOI: 10.1111/jnc.13093] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/18/2015] [Accepted: 02/24/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Richa Kapoor
- Department of Biological Sciences; Tata Institute of Fundamental Research; Mumbai India
| | - Sashaina E. Fanibunda
- Department of Biological Sciences; Tata Institute of Fundamental Research; Mumbai India
| | - Lynette A. Desouza
- Department of Biological Sciences; Tata Institute of Fundamental Research; Mumbai India
| | - Suman K. Guha
- Department of Biological Sciences; Tata Institute of Fundamental Research; Mumbai India
| | - Vidita A. Vaidya
- Department of Biological Sciences; Tata Institute of Fundamental Research; Mumbai India
| |
Collapse
|
42
|
Berbel P, Navarro D, Román GC. An evo-devo approach to thyroid hormones in cerebral and cerebellar cortical development: etiological implications for autism. Front Endocrinol (Lausanne) 2014; 5:146. [PMID: 25250016 PMCID: PMC4158880 DOI: 10.3389/fendo.2014.00146] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
The morphological alterations of cortical lamination observed in mouse models of developmental hypothyroidism prompted the recognition that these experimental changes resembled the brain lesions of children with autism; this led to recent studies showing that maternal thyroid hormone deficiency increases fourfold the risk of autism spectrum disorders (ASD), offering for the first time the possibility of prevention of some forms of ASD. For ethical reasons, the role of thyroid hormones on brain development is currently studied using animal models, usually mice and rats. Although mammals have in common many basic developmental principles regulating brain development, as well as fundamental basic mechanisms that are controlled by similar metabolic pathway activated genes, there are also important differences. For instance, the rodent cerebral cortex is basically a primary cortex, whereas the primary sensory areas in humans account for a very small surface in the cerebral cortex when compared to the associative and frontal areas that are more extensive. Associative and frontal areas in humans are involved in many neurological disorders, including ASD, attention deficit-hyperactive disorder, and dyslexia, among others. Therefore, an evo-devo approach to neocortical evolution among species is fundamental to understand not only the role of thyroid hormones and environmental thyroid disruptors on evolution, development, and organization of the cerebral cortex in mammals but also their role in neurological diseases associated to thyroid dysfunction.
Collapse
Affiliation(s)
- Pere Berbel
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Daniela Navarro
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Gustavo C. Román
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Methodist Neurological Institute, Houston, TX, USA
| |
Collapse
|
43
|
Kupferman JV, Basu J, Russo MJ, Guevarra J, Cheung SK, Siegelbaum SA. Reelin signaling specifies the molecular identity of the pyramidal neuron distal dendritic compartment. Cell 2014; 158:1335-1347. [PMID: 25201528 DOI: 10.1016/j.cell.2014.07.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/27/2014] [Accepted: 07/17/2014] [Indexed: 12/17/2022]
Abstract
The apical dendrites of many neurons contain proximal and distal compartments that receive synaptic inputs from different brain regions. These compartments also contain distinct complements of ion channels that enable the differential processing of their respective synaptic inputs, making them functionally distinct. At present, the molecular mechanisms that specify dendritic compartments are not well understood. Here, we report that the extracellular matrix protein Reelin, acting through its downstream, intracellular Dab1 and Src family tyrosine kinase signaling cascade, is essential for establishing and maintaining the molecular identity of the distal dendritic compartment of cortical pyramidal neurons. We find that Reelin signaling is required for the striking enrichment of HCN1 and GIRK1 channels in the distal tuft dendrites of both hippocampal CA1 and neocortical layer 5 pyramidal neurons, where the channels actively filter inputs targeted to these dendritic domains.
Collapse
Affiliation(s)
- Justine V Kupferman
- Department of Biology, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA; Department of Neuroscience, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Jayeeta Basu
- Department of Neuroscience, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Marco J Russo
- Department of Neuroscience, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Jenieve Guevarra
- Department of Biology, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Stephanie K Cheung
- Department of Neuroscience, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA; Department of Pharmacology, Kavli Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA; Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA.
| |
Collapse
|
44
|
A temporary compendium of thyroid hormone target genes in brain. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:122-9. [PMID: 24882357 DOI: 10.1016/j.bbagrm.2014.05.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/21/2014] [Accepted: 05/22/2014] [Indexed: 11/21/2022]
Abstract
BACKGROUND Thyroid hormone controls a number of developmental and physiological processes in the brain by directly acting on gene expression. Transcriptome analyses in rodent identified a number of thyroid hormone regulated genes in several brain areas at different stages. Genome wide analysis of chromatin occupancy in a neural cell line also identified a subset of genes which transcription is likely to be directly regulated by thyroid hormone receptors in neurons. However, the abundance of these data and apparent discrepancies between studies brought some confusion. RESULTS We present here a meta-analysis of available data to identify recurrent themes in thyroid hormone action in brain cells. This provides a curated list of 734 regulated genes in rodent brain, and highlights a small number of likely direct target genes. Some of these genes are also regulated in amphibians during metamorphosis. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
|
45
|
Andersen SL, Laurberg P, Wu CS, Olsen J. Attention deficit hyperactivity disorder and autism spectrum disorder in children born to mothers with thyroid dysfunction: a Danish nationwide cohort study. BJOG 2014; 121:1365-74. [PMID: 24605987 DOI: 10.1111/1471-0528.12681] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the association between maternal hyper- and hypothyroidism and the risk of attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) in the child. DESIGN A population-based cohort study. SETTING Singletons liveborn in Denmark between 1991 and 2004. POPULATION A total of 857 014 singletons alive and living in Denmark at the age of 3 years. METHODS Information on the diagnosis and/or treatment of maternal thyroid disease and the neurodevelopmental disorders ADHD and ASD in the child was obtained from Danish nationwide registers. The Cox proportional hazards model was used to estimate the hazard ratio (HR) with 95% confidence interval (95% CI) for risk of ADHD and ASD in children born to mothers with thyroid dysfunction, adjusting for potential confounding factors. MAIN OUTCOME MEASURES ADHD and ASD in the child. RESULTS Altogether, 30,295 singletons (3.5%) were born to mothers with thyroid dysfunction. Maternal hyperthyroidism diagnosed and treated for the first time after the birth of the child increased the risk of ADHD in the child (adjusted HR 1.23; 95% CI 1.05-1.44), whereas hypothyroidism increased the risk of ASD (adjusted HR 1.34; 95% CI 1.14-1.59). No significant association was seen for maternal diagnosis and treatment prior to the birth of the child. CONCLUSIONS Children born to mothers diagnosed and treated for the first time for thyroid dysfunction after their birth may have been exposed to abnormal levels of maternal thyroid hormone already present during the pregnancy, and this untreated condition could increase the risk of specific neurodevelopmental disorders in the child.
Collapse
Affiliation(s)
- S L Andersen
- Departments of Clinical Medicine and Endocrinology, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | | | | | | |
Collapse
|
46
|
Bianco AC, Anderson G, Forrest D, Galton VA, Gereben B, Kim BW, Kopp PA, Liao XH, Obregon MJ, Peeters RP, Refetoff S, Sharlin DS, Simonides WS, Weiss RE, Williams GR. American Thyroid Association Guide to investigating thyroid hormone economy and action in rodent and cell models. Thyroid 2014; 24:88-168. [PMID: 24001133 PMCID: PMC3887458 DOI: 10.1089/thy.2013.0109] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND An in-depth understanding of the fundamental principles that regulate thyroid hormone homeostasis is critical for the development of new diagnostic and treatment approaches for patients with thyroid disease. SUMMARY Important clinical practices in use today for the treatment of patients with hypothyroidism, hyperthyroidism, or thyroid cancer are the result of laboratory discoveries made by scientists investigating the most basic aspects of thyroid structure and molecular biology. In this document, a panel of experts commissioned by the American Thyroid Association makes a series of recommendations related to the study of thyroid hormone economy and action. These recommendations are intended to promote standardization of study design, which should in turn increase the comparability and reproducibility of experimental findings. CONCLUSIONS It is expected that adherence to these recommendations by investigators in the field will facilitate progress towards a better understanding of the thyroid gland and thyroid hormone dependent processes.
Collapse
Affiliation(s)
- Antonio C. Bianco
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Grant Anderson
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota Duluth, Duluth, Minnesota
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Valerie Anne Galton
- Department of Physiology and Neurobiology, Dartmouth Medical School, Lebanon, New Hampshire
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Brian W. Kim
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter A. Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Hui Liao
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Maria Jesus Obregon
- Institute of Biomedical Investigation (IIB), Spanish National Research Council (CSIC) and Autonomous University of Madrid, Madrid, Spain
| | - Robin P. Peeters
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - David S. Sharlin
- Department of Biological Sciences, Minnesota State University, Mankato, Minnesota
| | - Warner S. Simonides
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Roy E. Weiss
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Graham R. Williams
- Department of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| |
Collapse
|
47
|
Iodine Deficiency in Egyptian Autistic Children and Their Mothers: Relation to Disease Severity. Arch Med Res 2013; 44:555-61. [DOI: 10.1016/j.arcmed.2013.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/23/2013] [Indexed: 11/19/2022]
|
48
|
Román GC, Ghassabian A, Bongers‐Schokking JJ, Jaddoe VWV, Hofman A, Rijke YB, Verhulst FC, Tiemeier H. Association of gestational maternal hypothyroxinemia and increased autism risk. Ann Neurol 2013; 74:733-42. [DOI: 10.1002/ana.23976] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/21/2013] [Accepted: 07/03/2013] [Indexed: 01/18/2023]
Affiliation(s)
- Gustavo C. Román
- Methodist Neurological InstituteHouston TX
- Department of NeurologyWeill Cornell Medical CollegeNew York NY
| | - Akhgar Ghassabian
- Generation R Study GroupRotterdam the Netherlands
- Department of Child and Adolescent PsychiatryErasmus University Medical CenterRotterdam the Netherlands
| | - Jacoba J. Bongers‐Schokking
- Department of EpidemiologyErasmus University Medical CenterRotterdam the Netherlands
- Department of EndocrinologyErasmus University Medical CenterRotterdam the Netherlands
| | - Vincent W. V. Jaddoe
- Generation R Study GroupRotterdam the Netherlands
- Department of EpidemiologyErasmus University Medical CenterRotterdam the Netherlands
- Department of PediatricsErasmus University Medical CenterRotterdam the Netherlands
| | - Albert Hofman
- Generation R Study GroupRotterdam the Netherlands
- Department of EpidemiologyErasmus University Medical CenterRotterdam the Netherlands
| | - Yolanda B. Rijke
- Department of Internal MedicineErasmus University Medical CenterRotterdam the Netherlands
- Department of Clinical ChemistryErasmus University Medical CenterRotterdam the Netherlands
| | - Frank C. Verhulst
- Department of Child and Adolescent PsychiatryErasmus University Medical CenterRotterdam the Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent PsychiatryErasmus University Medical CenterRotterdam the Netherlands
- Department of EpidemiologyErasmus University Medical CenterRotterdam the Netherlands
- Department of PsychiatryErasmus University Medical CenterRotterdam the Netherlands
| |
Collapse
|
49
|
Abstract
About three-fourths of the total world population live in the tropics but consume only 6% of worldwide food production and contribute 15% of the world's net revenue explaining the short life expectancy, high infantile mortality, and poor daily caloric intake; moreover, lack of clean drinking water and deficient sanitation promote water-borne infections, diarrhea, and risk of malabsorption that contribute to the prevalence of malnutrition in the tropics. One-third of the world's population consumes insufficient iodine increasing the risk for mental retardation and deafness due to maternal hypothyroidism. The main nutritional syndromes comprise protein-energy malnutrition (marasmus and kwashiorkor); nutritional neuropathies, myelopathies and neuromyelopathies, as well as specific deficiencies of vitamins and micronutrients including iodine, iron, zinc, and selenium.
Collapse
Affiliation(s)
- Gustavo C Román
- Department of Neurology, Weill Cornell Medical College and Nantz National Alzheimer Center, Methodist Neurological Institute, Houston, TX, USA.
| |
Collapse
|
50
|
Tousson E, Beltagy DM, El-Gerbed MS, Gazia MA, Akela MA. The ameliorating role of folic acid in rat hippocampus after propylthiouracil-induced hypothyroidism. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.biomag.2012.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|