1
|
Koch K, Schlüppmann K, Hüsken S, Stark LM, Förster N, Masjosthusmann S, Klose J, Dönmez A, Fritsche E. Nuclear hormone receptors control fundamental processes of human fetal neurodevelopment: Basis for endocrine disruption assessment. ENVIRONMENT INTERNATIONAL 2025; 198:109400. [PMID: 40147140 DOI: 10.1016/j.envint.2025.109400] [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: 11/24/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Despite growing awareness of endocrine disrupting chemicals (EDCs), knowledge gaps remain regarding their effects on human brain development. EDC risk assessment focuses primarily on EATS modalities (estrogens, androgens, thyroid hormones, and steroidogenesis), overlooking the broader range of hormone receptors expressed in the developing brain. This limits the evaluation of chemicals for their potential to cause endocrine disruption-mediated developmental neurotoxicity (ED-DNT). The Neurosphere Assay, an in vitro test method for developmental neurotoxicity (DNT) evaluation, is an integral component of the DNT in vitro testing battery, which has been used to screen a broad domain of environmental chemicals. Here, we define the endocrine-related applicability domain of the Neurosphere Assay by assessing the impact and specificity of 14 hormone receptors on seven key neurodevelopmental processes (KNDPs), neural progenitor cell (NPC) proliferation, migration of radial glia, neurons, and oligodendrocytes, neurite outgrowth, and differentiation of neurons and oligodendrocytes. Comparative analyses in human and rat NPCs of both sexes revealed species- and sex-specific responses. Mechanistic insights were obtained through RNA sequencing and agonist/antagonist co-exposures. Most receptor agonists modulated KNDPs at concentrations in the range of physiologically relevant hormone concentrations. Phenotypic effects induced by glucocorticoid receptor (GR), liver X receptor (LXR), peroxisome proliferator-activated receptor beta/delta (PPARβδ), retinoic acid receptor (RAR) and retinoid X receptor (RXR) activation were counteracted by receptor antagonists, confirming specificity. Transcriptomics highlighted receptor crosstalk and the involvement of conserved developmental pathways (e.g. Notch and Wnt). Species comparisons identified limited concordance in hormone receptor-regulated KNDPs between human and rat NPCs. This study presents novel findings on cellular and molecular hormone actions in human fetal NPCs, highlights major species differences, and illustrates the Neurosphere Assay's relevance for detecting endocrine MoAs, supporting its application in human-based ED-DNT risk assessment.
Collapse
Affiliation(s)
- Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany.
| | - Kevin Schlüppmann
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Saskia Hüsken
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Louisa Merit Stark
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Nils Förster
- Bioinformatics Group, Faculty for Biology and Biotechnology, Ruhr-University Bochum, Germany; Center for Protein Diagnostics (ProDi), Ruhr-University Bochum, Bochum, Germany
| | | | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany
| | - Arif Dönmez
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany; Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany; SCAHT - Swiss Centre for Applied Human Toxicology, Basel, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
2
|
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
|
3
|
Wang B, Huang J, Chen L. Association between depression and anxiety disorders with euthyroid Hashimoto's thyroiditis: A systematic review and meta-analysis. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2024; 20:100279. [PMID: 39717384 PMCID: PMC11665666 DOI: 10.1016/j.cpnec.2024.100279] [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: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/25/2024] Open
Abstract
Background Hashimoto's thyroiditis (HT) affects up to 10 % of the population and is a common cause of hypothyroidism, which can lead to depression and anxiety. However, it remains unclear whether HT directly causes these conditions or if they arise due to HT-induced hypothyroidism. The present review aims to offer meta-analytic insights into the relationship between depression and anxiety in patients with euthyroid HT. Methods A comprehensive search was conducted in PubMed, Embase, Web of Science, Cochrane Library, CNKI, Wanfang Data, SinoMed, and VIP from their inception through May 2024. Case-control or cross-sectional studies examining the association between euthyroid HT and either depression, anxiety disorders, or both were included. Results For depression, 1365 patients (694 HT vs. 671 controls) from 11 articles were analyzed; for anxiety, 1009 patients (516 HT vs. 493 controls) from 8 articles were included. HT patients had 2.5 times higher odds of anxiety disorders (OR = 2.52, 95 % CI: 1.66-3.82). The Beck Depression Inventory showed a WMD of 4.26 (95 % CI: 1.28-7.24) and the Beck Anxiety Inventory a WMD of 5.10 (95 % CI: 1.55-8.66). Limitation The findings should be interpreted cautiously due to heterogeneity, potential publication bias, and variability in assessment tools, which may limit generalizability. Conclusions Euthyroid HT patients exhibit a higher prevalence of anxiety disorders compared to healthy control groups, and more susceptible to anxiety and depression symptoms based on the Beck Inventory. Thyroid antibodies themselves are also associated with depression and anxiety disorder.
Collapse
Affiliation(s)
- Bo Wang
- Department of Paediatric Surgery, Tianjin Medical University General Hospital Tianjin, China
| | - Jie Huang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Chen
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Khan K, Zahid M, Ali N, Attaullah S, Ullah M, Khan K, Muhammad I, Abusharha A, Aschner M, Khan H. STAT3 single-nucleotide variants in autoimmune thyroid disease in the Pakhtun population of Khyber Pakhtunkhwa, Pakistan. GENE REPORTS 2024; 36:101950. [PMID: 39385969 PMCID: PMC11463997 DOI: 10.1016/j.genrep.2024.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The current study was conducted to assess the relationship between the STAT3 gene variants rs744166 and rs2293152 and autoimmune thyroid disorder in the Pakhtun population of the province, of Khyber Pakhtunkhwa, Pakistan. Blood was collected from 100 healthy individuals and 400 thyroid-disordered patients. Of these, one hundred were diagnosed with Hashimoto's thyroiditis (HT), while 32 were confirmed as Grave's disease (GD) patients. T3, T4, and TSH serum levels were checked to diagnose thyroid disorders. The blood was analyzed for anti-thyroid peroxidase antibodies (Anti-TPOAb) (AESKULISA- ATPO - elisa kit), (Germany), and thyroid stimulating hormone receptor antibodies (TSHRAb), TSHR Ab elisa kit (Diametra Italy), respectively. PCR was used to amplify the targeted STAT3 gene polymorphisms from rs744166 (301 bp) and rs2293152 (365 bp) sequences and then digested by specific restriction endonucleases (AluI) and AciI respectively. The disease displayed a female predominance. The genotype TC and CC of rs744166 showed a significant relationship with Grave's disease (p = 0.002, OR = 0.28, 95 % CI = 0.11-0.77) in patients. The C allele contributed significantly to the disease in GD patients. The SNP rs2293152 significantly differed between GD patients and control (p = 0.032, OR = 0.29, 95 % CI = 0.09-0.86). Similarly, the G and C alleles showed a significant (p = 0.02) difference between GD patients and the control. No significant association was found for both SNPs in Hashimoto's thyroiditis disease. It is concluded that the STAT3 gene (rs744166 and rs2293152) was found to have a potential role in autoimmunity in GD patients. Still, it needs further studies with larger sample sizes in the Pakhtun population to understand this relationship.
Collapse
Affiliation(s)
- Khayyam Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Muhammad Zahid
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Niaz Ali
- Institute of Basic Medical Sciences Khyber Medical University, 25120 Peshawar, Pakistan
| | - Sobia Attaullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Mujeeb Ullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Khalid Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Ijaz Muhammad
- Department of Zoology, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| | - Ali Abusharha
- Optometry Department, Applied Medical Sciences Collage, King Saud University, P. O. Box 145111, Riyadh, Saudi Arabia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States of America
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| |
Collapse
|
5
|
Ma Q, Yang Z, Yang C, Lin M, Gong M, Deng P, He M, Lu Y, Zhang K, Pi H, Qu M, Yu Z, Zhou Z, Chen C. A single-cell transcriptomic landscape of cadmium-hindered brain development in mice. Commun Biol 2024; 7:997. [PMID: 39147853 PMCID: PMC11327346 DOI: 10.1038/s42003-024-06685-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
The effects of neurotoxicant cadmium (Cd) exposure on brain development have not been well elucidated. To investigate this, we have herein subjected pregnant mice to low-dose Cd throughout gestation. Using single-cell RNA sequencing (scRNA-seq), we explored the cellular responses in the embryonic brain to Cd exposure, and identified 18 distinct cell subpopulations that exhibited varied responses to Cd. Typically, Cd exposure impeded the development and maturation of cells in the brain, especially progenitor cells such as neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). It also caused significant cell subpopulation shifts in almost all the types of cells in the brain. Additionally, Cd exposure reduced the dendritic sophistication of cortical neurons in the offspring. Importantly, these changes led to aberrant Ca2+ activity in the cortex and neural behavior changes in mature offspring. These data contribute to our understanding of the effects and mechanisms of Cd exposure on brain development and highlight the importance of controlling environmental neurotoxicant exposure at the population level.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiqi Yang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chuanyan Yang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Lin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mingyue Gong
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Kuan Zhang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mingyue Qu
- The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
6
|
Salloum-Asfar S, Shin KC, Taha RZ, Khattak S, Park Y, Abdulla SA. The Potential Role of Thyroid Hormone Therapy in Neural Progenitor Cell Differentiation and Its Impact on Neurodevelopmental Disorders. Mol Neurobiol 2024; 61:3330-3342. [PMID: 37991699 PMCID: PMC11087352 DOI: 10.1007/s12035-023-03751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/28/2023] [Indexed: 11/23/2023]
Abstract
Thyroid hormone (T3) plays a vital role in brain development and its dysregulation can impact behavior, nervous system function, and cognitive development. Large case-cohort studies have associated abnormal maternal T3 during early pregnancy to epilepsy, autism, and attention deficit hyperactivity disorder (ADHD) in children. Recent experimental findings have also shown T3's influence on the fate of neural precursor cells and raise the question of its convergence with embryonic neural progenitors. Our objective was to investigate how T3 treatment affects neuronal development and functionality at the cellular level. In vitro experiments using neural precursor cells (NPCs) measured cell growth and numbers after exposure to varying T3 concentrations. Time points included week 0 (W0) representing NPCs treated with 100 nM T3 for 5 days, and differentiated cortical neurons assessed at weeks 3 (W3), 6 (W6), and 8 (W8). Techniques such as single-cell calcium imaging and whole-cell patch clamp were utilized to evaluate neuronal activity and function. IHC staining detected mature neuron markers, and RNA sequencing enabled molecular profiling. W6 and W8 neurons exhibited higher action potential frequencies, with W6 showing increased peak amplitudes and shortened inter-spike intervals by 50%, indicating enhanced activity. Transcriptomic analysis revealed that W6 T3-treated neurons formed a distinct cluster, suggesting accelerated maturation. Comparison with the whole transcriptome further unveiled a correlation between W6 neurons treated with T3 and neuronal regulatory elements associated with autism and ADHD. These findings provide insights into T3's impact on neuronal development and potential mechanisms of T3 dysregulation and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| | - Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Rowaida Z Taha
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Shahryar Khattak
- BESE and KAUST Smart-Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Sara A Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
7
|
Fame RM, Ali I, Lehtinen MK, Kanarek N, Petrova B. Optimized Mass Spectrometry Detection of Thyroid Hormones and Polar Metabolites in Rodent Cerebrospinal Fluid. Metabolites 2024; 14:79. [PMID: 38392972 PMCID: PMC10890085 DOI: 10.3390/metabo14020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
Thyroid hormones (TH) are required for brain development and function. Cerebrospinal fluid (CSF), which bathes the brain and spinal cord, contains TH as free hormones or as bound to transthyretin (TTR). Tight TH level regulation in the central nervous system is essential for developmental gene expression, which governs neurogenesis, myelination, and synaptogenesis. This integrated function of TH highlights the importance of developing precise and reliable methods for assessing TH levels in CSF. We report an optimized liquid chromatography-mass spectrometry (LC-MS)-based method to measure TH in rodent CSF and serum, applicable to both fresh and frozen samples. Using this new method, we find distinct differences in CSF TH in pregnant dams vs. non-pregnant adults and in embryonic vs. adult CSF. Further, targeted LC-MS metabolic profiling uncovers distinct central carbon metabolism in the CSF of these populations. TH detection and metabolite profiling of related metabolic pathways open new avenues of rigorous research into CSF TH and will inform future studies on metabolic alterations in CSF during normal development.
Collapse
Affiliation(s)
- Ryann M. Fame
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Ilhan Ali
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Maria K. Lehtinen
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Boryana Petrova
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Fame RM, Ali I, Lehtinen MK, Kanarek N, Petrova B. Optimized Mass Spectrometry Detection of Thyroid Hormones and Polar Metabolites in Rodent Cerebrospinal Fluid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570731. [PMID: 38116027 PMCID: PMC10729774 DOI: 10.1101/2023.12.07.570731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
BACKGROUND Thyroid hormones (TH) are required for brain development and function. Cerebrospinal fluid (CSF), which bathes the brain and spinal cord, contains TH as free or transthyretin (TTR)-bound. Tight thyroid hormone level regulation in the central nervous system is essential for developmental gene expression that governs neurogenesis, myelination, and synaptogenesis. This integrated function of TH highlights the importance of developing precise and reliable methods for assessing TH levels in CSF. METHODS we report an optimized LC-MS based method to measure thyroid hormones in rodent CSF and serum, applicable to both fresh and frozen samples. RESULTS We find distinct differences in CSF thyroid hormone in pregnant dams vs. non-pregnant adults and in embryonic vs. adult CSF. Further, targeted LC-MS metabolic profiling uncovers distinct central carbon metabolism in the CSF of these populations. CONCLUSIONS TH detection and metabolite profiling of related metabolic pathways open new avenues of rigorous research into CSF thyroid hormone and will inform future studies on metabolic alterations in CSF during normal development.
Collapse
|
9
|
Hwang E, Doolittle WKL, Zhu YJ, Zhu X, Zhao L, Yu Y, Cheng SY. Thyroid hormone receptor α1: a novel regulator of thyroid cancer cell differentiation. Oncogene 2023; 42:3075-3086. [PMID: 37634007 DOI: 10.1038/s41388-023-02815-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
Thyroid hormone receptor α1 (TRα1) mediates the genomic actions of thyroid hormone (T3). The biology of TRα1 in growth and development has been well studied, but the functional role of TRα1 in cancers remains to be elucidated. Analysis of the human thyroid cancer database of The Cancer Genome Atlas (TCGA) showed that THRA gene expression is lost in highly dedifferentiated anaplastic thyroid cancer (ATC). We, therefore, explored the effects of TRα1 on the progression of ATC. We stably expressed TRα1 in two human ATC cell lines, THJ-11T (11T-TRα1 #2, #7, and #8) and THJ-16T (16T-TRα1 #3, #4, and #8) cells. We found that the expressed TRα1 inhibited ATC cell proliferation and induced apoptosis. TCGA data showed that THRA gene expression was best correlated with the paired box gene 8 (PAX8). Consistently, we found that the PAX8 expression was barely detectable in parental 11T and 16T cells. However, PAX8 gene expression was elevated in 11T- and 16T-TRα1-expressing cells at the mRNA and protein levels. Using various molecular analyses, we found that TRα1 directly regulated the expression of the PAX8 gene. Single-cell transcriptomic analyses (scRNA-seq) demonstrated that TRα1 functions as a transcription factor through multiple signaling pathways to suppress tumor growth. Importantly, scRNA-seq analysis showed that TRα1-induced PAX8, via its transcription program, shifts the cell landscape of ATC toward a differentiated state. The present studies suggest that TRα1 is a newly identified regulator of thyroid differentiation and could be considered as a potential therapeutic target to improve the outcome of ATC patients.
Collapse
Affiliation(s)
- Eunmi Hwang
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Woo Kyung Lee Doolittle
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yuelin Jack Zhu
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuguang Zhu
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Li Zhao
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yanlin Yu
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
10
|
Di Criscio M, Lodahl JE, Stamatakis A, Kitraki E, Bakoyiannis I, Repouskou A, Bornehag CG, Gennings C, Lupu D, Rüegg J. A human-relevant mixture of endocrine disrupting chemicals induces changes in hippocampal DNA methylation correlating with hyperactive behavior in male mice. CHEMOSPHERE 2023; 313:137633. [PMID: 36565761 DOI: 10.1016/j.chemosphere.2022.137633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Humans are ubiquitously exposed to endocrine disrupting chemicals (EDCs), substances that interfere with endogenous hormonal signaling. Exposure during early development is of particular concern due to the programming role of hormones during this period. A previous epidemiological study has shown association between prenatal co-exposure to 8 EDCs (Mixture N1) and language delay in children, suggesting an effect of this mixture on neurodevelopment. Furthermore, in utero exposure to Mixture N1 altered gene expression and behavior in adult mice. In this study, we investigated whether epigenetic mechanisms could underlie the long term effects of Mixture N1 on gene expression and behavior. To this end, we analyzed DNA methylation at regulatory regions of genes whose expression was affected by Mixture N1 in the hippocampus of in utero exposed mice using bisulfite-pyrosequencing. We show that Mixture N1 decreases DNA methylation in males at three genes that are part of the hypothalamus-pituitary-adrenal (HPA) axis: Nr3c1, Nr3c2, and Crhr1, coding for the glucocorticoid receptor, the mineralocorticoid receptor, and the corticotropin releasing hormone receptor 1, respectively. Furthermore, we show that the decrease in Nr3c1 methylation correlates with increased gene expression, and that Nr3c1, Nr3c2, and Crhr1 methylation correlates with hyperactivity and reduction in social behavior. These findings indicate that an EDC mixture corresponding to a human exposure scenario induces epigenetic changes, and thus programming effects, on the HPA axis that are reflected in the behavioral phenotypes of the adult male offspring.
Collapse
Affiliation(s)
- Michela Di Criscio
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Jennifer Ekholm Lodahl
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens 11527, Greece
| | - Efthymia Kitraki
- Basic Sciences Lab, Faculty of Dentistry, School of Health Sciences, NKUA, Athens 15272, Greece
| | - Ioannis Bakoyiannis
- Biology-Biochemistry Lab, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens 11527, Greece
| | - Anastasia Repouskou
- Basic Sciences Lab, Faculty of Dentistry, School of Health Sciences, NKUA, Athens 15272, Greece
| | - Carl-Gustaf Bornehag
- Faculty of Health, Science and Technology, Department of Health Sciences, Karlstad University, SE- 651 88 Karlstad, Sweden
| | - Chris Gennings
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Diana Lupu
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Joëlle Rüegg
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden.
| |
Collapse
|
11
|
Subclinical hypothyroidism in pregnancy rats impaired offspring's spatial learning and memory and the cerebellar development. Biochem Biophys Res Commun 2022; 602:63-69. [DOI: 10.1016/j.bbrc.2022.02.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/02/2022] [Accepted: 02/26/2022] [Indexed: 11/21/2022]
|
12
|
Kapri D, Fanibunda SE, Vaidya VA. Thyroid hormone regulation of adult hippocampal neurogenesis: Putative molecular and cellular mechanisms. VITAMINS AND HORMONES 2021; 118:1-33. [PMID: 35180924 DOI: 10.1016/bs.vh.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adult hippocampal neurogenesis is sensitive to perturbations in thyroid hormone signaling, with evidence supporting a key role for thyroid hormone and thyroid hormone receptors (TRs) in the regulation of postmitotic progenitor survival and neuronal differentiation. In this book chapter we summarize the current understanding of the effects of thyroid hormone signaling on adult hippocampal progenitor development, and also critically address the role of TRs in regulation of distinct aspects of stage-specific hippocampal progenitor progression. We highlight actions of thyroid hormone on thyroid hormone responsive target genes, and the implications for hippocampal progenitor regulation. Given the influence of thyroid hormone on both mitochondrial and lipid metabolism, we discuss a putative role for regulation of metabolism in the effects of thyroid hormone on adult hippocampal neurogenesis. Finally, we highlight specific ideas that require detailed experimental investigation, and the need for future studies to obtain a deeper mechanistic insight into the influence of thyroid hormone and TRs in the developmental progression of adult hippocampal progenitors.
Collapse
Affiliation(s)
- Darshana Kapri
- 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; Medical Research Centre, Kasturba Health Society, Mumbai, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
13
|
Achour M, Ferdousi F, Sasaki K, Isoda H. Luteolin Modulates Neural Stem Cells Fate Determination: In vitro Study on Human Neural Stem Cells, and in vivo Study on LPS-Induced Depression Mice Model. Front Cell Dev Biol 2021; 9:753279. [PMID: 34790666 PMCID: PMC8591246 DOI: 10.3389/fcell.2021.753279] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/11/2021] [Indexed: 11/23/2022] Open
Abstract
Luteolin is a natural flavone with neurotrophic effects observed on different neuronal cell lines. In the present study, we aimed to assess the effect of luteolin on hNSCs fate determination and the LPS-induced neuroinflammation in a mouse model of depression with astrocytogenesis defect. hNSCs were cultured in basal cell culture medium (control) or medium supplemented with luteolin or AICAR, a known inducer of astrogenesis. A whole-genome transcriptomic analysis showed that luteolin upregulated the expressions of genes related to neurotrophin, dopaminergic, hippo, and Wnt signaling pathways, and downregulated the genes involved in p53, TNF, FOXO, and Notch signaling pathways. We also found that astrocyte-specific gene GFAP, as well as other genes of the key signaling pathways involved in astrogenesis such as Wnt, BMP, and JAK-STAT pathways were upregulated in luteolin-treated hNSCs. On the other hand, neurogenesis and oligodendrogenesis-related genes, TUBB3, NEUROD 1 and 6, and MBP, were downregulated in luteolin-treated hNSCs. Furthermore, immunostaining showed that percentages of GFAP+ cells were significantly higher in luteolin- and AICAR-treated hNSCs compared to control hNSCs. Additionally, RT-qPCR results showed that luteolin upregulated the expressions of GFAP, BMP2, and STAT3, whereas the expression of TUBB3 remained unchanged. Next, we evaluated the effects of luteolin in LPS-induced mice model of depression that represents defects in astrocytogenesis. We found that oral administration of luteolin (10 mg/Kg) for eight consecutive days could decrease the immobility time on tail suspension test, a mouse behavioral test measuring depression-like behavior, and attenuate LPS-induced inflammatory responses by significantly decreasing IL-6 production in mice brain-derived astrocytes and serum, and TNFα and corticosterone levels in serum. Luteolin treatment also significantly increased mature BDNF, dopamine, and noradrenaline levels in the hypothalamus of LPS-induced depression mice. Though the behavioral effects of luteolin did not reach statistical significance, global gene expression analyses of mice hippocampus and brain-derived NSCs highlighted the modulatory effects of luteolin on different signaling pathways involved in the pathophysiology of depression. Altogether, our findings suggest an astrocytogenic potential of luteolin and its possible therapeutic benefits in neuroinflammatory and neurodegenerative diseases. However, further studies are required to identify the specific mechanism of action of luteolin.
Collapse
Affiliation(s)
- Mariem Achour
- Laboratory of Metabolic Biophysics and Applied Pharmacology, Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia.,Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
14
|
Wang L, Liu J, Xu J, Zhang W, Wang R. Coupling of GPR30 mediated neurogenesis and protection with astroglial Aromatase-STAT3 signaling in rat hippocampus after global cerebral ischemia. Mol Cell Endocrinol 2021; 535:111394. [PMID: 34274445 DOI: 10.1016/j.mce.2021.111394] [Citation(s) in RCA: 8] [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: 02/27/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 11/28/2022]
Abstract
Our previous study revealed that G-protein-coupled estrogen receptor-30 (GPR30) agonist G1 serves as a viable alternative neuroprotectant of 17β-estradiol (E2) to attenuate neuroinflammation and improve cognitive function after global cerebral ischemia (GCI). Aromatase, the key enzyme of E2 biosynthesis, is widely expressed in animal and human brain, and its expression and activity are mediated by selective estrogen receptor modulators. In the present study, we explored the long-term protective and reparative effects of G1 in ovariectomized rats after GCI. We used the aromatase inhibitor letrozole to elucidate whether G1 and brain-derived E2 together induce beneficial effects. Our results showed that G1 administration for 28 days a) significantly increased neurogenesis in the hippocampal sub-granular zone and CA1 regions; b) declined CA1 neuronal impairment in a long-term fashion; c) enhanced expression of synaptic proteins and cognitive function; d) and prevented reactive astrocytes loss, wherein aromatase and brain-derived estrogen levels were markedly increased. Additionally, expression and activation of transducer and activator of transcription 3 (STAT3) were increased in CA1 astrocytes of G1-treated animals. Letrozole abolished all of the observed benefits of G1. Our results suggest that GPR30 activation mediates long-term neuroprotection and neurogenesis in the hippocampus following GCI, with potential mechanism coupling with the activation of astroglial aromatase-STAT3 signaling.
Collapse
Affiliation(s)
- Lu Wang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Jiahao Liu
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Jing Xu
- Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Wenli Zhang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Ruimin Wang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
15
|
Costa Reis LT, Sena de Souza J, Hirochi Herai R, Cunha EB, Ribeiro Pereira Soares J, Santos El-Bachá R, Diogenes Amaral da Silva V, Aurelio Romano M, Marino Romano R, Izabel Chiamolera M, Giannocco G, Lima Costa S, Dias da Silva MR, Telles da Cunha Lima S. Intergenerational thyroid hormone homeostasis imbalance in cerebellum of rats perinatally exposed to glyphosate-based herbicide. ENVIRONMENTAL TOXICOLOGY 2021; 36:1031-1042. [PMID: 33512083 DOI: 10.1002/tox.23102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 05/24/2023]
Abstract
Agrochemicals became a public health concern due to increased human exposure and possible endocrine disruption effects in several organs, including the brain. Thyroid hormones controls neurodevelopment, which turn them sensitive to endocrine disruptors (EDs). In this work, we evaluated the effect of glyphosate-based herbicides (GBH) as an intergenerational endocrine disrupter on thyroid homeostasis in cerebellar cells. Female pregnant Wistar rats were exposed to Roundup Transorb® solution at 5 and 50 mg/kg/day, from gestation day 18 to post-natal day 5 (P5). Cerebellum of male offspring was used to evaluate gene expression. The mRNA levels of thyroid hormone receptors, hormonal conversion enzymes, hormone transporters, as well as, de novo epigenetic regulators were altered, with some of these genes presenting a non-monotonic dose response. Furthermore, metabolomic profile correlation with tested dose demonstrated altered metabolic profile, in agreement with cerebellar gene alterations. Moreover, cerebellar primary cultures exposed to non-toxic GBH concentration presented a decrease level in glial fibrillary acidic protein, a protein regulated by endocrine signals. In conclusion, our results indicate that animals exposed to non-toxic GBH doses during perinatal phase carry intergenerational alterations in key regulators of cellular thyroid hormone homeostasis and epigenetic controllers in adulthood, indicating the possible ED effect of GBH based on epigenetic alterations.
Collapse
Affiliation(s)
- Luã Tainã Costa Reis
- Laboratório de Bioprospecção e Biotecnologia Instituto de Biologia, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Janaina Sena de Souza
- Departamento de Medicina Disciplina de Endocrinologia Clínica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Roberto Hirochi Herai
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
- Research Department, Lico Kaesemodel Institute, Curitiba, Paraná, Brazil
| | - Eduardo Brunetti Cunha
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | | | - Ramon Santos El-Bachá
- Laboratório de Neuroquímica e Biologia Celular, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | | | - Marco Aurelio Romano
- Departamento de Farmácia, Universidade Estadual do Centro-Oeste (UNICENTRO), Guarapuava, Brazil
| | - Renata Marino Romano
- Departamento de Farmácia, Universidade Estadual do Centro-Oeste (UNICENTRO), Guarapuava, Brazil
| | - Maria Izabel Chiamolera
- Departamento de Medicina Disciplina de Endocrinologia Clínica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Gisele Giannocco
- Departamento de Medicina Disciplina de Endocrinologia Clínica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Silvia Lima Costa
- Laboratório de Neuroquímica e Biologia Celular, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Magnus Régios Dias da Silva
- Departamento de Medicina Disciplina de Endocrinologia Clínica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Suzana Telles da Cunha Lima
- Laboratório de Bioprospecção e Biotecnologia Instituto de Biologia, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- AnaclinGENE, Curitiba, Paraná, Brazil
| |
Collapse
|
16
|
Schiera G, Di Liegro CM, Di Liegro I. Involvement of Thyroid Hormones in Brain Development and Cancer. Cancers (Basel) 2021; 13:2693. [PMID: 34070729 PMCID: PMC8197921 DOI: 10.3390/cancers13112693] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/21/2022] Open
Abstract
The development and maturation of the mammalian brain are regulated by thyroid hormones (THs). Both hypothyroidism and hyperthyroidism cause serious anomalies in the organization and function of the nervous system. Most importantly, brain development is sensitive to TH supply well before the onset of the fetal thyroid function, and thus depends on the trans-placental transfer of maternal THs during pregnancy. Although the mechanism of action of THs mainly involves direct regulation of gene expression (genomic effects), mediated by nuclear receptors (THRs), it is now clear that THs can elicit cell responses also by binding to plasma membrane sites (non-genomic effects). Genomic and non-genomic effects of THs cooperate in modeling chromatin organization and function, thus controlling proliferation, maturation, and metabolism of the nervous system. However, the complex interplay of THs with their targets has also been suggested to impact cancer proliferation as well as metastatic processes. Herein, after discussing the general mechanisms of action of THs and their physiological effects on the nervous system, we will summarize a collection of data showing that thyroid hormone levels might influence cancer proliferation and invasion.
Collapse
Affiliation(s)
- Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Dipartimento di Biomedicina, Neuroscienze e Diagnostica avanzata) (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
17
|
Sharma KD, Schaal D, Kore RA, Hamzah RN, Pandanaboina SC, Hayar A, Griffin RJ, Srivatsan M, Reyna NS, Xie JY. Glioma-derived exosomes drive the differentiation of neural stem cells to astrocytes. PLoS One 2020; 15:e0234614. [PMID: 32649728 PMCID: PMC7351166 DOI: 10.1371/journal.pone.0234614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/29/2020] [Indexed: 01/15/2023] Open
Abstract
Exosomes appear to be effective inter-cellular communicators delivering several types of molecules, such as proteins and RNAs, suggesting that they could influence neural stem cell (NSC) differentiation. Our RNA sequencing studies demonstrated that the RNAs related to cell proliferation and astrocyte differentiation were upregulated in human mesenchymal stem cells (hMSC) when co-cultured with exosomes obtained from the culture medium of human glioma cells (U87). Metallothionein 3 and elastin genes, which are related to cell proliferation, increased 10 and 7.2 fold, respectively. Expression of genes for astrocyte differentiation, such as tumor growth factor alpha, induced protein 3 of the NOTCH1 family, colony stimulating factor and interleukin 6 of the STAT3 family and Hes family bHLH transcription factor 1 also increased by 2.3, 10, 4.7 and 2.9 fold, respectively. We further examined the effects of these exosomes on rat fetal neural stem cell (rNSC) differentiation using the secreted exosomes from U87 glioma cells or exosomes from U87 cells that were stimulated with interleukin 1β (IL-1β). The rNSCs, extracted from rat brains at embryonic day 14 (E14), underwent a culture protocol that normally leads to predominant (~90%) differentiation to ODCs. However, in the presence of the exosomes from untreated or IL-1β-treated U87 cells, significantly more cells differentiated into astrocytes, especially in the presence of exosomes obtained from the IL-1β-challenged glioma cells. Moreover, glioma-derived exosomes appeared to inhibit rNSC differentiation into ODCs or astrocytes as indicated by a significantly increased population of unlabeled cells. A portion of the resulting astrocytes co-expressed both CD133 and glial fibrillary acidic protein (GFAP) suggesting that exosomes from U87 cells could promote astrocytic differentiation of NSCs with features expected from a transformed cell. Our data clearly demonstrated that exosomes secreted by human glioma cells provide a strong driving force for rat neural stem cells to differentiate into astrocytes, uncovering potential pathways and therapeutic targets that might control this aggressive tumor type.
Collapse
Affiliation(s)
- Krishna D. Sharma
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Danielle Schaal
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Rajshekhar A. Kore
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Rabab N. Hamzah
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Sahitya Chetan Pandanaboina
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Abdallah Hayar
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Malathi Srivatsan
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Nathan S. Reyna
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Jennifer Yanhua Xie
- Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, Arkansas, United States of America
| |
Collapse
|
18
|
Sonic Hedgehog and Triiodothyronine Pathway Interact in Mouse Embryonic Neural Stem Cells. Int J Mol Sci 2020; 21:ijms21103672. [PMID: 32456161 PMCID: PMC7279276 DOI: 10.3390/ijms21103672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 11/17/2022] Open
Abstract
Neural stem cells are fundamental to development of the central nervous system (CNS)-as well as its plasticity and regeneration-and represent a potential tool for neuro transplantation therapy and research. This study is focused on examination of the proliferation dynamic and fate of embryonic neural stem cells (eNSCs) under differentiating conditions. In this work, we analyzed eNSCs differentiating alone and in the presence of sonic hedgehog (SHH) or triiodothyronine (T3) which play an important role in the development of the CNS. We found that inhibition of the SHH pathway and activation of the T3 pathway increased cellular health and survival of differentiating eNSCs. In addition, T3 was able to increase the expression of the gene for the receptor smoothened (Smo), which is part of the SHH signaling cascade, while SHH increased the expression of the T3 receptor beta gene (Thrb). This might be the reason why the combination of SHH and T3 increased the expression of the thyroxine 5-deiodinase type III gene (Dio3), which inhibits T3 activity, which in turn affects cellular health and proliferation activity of eNSCs.
Collapse
|
19
|
Lin L, Li C, Zhang D, Yuan M, Chen CH, Li M. Synergic Effects of Berberine and Curcumin on Improving Cognitive Function in an Alzheimer's Disease Mouse Model. Neurochem Res 2020; 45:1130-1141. [PMID: 32080784 DOI: 10.1007/s11064-020-02992-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and no effective therapies have been found to prevent or cure AD to date. Berberine and curcumin are extracts from traditional Chinese herbs that have a long history of clinical benefits for AD. Here, using a transgenic AD mouse model, we found that the combined berberine and curcumin treatment had a much better effect on improving the cognitive function of mice than the single-drug treatment, suggesting synergic effects of the combined berberine and curcumin treatment. In addition, we found that the combined berberine and curcumin treatment had significant synergic effects on reducing soluble amyloid-β-peptide(1-42) production. Furthermore, the combination treatment also had remarkable synergic effects on decreasing inflammatory responses and oxidative stress in both the cortex and hippocampus of AD mice. We also found that the combination treatment performed much better than the single drugs in reducing the APP and BACE1 levels and increasing AMPKα phosphorylation and cell autophagy, which might be the underlying mechanism of the synergic effects. Taken together, the result of this study reveal the synergic effects and potential underlying mechanisms of the combined berberine and curcumin treatment in improving the symptoms of AD in mice. This study sheds light on a new strategy for exploring new phytotherapies for AD and also emphasizes that more research should focus on the synergic effects of herbal drugs in the future.
Collapse
Affiliation(s)
- Lin Lin
- Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Cheng Li
- Department of Public Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Deyi Zhang
- Department of Anesthesiology, Mianyang People's Hospital, Mianyang, 621000, Sichuan, China
| | - Mingxiang Yuan
- Department of Gynaecology and Obstetrics, Mianyang People's Hospital, Mianyang, 621000, Sichuan, China
| | - Chun-Hai Chen
- Department of Occupational Health, Amy Medical University, Chongqing, 400038, China.
| | - Maoquan Li
- Affiliated Traditional Chinese Medicine Hospital of Chengdu Medical College, Chengdu, 610300, Sichuan, China. .,Chengdu Qingbaijiang District Traditional Chinese Medicine Hospital, Chengdu, 610300, Sichuan, China. .,Department of Public Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|
20
|
Transcriptomic insight into cadmium-induced neurotoxicity in embryonic neural stem/progenitor cells. Toxicol In Vitro 2020; 62:104686. [DOI: 10.1016/j.tiv.2019.104686] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
|
21
|
Gothié J, Vancamp P, Demeneix B, Remaud S. Thyroid hormone regulation of neural stem cell fate: From development to ageing. Acta Physiol (Oxf) 2020; 228:e13316. [PMID: 31121082 PMCID: PMC9286394 DOI: 10.1111/apha.13316] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022]
Abstract
In the vertebrate brain, neural stem cells (NSCs) generate both neuronal and glial cells throughout life. However, their neuro‐ and gliogenic capacity changes as a function of the developmental context. Despite the growing body of evidence on the variety of intrinsic and extrinsic factors regulating NSC physiology, their precise cellular and molecular actions are not fully determined. Our review focuses on thyroid hormone (TH), a vital component for both development and adult brain function that regulates NSC biology at all stages. First, we review comparative data to analyse how TH modulates neuro‐ and gliogenesis during vertebrate brain development. Second, as the mammalian brain is the most studied, we highlight the molecular mechanisms underlying TH action in this context. Lastly, we explore how the interplay between TH signalling and cell metabolism governs both neurodevelopmental and adult neurogenesis. We conclude that, together, TH and cellular metabolism regulate optimal brain formation, maturation and function from early foetal life to adult in vertebrate species.
Collapse
Affiliation(s)
- Jean‐David Gothié
- Department of Neurology & Neurosurgery Montreal Neurological Institute & Hospital, McGill University Montreal Quebec Canada
| | - Pieter Vancamp
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| | | | - Sylvie Remaud
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| |
Collapse
|
22
|
Churilov LP, Sobolevskaia PA, Stroev YI. Thyroid gland and brain: Enigma of Hashimoto's encephalopathy. Best Pract Res Clin Endocrinol Metab 2019; 33:101364. [PMID: 31801687 DOI: 10.1016/j.beem.2019.101364] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The versatile clinical manifestations of the Hashimoto's chronic autoimmune thyroiditis often include psycho-neurological disorders. Although hypothyroidism disturbs significantly the ontogenesis and functions of central nervous system, causing in severe cases of myxedema profound impairment of cognitive abilities and even psychosis, the behavioral, motor and other psychoneurological disorders accompany euthyroid and slightly hypothyroid cases and periods of Hashimoto's disease as well, thus constituting the picture of so called "Hashimoto's encephalopathy". The entity, although discussed and explored for more than 50 years since its initial descriptions, remains an enigma of thyroidology and psychiatry, because its etiology and pathogenesis are obscure. The paper describes the development of current views on the role of thyroid in ontogeny and functions of brain, as well as classical and newest ideas on the etiology and pathogenesis of Hashimot's encephalopathy. The synopsis of the world case reports and research literature on this disorder is added with authors' own results obtained by study of 17 cases of Hashimoto's thyroiditis with schizophrenia-like clinical manifestations. The relation of the disease to adjuvant-like etiological factors is discussed. Three major mechanistic concepts of Hashimoto's encephalopathy are detailed, namely cerebral vasculitis theory, hormone dysregulation theory and concept, explaining the disease via direct action of the autoantibodies against various thyroid (thyroperoxidase, thyroglobulin, and TSH-receptor) and several extrathyroid antigens (alpha-enolase and other enzymes, gangliosides and MOG-protein, onconeuronal antigens) - all of them expressed in the brain. The article demonstrates that all above mentioned concepts intermingle and prone to unification, suggesting the unified scheme of pathogenesis for the Hashimoto's encephalopathy. The clinical manifestations, criteria, forms, course, treatment and prognosis of Hashimoto's encephalopathy and its comorbidity to other diseases - are also discussed in brief. The relation between Hashimoto's encephalopathy and non-vasculitis autoimmune encephalomyelitides of paraneoplastic and non-paraneoplastic origin is emphasized [1 figure, bibliography - 200 references].
Collapse
Affiliation(s)
- Leonid P Churilov
- Laboratory of the Mosaic of Autoimmunity, Saint Petersburg State University, Russia.
| | - Polina A Sobolevskaia
- Laboratory of the Mosaic of Autoimmunity, Saint Petersburg State University, Russia.
| | - Yuri I Stroev
- Laboratory of the Mosaic of Autoimmunity, Saint Petersburg State University, Russia.
| |
Collapse
|
23
|
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
|
24
|
Xiu L, Xing Q, Mao J, Sun H, Teng W, Shan Z. miRNA-125b-5p Suppresses Hypothyroidism Development by Targeting Signal Transducer and Activator of Transcription 3. Med Sci Monit 2018; 24:5041-5049. [PMID: 30027933 PMCID: PMC6067029 DOI: 10.12659/msm.907510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background A deficiency of maternal thyroid hormones (THs) during pregnancy has severe impacts on fetal brain development. Neural stem cells (NSCs) are major targets of THs and provided a powerful model to explore the underlying mechanism of THs during brain development. Although miRNA-125 might be associated with the NSCs differentiation, the relationship between miR-125 and hypothyroidism (HypoT) development remains unclear. Material/Methods In our study, we screened a differentially expressed gene miR-125b-5p from brain between euthyroid (EuT) and HypoT rats. In vitro, we employed anion exchange resin to remove THs to stimulate HypoT. QRT-PCR and Western blot were used to examine the expression of signal transducer and activator of transcription 3 (Stat3). The relationship between miR-125b-5p and Stat3 was detected via a dual-luciferase assay. Results QRT-PCR results showed that the level of miR-125b-5p in HypoT rat brains was significantly suppressed, suggesting some relationship between miR-125b-5p and HypoT. In C17.2, miR-125b-5p promoted cell differentiation into neurons by regulating the expression of tubulin beta chain 3 (TUBB3) and glial fibrillary acid protein (GFAP). QRT-PCR and Western blot results revealed that miR-125b-5p mimic modulated the contents of total Stat3 and p-Stat3. A dual-luciferase assay showed that miR-125b-5p negatively regulated the expression of Stat3 by binding with the first site in 3′ UTR of Stat3. Conclusions These results revealed Stat3 is a new target of miR-125b-5p and revealed the mechanism of miR-125b-5p suppressing HypoT development. These findings provide a new target for HypoT therapy.
Collapse
Affiliation(s)
- Liu Xiu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,Shijiazhuang First Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Qian Xing
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Jinyuan Mao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Huakun Sun
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning, Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
25
|
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]
|
26
|
Gkikas D, Tsampoula M, Politis PK. Nuclear receptors in neural stem/progenitor cell homeostasis. Cell Mol Life Sci 2017; 74:4097-4120. [PMID: 28638936 PMCID: PMC11107725 DOI: 10.1007/s00018-017-2571-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022]
Abstract
In the central nervous system, embryonic and adult neural stem/progenitor cells (NSCs) generate the enormous variety and huge numbers of neuronal and glial cells that provide structural and functional support in the brain and spinal cord. Over the last decades, nuclear receptors and their natural ligands have emerged as critical regulators of NSC homeostasis during embryonic development and adult life. Furthermore, substantial progress has been achieved towards elucidating the molecular mechanisms of nuclear receptors action in proliferative and differentiation capacities of NSCs. Aberrant expression or function of nuclear receptors in NSCs also contributes to the pathogenesis of various nervous system diseases. Here, we review recent advances in our understanding of the regulatory roles of steroid, non-steroid, and orphan nuclear receptors in NSC fate decisions. These studies establish nuclear receptors as key therapeutic targets in brain diseases.
Collapse
Affiliation(s)
- Dimitrios Gkikas
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece.
| |
Collapse
|
27
|
Chen C, Ma Q, Deng P, Yang J, Yang L, Lin M, Yu Z, Zhou Z. Critical role of TRPC1 in thyroid hormone-dependent dopaminergic neuron development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1900-1912. [PMID: 28779972 DOI: 10.1016/j.bbamcr.2017.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/21/2017] [Accepted: 07/31/2017] [Indexed: 01/20/2023]
Abstract
Thyroid hormones play a crucial role in midbrain dopaminergic (DA) neuron development. However, the underlying molecular mechanisms remain largely unknown. In this study, we revealed that thyroid hormone treatment evokes significant calcium entry through canonical transient receptor potential (TRPC) channels in ventral midbrain neural stem cells and this calcium signaling is essential for thyroid hormone-dependent DA neuronal differentiation. We also found that TRPC1 is the dominant TRPC channel expressed in ventral midbrain neural stem cells which responds to thyroid hormone. In addition, thyroid hormone increases TRPC1 expression through its receptor alpha 1 during DA neuron differentiation, and, importantly, produces calcium signals by activating TRPC1 channels. In vivo and in vitro gene silencing experiments indicate that TRPC1-mediated calcium signaling is required for thyroid hormone-dependent DA neuronal differentiation. Finally, we confirmed that the activation of OTX2, a determinant of DA neuron development and the expression of which is induced by thyroid hormone, is dependent on TRPC1-mediated calcium signaling. These data revealed the molecular mechanisms of how thyroid hormone regulates DA neuron development from ventral midbrain neural stem cells, particularly endowing a novel physiological relevance to TRPC1 channels.
Collapse
Affiliation(s)
- Chunhai Chen
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China.
| | - Qinglong Ma
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China
| | - Jianjing Yang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9148, USA
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China
| | - Min Lin
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China
| | - Zhou Zhou
- Department of Occupational Health, Third Military Medical University, No.30 Gaotanyan Street, Chongqing 400038, China.
| |
Collapse
|
28
|
Dong J, Song H, Wang Y, Li M, Yu Y, Wang Y, Chen J. Maternal Different Degrees of Iodine Deficiency during Pregnant and Lactation Impair the Development of Cerebellar Pinceau in Offspring. Front Neurosci 2017; 11:298. [PMID: 28611576 PMCID: PMC5446996 DOI: 10.3389/fnins.2017.00298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/12/2017] [Indexed: 11/13/2022] Open
Abstract
Aims: Iodine is critical for synthesis of thyroid hormones (TH). And iodine deficiency (ID) is one of the most significant reasons of intellectual disability and motor memory impairment, although the potential mechanisms are still under investigation. Presently, mild ID and marginal ID are largely ignored problems for women of child bearing age. Mild ID is a subtle form of TH deficiency, which shows low levels of free thyroxine (FT4) and relatively normal free triiodothyronine (FT3) or thyroid stimulation hormone (TSH). And marginal ID is a milder form of ID with decreased total T4 (TT4) but relatively normal FT3, FT4, and TSH. Therefore, we investigated the effects of maternal different degrees of ID on the development of pinceau in cerebellar purkinje cells (PCs) and studied the expression of pinceau related protein, which is crucial for the development and maturation of pinceau. Methods and Results: Three developmental iodine deficient rat models were created by feeding dam rats with an iodine-deficient diet and deionized water supplemented with potassiumiodide. Our study showed that different degrees of ID inhibited cerebellar pinceau synapse development and maturation on postnatal day (PN) 14 and PN21. What's more, mild and severe ID reduced the expression of AnkG, β4-spectrin, neurofascin186 and NrCAM on PN7, PN14, and PN21. However, marginal ID rarely altered expression of these proteins in the offspring. Conclusion: These results suggested that maternal mild and severe ID impaired the development and maturation of cerebellar pinceau, which may be attributed to the decrease of AnkG, β4-spectrin, neurofascin 186, and NrCAM. And the alteration of development and maturation in cerebellar pinceau in the offspring were also observed following maternal marginal ID, which is slighter than that of mild ID.
Collapse
Affiliation(s)
- Jing Dong
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Heling Song
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yuan Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Min Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Ye Yu
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
29
|
Dach K, Bendt F, Huebenthal U, Giersiefer S, Lein PJ, Heuer H, Fritsche E. BDE-99 impairs differentiation of human and mouse NPCs into the oligodendroglial lineage by species-specific modes of action. Sci Rep 2017; 7:44861. [PMID: 28317842 PMCID: PMC5357893 DOI: 10.1038/srep44861] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 02/15/2017] [Indexed: 01/02/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are bioaccumulating flame retardants causing developmental neurotoxicity (DNT) in humans and rodents. Their DNT effects are suspected to involve thyroid hormone (TH) signaling disruption. Here, we tested the hypothesis whether disturbance of neural progenitor cell (NPC) differentiation into the oligodendrocyte lineage (O4+ cells) by BDE-99 involves disruption of TH action in human and mouse (h,m)NPCs. Therefore, we quantified differentiation of NPCs into O4+ cells and measured their maturation via expression of myelin-associated genes (hMBP, mMog) in presence and absence of TH and/or BDE-99. T3 promoted O4+ cell differentiation in mouse, but not hNPCs, and induced hMBP/mMog gene expression in both species. BDE-99 reduced generation of human and mouse O4+ cells, but there is no indication for BDE-99 interfering with cellular TH signaling during O4+ cell formation. BDE-99 reduced hMBP expression due to oligodendrocyte reduction, but concentrations that did not affect the number of mouse O4+ cells inhibited TH-induced mMog transcription by a yet unknown mechanism. In addition, ascorbic acid antagonized only the BDE-99-dependent loss of human, not mouse, O4+ cells by a mechanism probably independent of reactive oxygen species. These data point to species-specific modes of action of BDE-99 on h/mNPC development into the oligodendrocyte lineage.
Collapse
Affiliation(s)
- Katharina Dach
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Farina Bendt
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Ulrike Huebenthal
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Susanne Giersiefer
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California 95616, United States
| | - Heike Heuer
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| |
Collapse
|
30
|
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: 256] [Impact Index Per Article: 32.0] [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
|
31
|
Sak S, Agacayak E, Tunc SY, Icen MS, Findik FM, Sak ME, Yalinkaya A, Gul T. Amniotic fluid paraoxonase-1 activity, thyroid hormone concentration and oxidant status in neural tube defects. J Obstet Gynaecol Res 2016; 42:1080-5. [DOI: 10.1111/jog.13026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/12/2016] [Accepted: 03/11/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Sibel Sak
- Department of Obstetrics and Gynaecology; Harran University; Sanliurfa Turkey
| | - Elif Agacayak
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Senem Yaman Tunc
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Mehmet Sait Icen
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Fatih Mehmet Findik
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Muhammet Erdal Sak
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Ahmet Yalinkaya
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| | - Talip Gul
- Department of Obstetrics and Gynaecology; Dicle University, School of Medicine; Diyarbakir Turkey
| |
Collapse
|
32
|
Heng BC, Lim LW, Wu W, Zhang C. An Overview of Protocols for the Neural Induction of Dental and Oral Stem Cells In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:220-50. [PMID: 26757369 DOI: 10.1089/ten.teb.2015.0488] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To date, various adult stem cells have been identified within the oral cavity, including dental pulp stem cells, dental follicle stem cells, stem cells from apical papilla, stem cells from human exfoliated deciduous teeth, periodontal ligament stem cells, and mesenchymal stem cells from the gingiva. All of these possess neurogenic potential due to their common developmental origin from the embryonic neural crest. Besides the relative ease of isolation of these adult stem cells from readily available biological waste routinely produced during dental treatment, these cells also possess the advantage of immune compatibility in autologous transplantation. In recent years, much interest has been focused on the derivation of neural lineages from these adult stem cells for therapeutic applications in the brain, spinal cord, and peripheral nerve regeneration. In addition, there are also promising nontherapeutic applications of stem cell-derived neurons in pharmacological and toxicological screening of neuroactive drugs, and for in vitro modeling of neurodevelopmental and neurodegenerative diseases. Hence, this review will critically examine the diverse array of in vitro neural induction protocols that have been devised for dental and oral-derived stem cells. These protocols are defined not only by the culture milieu comprising the basal medium plus growth factors, small molecules, and other culture supplements but also by the substrata/surface coatings utilized, the presence of multiple culture stages, the total culture duration, the initial seeding density, and whether the spheroid/neurosphere formation is being utilized to recapitulate the three-dimensional neural differentiation microenvironment that is naturally present physiologically in vivo.
Collapse
Affiliation(s)
- Boon Chin Heng
- 1 Comprehensive Dental Care, Endodonthics, Faculty of Dentistry, The University of Hong Kong , Pokfulam, Hong Kong
| | - Lee Wei Lim
- 2 School of Biomedical Sciences, The University of Hong Kong , Pokfulam, Hong Kong
| | - Wutian Wu
- 2 School of Biomedical Sciences, The University of Hong Kong , Pokfulam, Hong Kong
| | - Chengfei Zhang
- 1 Comprehensive Dental Care, Endodonthics, Faculty of Dentistry, The University of Hong Kong , Pokfulam, Hong Kong
| |
Collapse
|
33
|
Ma Q, Chen C, Deng P, Zhu G, Lin M, Zhang L, Xu S, He M, Lu Y, Duan W, Pi H, Cao Z, Pei L, Li M, Liu C, Zhang Y, Zhong M, Zhou Z, Yu Z. Extremely Low-Frequency Electromagnetic Fields Promote In Vitro Neuronal Differentiation and Neurite Outgrowth of Embryonic Neural Stem Cells via Up-Regulating TRPC1. PLoS One 2016; 11:e0150923. [PMID: 26950212 PMCID: PMC4780708 DOI: 10.1371/journal.pone.0150923] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Gang Zhu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Min Lin
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Shangcheng Xu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Weixia Duan
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zhengwang Cao
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Liping Pei
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Li
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chuan Liu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yanwen Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Zhong
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| | - Zhengping Yu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| |
Collapse
|
34
|
Zhou B, Osinski JM, Mateo JL, Martynoga B, Sim FJ, Campbell CE, Guillemot F, Piper M, Gronostajski RM. Loss of NFIX Transcription Factor Biases Postnatal Neural Stem/Progenitor Cells Toward Oligodendrogenesis. Stem Cells Dev 2015; 24:2114-26. [DOI: 10.1089/scd.2015.0136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Bo Zhou
- Department of Biochemistry, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Jason M. Osinski
- Department of Biochemistry, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Juan L. Mateo
- Centre for Organismal Studies Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Ben Martynoga
- Division of Molecular Neurobiology, MRC, London, United Kingdom
| | - Fraser J. Sim
- Department of Genetics, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| | - Christine E. Campbell
- Department of Biochemistry, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
| | | | - Michael Piper
- School of Biomedical Sciences, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Richard M. Gronostajski
- Department of Biochemistry, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
- Department of Genetics, Genomics and Bioinformatics Program, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
35
|
Calzà L, Fernández M, Giardino L. Role of the Thyroid System in Myelination and Neural Connectivity. Compr Physiol 2015; 5:1405-21. [DOI: 10.1002/cphy.c140035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
36
|
Chen C, Ma Q, Chen X, Zhong M, Deng P, Zhu G, Zhang Y, Zhang L, Yang Z, Zhang K, Guo L, Wang L, Yu Z, Zhou Z. Thyroid Hormone-Otx2 Signaling Is Required for Embryonic Ventral Midbrain Neural Stem Cells Differentiated into Dopamine Neurons. Stem Cells Dev 2015; 24:1751-65. [PMID: 25867707 DOI: 10.1089/scd.2014.0489] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Midbrain dopamine (DA) neurons are essential for maintaining multiple brain functions. These neurons have also been implicated in relation with diverse neurological disorders. However, how these neurons are developed from neuronal stem cells (NSCs) remains largely unknown. In this study, we provide both in vivo and in vitro evidence that the thyroid hormone, an important physiological factor for brain development, promotes DA neuron differentiation from embryonic ventral midbrain (VM) NSCs. We find that thyroid hormone deficiency during development reduces the midbrain DA neuron number, downregulates the expression of tyrosine hydroxylase (TH) and the dopamine transporter (DAT), and impairs the DA neuron-dependent motor behavior. In addition, thyroid hormone treatment during VM NSC differentiation in vitro increases the production of DA neurons and upregulates the expression of TH and DAT. We also found that the thyroid hormone enhances the expression of Otx2, an important determinant of DA neurogenesis, during DA neuron differentiation. Our in vitro gene silencing experiments indicate that Otx2 is required for thyroid hormone-dependent DA neuron differentiation from embryonic VM NSCs. Finally, we revealed both in vivo and in vitro that the thyroid hormone receptor alpha 1 is expressed in embryonic VM NSCs. Furthermore, it participates in the effects of thyroid hormone-induced Otx2 upregulation and DA neuron differentiation. These data demonstrate the role and molecular mechanisms of how the thyroid hormone regulates DA neuron differentiation from embryonic VM NSCs, particularly providing new mechanisms and a potential strategy for generating dopamine neurons from NSCs.
Collapse
Affiliation(s)
- Chunhai Chen
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Qinglong Ma
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Xiaowei Chen
- 2 Brain Research Center, Third Military Medical University , Chongqing, China
| | - Min Zhong
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Ping Deng
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Gang Zhu
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Yanwen Zhang
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Lei Zhang
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Zhiqi Yang
- 2 Brain Research Center, Third Military Medical University , Chongqing, China
| | - Kuan Zhang
- 2 Brain Research Center, Third Military Medical University , Chongqing, China
| | - Lu Guo
- 3 Department of Neurology, Daping Hospital, Third Military Medical University , Chongqing, China
| | - Liting Wang
- 4 Biomedical Analysis Center, Third Military Medical University , Chongqing, China
| | - Zhengping Yu
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| | - Zhou Zhou
- 1 Department of Occupational Health, Third Military Medical University , Chongqing, China
| |
Collapse
|
37
|
Lin L, Yuan J, Sander B, Golas MM. In Vitro Differentiation of Human Neural Progenitor Cells Into Striatal GABAergic Neurons. Stem Cells Transl Med 2015; 4:775-88. [PMID: 25972145 DOI: 10.5966/sctm.2014-0083] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 03/05/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : Huntington's disease (HD) results from a CAG repeat expansion in the gene encoding the huntingtin protein. This inherited disorder is characterized by progressive neurodegeneration. In particular, HD progression involves the loss of striatal projection neurons. The limited availability of reliable sources of human striatal projection neurons currently hampers our understanding of HD mechanisms and hinders the development of novel HD treatments. In this paper, we described two- and three-step methods for differentiating human neural progenitor cells toward striatal projection neurons. In the two-step differentiation protocol, 90%, 54%, and 6% of MAP2-positive cells were immunopositive for GABA, calbindin (CALB1), and DARPP-32/PPP1R1B, respectively. In the three-step differentiation protocol, 96%, 84%, and 21% of MAP2-positive cells were immunopositive for GABA, calbindin, and DARPP-32/PPP1R1B, respectively. In line with a striatal projection neuron phenotype, cells differentiated with our protocols displayed significantly increased expression of MAP2, CALB1, DARPP-32/PPP1R1B, ARPP21, and CTIP2. Application of glutamate receptor agonists induced calcium influx; accordingly, the cells also expressed various ionotropic glutamate receptor subunits. Differentiated cells also released GABA on stimulation. We suggest that our three-step differentiation protocol presents a reliable and simplified method for the generation of striatal projection neurons, yielding a critical resource for neuronal physiology and neurodegenerative disorder studies. SIGNIFICANCE The earliest changes in the neurodegenerative disorder Huntington's disease affect a specific type of brain neurons, the so-called medium spiny neurons of the striatum. In this study, two protocols were developed for the differentiation of neural progenitor cells into striatal medium spiny neurons, and the differentiated neurons were extensively characterized. The data indicate that the three-step differentiation protocol presents a reliable and simplified method for the generation of striatal medium spiny neurons. The generated striatal medium spiny neurons could represent a critical resource for the study of neurodegenerative disorders, a model system for drug discovery, and a step toward cell-based regeneration therapies.
Collapse
Affiliation(s)
- Lin Lin
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Juan Yuan
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Bjoern Sander
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Monika M Golas
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| |
Collapse
|
38
|
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
|
39
|
Min H, Dong J, Wang Y, Wang Y, Teng W, Xi Q, Chen J. Maternal Hypothyroxinemia-Induced Neurodevelopmental Impairments in the Progeny. Mol Neurobiol 2015; 53:1613-1624. [PMID: 25666160 DOI: 10.1007/s12035-015-9101-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/15/2015] [Indexed: 12/16/2022]
Abstract
Maternal hypothyroxinemia can induce neurodevelopmental impairments in the developing fetus. We here review recent studies on the epidemiology and molecular mechanisms associated with this important public health issue. In 2011, the American Thyroid Association defined maternal hypothyroxinemia as low serum free thyroxine (FT4) levels (<5th or <10th percentile) existing in conjunction with normal serum free triiodothyronine (FT3) or thyroid stimulating hormone (TSH) levels during pregnancy. Compared to clinical or subclinical hypothyroidism, hypothyroxinemia is more commonly found in pregnant women. Hypothyroxinemia usually ensues in response to several factors, such as mild iodine deficiency, environmental endocrine disrupters, or certain thyroid diseases. Unequivocal evidence demonstrates that maternal hypothyroxinemia leads to negative effects on fetal brain development, increasing the risks for cognitive deficits and poor psychomotor development in resulting progeny. In support of this, rodent models provide direct evidence of neurodevelopmental damage induced by maternal hypothyroxinemia, including dendritic and axonal growth limitation, neural abnormal location, and synaptic function alteration. The neurodevelopmental impairments induced by hypothyroxinemia suggest an independent role of T4. Increasing evidence indicates that adequate thyroxine is required for the mothers in order to protect against the abnormal brain development in their progeny.
Collapse
Affiliation(s)
- Hui Min
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110013, People's Republic of China
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jing Dong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110013, People's Republic of China
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110013, People's Republic of China
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yuan Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110013, People's Republic of China
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Weiping Teng
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Qi Xi
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110013, People's Republic of China.
- Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
40
|
Sirakov M, Kress E, Nadjar J, Plateroti M. Thyroid hormones and their nuclear receptors: new players in intestinal epithelium stem cell biology? Cell Mol Life Sci 2014; 71:2897-907. [PMID: 24604390 PMCID: PMC11113153 DOI: 10.1007/s00018-014-1586-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/31/2014] [Accepted: 02/12/2014] [Indexed: 12/14/2022]
Abstract
Thyroid hormones participate in the development and homeostasis of several organs and tissues. It is well documented that they act via nuclear receptors, the TRs, which are transcription factors whose function is modulated by the hormone T3. Importantly, T3-induced physiological response within a cell depends on the specific TR expression and on the T3 bioavailability. However, in addition to this T3-dependent control of TR functionality, increasing data show that the action of TRs is coordinated and integrated with other signaling pathways, specifically at the level of stem/progenitor cell populations. By focusing on the intestinal epithelium of both amphibians and mammals we summarize here new data in support of a role for thyroid hormones and the TR nuclear receptors in stem cell biology. This new concept may be extended to other organs and have biological relevance in therapeutic approaches aimed to target stem cells such as tissue engineering and cancer.
Collapse
Affiliation(s)
- Maria Sirakov
- Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Brussels, Belgium
| | - Elsa Kress
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, 69622 Villeurbanne, France
| | - Julien Nadjar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, 69622 Villeurbanne, France
| | - Michelina Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, 69622 Villeurbanne, France
| |
Collapse
|
41
|
Exposure to 1800 MHz radiofrequency radiation impairs neurite outgrowth of embryonic neural stem cells. Sci Rep 2014; 4:5103. [PMID: 24869783 PMCID: PMC4037711 DOI: 10.1038/srep05103] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/09/2014] [Indexed: 01/31/2023] Open
Abstract
A radiofrequency electromagnetic field (RF-EMF) of 1800 MHz is widely used in mobile communications. However, the effects of RF-EMFs on cell biology are unclear. Embryonic neural stem cells (eNSCs) play a critical role in brain development. Thus, detecting the effects of RF-EMF on eNSCs is important for exploring the effects of RF-EMF on brain development. Here, we exposed eNSCs to 1800 MHz RF-EMF at specific absorption rate (SAR) values of 1, 2, and 4 W/kg for 1, 2, and 3 days. We found that 1800 MHz RF-EMF exposure did not influence eNSC apoptosis, proliferation, cell cycle or the mRNA expressions of related genes. RF-EMF exposure also did not alter the ratio of eNSC differentiated neurons and astrocytes. However, neurite outgrowth of eNSC differentiated neurons was inhibited after 4 W/kg RF-EMF exposure for 3 days. Additionally, the mRNA and protein expression of the proneural genes Ngn1 and NeuroD, which are crucial for neurite outgrowth, were decreased after RF-EMF exposure. The expression of their inhibitor Hes1 was upregulated by RF-EMF exposure. These results together suggested that 1800 MHz RF-EMF exposure impairs neurite outgrowth of eNSCs. More attention should be given to the potential adverse effects of RF-EMF exposure on brain development.
Collapse
|
42
|
Remaud S, Gothié JD, Morvan-Dubois G, Demeneix BA. Thyroid hormone signaling and adult neurogenesis in mammals. Front Endocrinol (Lausanne) 2014; 5:62. [PMID: 24808891 PMCID: PMC4009442 DOI: 10.3389/fendo.2014.00062] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/14/2014] [Indexed: 12/31/2022] Open
Abstract
The vital roles of thyroid hormone in multiple aspects of perinatal brain development have been known for over a century. In the last decades, the molecular mechanisms underlying effects of thyroid hormone on proliferation, differentiation, migration, synaptogenesis, and myelination in the developing nervous system have been gradually dissected. However, recent data reveal that thyroid signaling influences neuronal development throughout life, from early embryogenesis to the neurogenesis in the adult brain. This review deals with the latter phase and analyses current knowledge on the role of T3, the active form of thyroid hormone, and its receptors in regulating neural stem cell function in the hippocampus and the subventricular zone, the two principal sites harboring neurogenesis in the adult mammalian brain. In particular, we discuss the critical roles of T3 and TRα1 in commitment to a neuronal phenotype, a process that entails the repression of a number of genes notably that encoding the pluripotency factor, Sox2. Furthermore, the question of the relevance of thyroid hormone control of adult neurogenesis is considered in the context of brain aging, cognitive decline, and neurodegenerative disease.
Collapse
Affiliation(s)
- Sylvie Remaud
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d’Histoire Naturelle, Paris, France
| | - Jean-David Gothié
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d’Histoire Naturelle, Paris, France
| | - Ghislaine Morvan-Dubois
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d’Histoire Naturelle, Paris, France
| | - Barbara A. Demeneix
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d’Histoire Naturelle, Paris, France
- *Correspondence: Barbara A. Demeneix, UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d’Histoire Naturelle, Paris 75231, France e-mail:
| |
Collapse
|
43
|
Minato K, Tomimatsu T, Mimura K, Jugder O, Kakigano A, Kanayama T, Fujita S, Taniguchi Y, Kanagawa T, Endo M, Kimura T. Hypoxic preconditioning increases triiodothyronine (T3) level in the developing rat brain. Brain Res 2013; 1501:89-97. [PMID: 23376195 DOI: 10.1016/j.brainres.2013.01.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 01/11/2013] [Accepted: 01/20/2013] [Indexed: 11/24/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the major causes of neurodegeneration and mortality in the neonatal period. Although hypoxic preconditioning (HPC) provided strong neuroprotection against HIE in an animal model, the mechanism underlying this effect is not fully understood especially in the immature brain. Here, we investigated whether thyroid hormones (THs), especially triiodothyronine (T3), which are essential during normal brain development, contribute to the neuroprotective mechanisms of HPC by using an established model of HPC in neonatal rats. HPC treatment (8% O2 for 2.5h at 37°C) was performed in immature rats at postnatal day 6 (P6). Subsequently, we investigated the levels of THs, TH receptors (TRs) and type 2 and 3 deiodinase (D2 and D3) mRNA, and glutamate transporter 1 (GLT1) at 24h after HPC treatment, and myelin basic protein (MBP) at 6, 12 and 24h after HPC treatment. The HIE procedure was performed at 24h after HPC, and the neuroprotective effect of HPC was assessed via microtubule-associated protein 2 (MAP2) and MBP immunohistochemical staining at 14 days after HIE (P21). HPC treatment afforded marked neuroprotection at 14 days after HIE. The local level of T3 was upregulated 24h after HPC treatment in the developing rat brain, probably via the upregulation of D2. In addition, the expression of MBP and GLT1, which are the downstream protein of T3, were significantly increased 24h after HPC treatment. The present study indicates that thyroid hormones and their associated molecules may be involved in neuroprotective mechanisms of HPC during the developmental period.
Collapse
Affiliation(s)
- Kenji Minato
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Buss C, Entringer S, Wadhwa PD. Fetal programming of brain development: intrauterine stress and susceptibility to psychopathology. Sci Signal 2012; 5:pt7. [PMID: 23047922 DOI: 10.1126/scisignal.2003406] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetal brain is highly plastic and is not only receptive to but requires cues from its environment to develop properly. Based on an understanding of evolutionary biology, developmental plasticity, and life history theory, one can predict that stressors are an important environmental condition that may influence brain development. In fact, the available empirical evidence appears to support the notion that exposure to excess stress in intrauterine life has the potential to adversely affect short- and long-term neurodevelopmental outcomes with implications for altered susceptibility for mental health disorders in childhood and adult life. In this presentation, we provide a rationale for proposing that endocrine and inflammatory stress mediators are key candidate pathways for programming brain development. These mediators are responsive to a diverse set of intrauterine perturbations and alter key signaling pathways critical for brain development, including but not limited to mammalian target of rapamycin, Wnt (wingless), Sonic hedgehog, and reelin signaling. We suggest that recent advances in neuroimaging and other methods now afford us an unprecedented opportunity to advance our understanding of this important topic. Additionally, we provide empirical evidence from two recently published papers for fetal programming of human brain development. We conclude by suggesting some future directions for expanding this field of research.
Collapse
Affiliation(s)
- Claudia Buss
- Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA.
| | | | | |
Collapse
|