1
|
Li R, Tang Y, Wang H, Hu P, Yu L, Lv C, Zhang Y, Gerdes AM, Wang Y. Local DIO2 Elevation Is an Adaption in Malformed Cerebrovasculature. Circ Res 2025; 136:1010-1027. [PMID: 40130314 DOI: 10.1161/circresaha.124.325857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Cerebrovascular malformations are a pivotal cause of hemorrhage and neurological disability alongside lacking effective medication. Thyroid hormones (THs), including thyroxine and triiodothyronine, are essential for vascular development, yet whether they participate in malformed cerebrovascular pathology remains elusive. METHODS Single-cell transcriptome analysis characterized human cerebral cavernous malformations and brain arteriovenous malformations, 2 typical cerebrovascular malformation diseases. Adeno-associated virus-mediated Dio2 (iodothyronine deiodinase 2; an enzyme that converts thyroxine to active triiodothyronine) overexpression/knockdown or triiodothyronine/methimazole (an antithyroid drug) treatment was applied to mouse models of cerebral cavernous malformations (endothelial-specific Pdcd10 knockout mice, Pdcd10 endothelial-specific knockout [KO]) and brain arteriovenous malformations (endothelial-specific KrasG12D mutant mice, KrasG12D) to evaluate the involvement of DIO2 and TH signaling in cerebrovascular malformations. RESULTS TH signaling was markedly activated in fibroblasts of human cerebral cavernous malformation and arteriovenous malformation single-cell samples, accompanied by elevated DIO2 expression. Similar DIO2 upregulation was observed in cerebrovascular fibroblasts of Pdcd10 KO/KrasG12D mice and patient brain sections. Exogenous Dio2 or triiodothyronine replenishment effectively reduced brain hemorrhage, excessive ECM (extracellular matrix) remodeling, and vascular leakage in juvenile and adult male and female Pdcd10 KO/KrasG12D mice. In contrast, Dio2 silencing or TH inhibition deteriorated vascular anomalies. Mechanistically, transcription factor FOXK1 (forkhead box K1) was determined to interact with the DIO2 promoter region. The activation of fibroblast PI3K (phosphoinositide 3-kinase)-Akt (protein kinase B)-mTOR (mammalian target of rapamycin) signaling in Pdcd10 KO/KrasG12D mice triggered Foxk1 nuclear translocation to promote Dio2 transcription. Triiodothyronine treatment mitigated inflammatory infiltration, normalized mitochondrial morphology, and restored mitochondrial biogenesis in malformed brain vessels by activating the Pgc1a (peroxisome proliferator-activated receptor gamma coactivator 1-alpha)-Sod2 (superoxide dismutase 2)/Prdx3 (peroxiredoxin 3)/Gpx1 (glutathione peroxidase 1) axis to reduce reactive oxygen species accumulation. We also determined that the vascular repair effects of triiodothyronine were Pgc1a-dependent. CONCLUSIONS We delineate a novel DIO2-mediated adaption in malformed cerebrovasculature and conclude that targeting TH signaling may represent a potential therapy for cerebrovascular disorders.
Collapse
Affiliation(s)
- Ruofei Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Yushan Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Haiyue Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Pengyan Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Liang Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Cheng Lv
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
| | - A Martin Gerdes
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury (A.M.G.)
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (R.L., Y.T., H.W., P.H., L.Y., C.L., Y.Z., Y.W.)
- Central China Sub-center of the National Center for Cardiovascular Diseases, Zhengzhou, Henan, China (Y.W.)
- Institute of Cardiovascular Disease, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China (Y.W.)
| |
Collapse
|
2
|
Rao Z, Zhang Y, Zhu C. Association between thyroid hormone levels and early neurological deterioration in acute ischemic stroke. J Clin Neurosci 2025; 136:111253. [PMID: 40273598 DOI: 10.1016/j.jocn.2025.111253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Acute ischemic stroke (AIS) prognosis is significantly influenced by early neurological deterioration (END). Emerging evidence suggests that thyroid function, particularly free triiodothyronine (FT3) levels and the FT3/FT4 ratio, may play a crucial role in END occurrence. This study aimed to investigate the association between thyroid function parameters and END. METHODS This study included a total of 1767 AIS patients. Multivariable regression analysis was conducted to assess the associations between FT3, the FT3/FT4 ratio, and other thyroid function parameters with END. Subgroup analyses were performed to explore the potential moderating effects of age, smoking status, and sex on these relationships. The predictive performance of the models was evaluated using receiver operating characteristic (ROC) curves. RESULTS The FT3 levels (3.66 vs. 4.03 pmol/L, P < 0.001) and the FT3/FT4 ratio (0.28 vs. 0.30, P < 0.001) were significantly lower in the END group compared to the non-END group. Multivariable regression analysis identified a higher FT3/FT4 ratio as an independent protective factor against END (OR = 1.20, 95 % CI: 1.07-1.35, P = 0.0014). Subgroup analyses demonstrated that this protective effect was more pronounced inpatients older than 70 years, and non-smokers. Furthermore, the predictive model incorporating thyroid function parameters demonstrated certain predictive advantages over the CRP-based model (AUC = 0.688 vs. 0.624, P = 0.0036), suggesting its potential utility in predicting early neurological deterioration in AIS patients. CONCLUSIONS Lower FT3 levels and a reduced FT3/FT4 ratio are significantly associated with a higher risk of END. The FT3/FT4 ratio may serve as a valuable biomarker for risk stratification in AIS patients, underscoring the potential role of thyroid function in stroke management.
Collapse
Affiliation(s)
- Zichen Rao
- Department of Endocrinology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhenjiang, China.
| | - Yiming Zhang
- Department of Endocrinology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhenjiang, China.
| | - Chunyan Zhu
- Department of Endocrinology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhenjiang, China.
| |
Collapse
|
3
|
He Y, Li J, Chen Y, Ren B, Zhou Z, Liu J, Gao H, Li F, Li B, Liu L, Shen H. Expression and Function of Long Non-coding RNA in Endemic Cretinism. Mol Neurobiol 2025; 62:1770-1787. [PMID: 39031326 DOI: 10.1007/s12035-024-04358-3] [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: 03/02/2024] [Accepted: 07/09/2024] [Indexed: 07/22/2024]
Abstract
Endemic cretinism (EC) is one of the most severe iodine deficiency disorders, leading to typical symptoms such as neurodevelopmental impairments or mental deficits. In addition to environmental factors, the pathogenesis of its genetic contribution remains unclear. The study revealed the differential expression profiles of long non-coding RNA(lncRNA) and messenger RNA(mRNA) based on high-throughput RNA-seq. GO and KEGG analyses were used to annotate the function and pathway of differentially expressed (DE) mRNA and co-expressed mRNA. The protein-protein interaction(PPI) network was established. The expression levels of three lncRNAs and six mRNAs were validated by quantitative real-time PCR analysis (qRT-PCR) and subjected to correlation analysis. Compared to controls, a total of 864 lncRNAs and 393 mRNAs were differentially expressed. The PPI network had 149 nodes and 238 edges, and three key protein-coding genes were observed. Levels of LINC01220 and target mRNA IDO1 were statistically elevated in EC patients. Differentially expressed lncRNA may be a new potential player in EC. LINC01220 and IDO1 might interact with each other to participate in EC. The biological process of regulation of postsynaptic membrane potential and the Rap1 signaling pathway might exert a regulating role in the pathophysiological process of EC. Our findings could provide more theoretical and experimental evidence for investigating the pathophysiological mechanisms of EC.
Collapse
Affiliation(s)
- Yanhong He
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Jianshuang Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Heilongjiang Province 163319, Daqing City, People's Republic of China
| | - Yun Chen
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Bingxuan Ren
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Zheng Zhou
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Jinjin Liu
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Haiyan Gao
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Fan Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Baoxiang Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Lixiang Liu
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Hongmei Shen
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
| |
Collapse
|
4
|
Lin W, Liao L, Ling L, Luo H, Jiang Y, Li X, Yao Y, Yang P. Combined effects of co-exposure to microcystin-LR and polystyrene microplastics on growth, brain pathology and thyroid hormone homeostasis in adult zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117855. [PMID: 39919588 DOI: 10.1016/j.ecoenv.2025.117855] [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: 12/07/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
The concurrent presence of algal blooms and microplastics pollution in natural water bodies poses a novel threat. However, the joint effects of microcystin-LR (MCLR) in combination with polystyrene microplastics (PSMPs) on the thyroid endocrine system of adult fish remains unclear. In our study, male zebrafish (Danio rerio) were exposed to environmentally relevant concentrations of MCLR alone (0, 0.8, 4, 20 μg/L) and a mix of MCLR and PSMPs (100 μg/L) for 60 days. Alterations in brain histology, thyroid hormone (TH) levels, and the transcription levels of hypothalamic-pituitary-thyroid (HPT)-axis genes were used to assess the thyroid function. In the MCLR-only treatment groups, we observed mild brain tissue damage characterized by glial scarring and hyperemia. The presence of PSMPs exacerbated the brain damage cause by MCLR, resulting in more pronounced ventriculomegaly and hyperemia. No significant changes in whole-body thyroxine (T4) and triiodothyronine (T3) levels were observed in the MCLR-only groups, while a significant decrease was noted in the groups co-exposed to MCLR and PSMPs. Additionally, significant alterations in crh, tshβ, ttr, trα, and trβ expression levels in the combined exposure groups provided further confirmation that MCLR and PSMPs jointly cause thyroid endocrine disruption. Our findings suggest that the fish can trigger a compensatory mechanism to maintain thyroid hormone homeostasis in response to environmentally relevant concentrations of MCLR. However, the presence of PSMPs disrupts this self-regulatory equilibrium, thereby exacerbates the thyroid endocrine disruption cause by MCLR in zebrafish.
Collapse
Affiliation(s)
- Wang Lin
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China; Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Science, Yunnan University, Kunming 650500, China; Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Changde 415000, China
| | - Ling Liao
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China
| | - Ling Ling
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China
| | - Huimin Luo
- School of Chemistry and Materials Science, Hunan Agricultural University, Changsha 410125, China
| | - Ying Jiang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China
| | - Xinru Li
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China
| | - Yilong Yao
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China
| | - Pinhong Yang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China; College of Agricultural and Forestry Science and Technology, Hunan Applied Technology University, Changde 415000, China; Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Changde 415000, China.
| |
Collapse
|
5
|
Huang FBQ, Liao K, Sun YN, Li ZH, Zhang YR, Liao PF, Jiang SY, Zhu ZY, Chen DY, Lei Y, Liu SP, Lin YN, Zhuang ZK. Cross-species single-cell transcriptomics reveals neuronal similarities and heterogeneity in amniote pallium. Zool Res 2025; 46:193-208. [PMID: 39846196 DOI: 10.24272/j.issn.2095-8137.2024.102] [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] [Indexed: 01/24/2025] Open
Abstract
The amniote pallium, a vital component of the forebrain, exhibits considerable evolutionary divergence across species and mediates diverse functions, including sensory processing, memory formation, and learning. However, the relationships among pallial subregions in different species remain poorly characterized, particularly regarding the identification of homologous neurons and their transcriptional signatures. In this study, we utilized single-nucleus RNA sequencing to examine over 130 000 nuclei from the macaque ( Macaca fascicularis) neocortex, complemented by datasets from humans ( Homo sapiens), mice ( Mus musculus), zebra finches ( Taeniopygia guttata), turtles ( Chrysemys picta bellii), and lizards ( Pogona vitticeps), enabling comprehensive cross-species comparison. Results revealed transcriptomic conservation and species-specific distinctions within the amniote pallium. Notable similarities were observed among cell subtypes, particularly within PVALB + inhibitory neurons, which exhibited species-preferred subtypes. Furthermore, correlations between pallial subregions and several transcription factor candidates were identified, including RARB, DLX2, STAT6, NR3C1, and THRB, with potential regulatory roles in gene expression in mammalian pallial neurons compared to their avian and reptilian counterparts. These results highlight the conserved nature of inhibitory neurons, remarkable regional divergence of excitatory neurons, and species-specific gene expression and regulation in amniote pallial neurons. Collectively, these findings provide valuable insights into the evolutionary dynamics of the amniote pallium.
Collapse
Affiliation(s)
- Fu-Bao-Qian Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Yu-Nong Sun
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Zi-Hao Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Yan-Ru Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Ping-Fang Liao
- BGI Research, Hangzhou, Zhejiang 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si-Yuan Jiang
- BGI Research, Hangzhou, Zhejiang 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhi-Yong Zhu
- BGI Research, Hangzhou, Zhejiang 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Duo-Yuan Chen
- BGI Research, Hangzhou, Zhejiang 310030, China
- BGI Research, Shenzhen, Guangdong 518083, China
| | - Ying Lei
- BGI Research, Hangzhou, Zhejiang 310030, China
- BGI Research, Shenzhen, Guangdong 518083, China
| | - Shi-Ping Liu
- BGI Research, Hangzhou, Zhejiang 310030, China
- BGI Research, Shenzhen, Guangdong 518083, China
| | - You-Ning Lin
- BGI Research, Hangzhou, Zhejiang 310030, China
- BGI Research, Shenzhen, Guangdong 518083, China. E-mail:
| | - Zhen-Kun Zhuang
- BGI Research, Hangzhou, Zhejiang 310030, China
- BGI Research, Shenzhen, Guangdong 518083, China. E-mail:
| |
Collapse
|
6
|
Mooradian AD, Haas MJ. Role of Thyroid Hormone in Neurodegenerative Disorders of Older People. Cells 2025; 14:140. [PMID: 39851568 PMCID: PMC11763745 DOI: 10.3390/cells14020140] [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: 12/20/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Thyroid dysfunction is associated with a number of neuropsychiatric manifestations. Cognitive decline is a common feature of hypothyroidism and clinical or subclinical hyperthyroidism. In addition, there is a significant association between thyroid hormone (TH) levels and the degree of cognitive impairment in Parkinson's disease (PD). The pathophysiology of TH-related neurodegeneration include changes in the blood-brain barrier, increased cellular stress, altered processing of β-amyloid precursor protein and the effect of TH on neuronal cell viability. The neurotoxicity of TH is partially mediated by the thyroid hormone responsive protein (THRP). This protein is 83% homologous to mouse c-Abl-interacting protein-2 (Abi2), a c-Abl-modulating protein with tumor suppressor activity. In cell cultures, increasing THRP expression either with TH treatment or exogenously through transfecting neuronal or PC 12 cells causes cell necrosis. The expression of exogenous THRP in other cells such as the colonic epithelial cell line Caco-2 and the glial cell line U251 has no effect on cell viability. The effect of THRP on cell viability is not modulated by c-Abl tyrosine kinase. The causal relationship between specific biochemical perturbations in cerebral tissue and thyroid dysfunction remains to be elucidated.
Collapse
Affiliation(s)
- Arshag D. Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street, Jacksonville, FL 32209, USA;
| | | |
Collapse
|
7
|
Beyoğlu D, Hamberg P, IJzerman NS, Mathijssen RHJ, Idle JR. New metabolic insights into the mechanism of ifosfamide encephalopathy. Biomed Pharmacother 2025; 182:117773. [PMID: 39693904 DOI: 10.1016/j.biopha.2024.117773] [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: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024] Open
Abstract
Ifosfamide causes neurotoxicity, including sometimes fatal encephalopathy, in a small number of patients. Why and how this occurs is not fully understood. It is generally believed that N-dechloroethylation of ifosfamide to 2-chloroacetaldehyde is the cause. A total of 61 patients were investigated, 49 who received ifosfamide and pazopanib and 12 treated with ifosfamide and sunitinib. Plasmas were analyzed by ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOFMS) and by gas chromatography-mass spectrometry (GC-MS). Neurotoxicity occurred in 25/61 patients, including four with encephalopathy. UPLC-QTOFMS revealed that N-dechloroethylation was unlikely to be the cause but did divulge in plasma that 2-chloroethylamine, 3-phosphoserine, uridine 3'-diphosphate 5'-diphosphate, Cer(d16:1/17:0), Cer(d16:0/16:0), and thyroxine were associated with encephalopathy. GC-MS analysis showed that palmitic, oleic and stearic acids increased significantly in plasma only in nonencephalopathic patients, suggesting impaired long-chain fatty acid oxidation but an alternative metabolic pathway in encephalopathic patients. Glycine, alanine, serine, glutamate and 5-oxoproline all decreased significantly only in encephalopathic plasmas, signifying increased de novo GSH synthesis. Taken together, these findings indicate three new putative mechanisms of ifosfamide encephalopathy: (i) failure to convert 3-phosphoserine to serine due to inhibition of O-phosphoserine phosphohydratase; (ii) failure to incorporate ceramides into cerebrosides and (iii) oxidative injury of the cerebral cortex requiring de novo GSH synthesis.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Biomedical Research, University of Bern, Bern, Switzerland; Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA.
| | - Paul Hamberg
- Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands.
| | | | | | - Jeffrey R Idle
- Department of Biomedical Research, University of Bern, Bern, Switzerland; Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA.
| |
Collapse
|
8
|
Sagliocchi S, Restolfer F, Cossidente A, Dentice M. The key roles of thyroid hormone in mitochondrial regulation, at interface of human health and disease. J Basic Clin Physiol Pharmacol 2024; 35:231-240. [PMID: 39023546 PMCID: PMC11522957 DOI: 10.1515/jbcpp-2024-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/06/2024] [Indexed: 07/20/2024]
Abstract
Mitochondria are highly plastic and dynamic organelles long known as the powerhouse of cellular bioenergetics, but also endowed with a critical role in stress responses and homeostasis maintenance, supporting and integrating activities across multifaced cellular processes. As a such, mitochondria dysfunctions are leading causes of a wide range of diseases and pathologies. Thyroid hormones (THs) are endocrine regulators of cellular metabolism, regulating intracellular nutrients fueling of sugars, amino acids and fatty acids. For instance, THs regulate the balance between the anabolism and catabolism of all the macro-molecules, influencing energy homeostasis during different nutritional conditions. Noteworthy, not only most of the TH-dependent metabolic modulations act via the mitochondria, but also THs have been proved to regulate the mitochondrial biosynthesis, dynamics and function. The significance of such an interplay is different in the context of specific tissues and strongly impacts on cellular homeostasis. Thus, a comprehensive understanding of THs-dependent mitochondrial functions and dynamics is required to develop more precise strategies for targeting mitochondrial function. Herein, we describe the mechanisms of TH-dependent metabolic regulation with a focus on mitochondrial action, in different tissue contexts, thus providing new insights for targeted modulation of mitochondrial dynamics.
Collapse
Affiliation(s)
- Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| | - Alessandro Cossidente
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
9
|
Yu X, Guo J, Song Y, Wei B, Shi Y, Zhao Y, Zhao Z, Gao Q, Wang B, Sun M. HDAC1/2/3-mediated downregulation of neurogranin is involved in cognitive impairment in offspring exposed to maternal subclinical hypothyroidism. FASEB J 2024; 38:e23736. [PMID: 38865202 DOI: 10.1096/fj.202400389r] [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: 02/20/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Subclinical hypothyroidism (SCH) in pregnancy is the most common form of thyroid dysfunction in pregnancy, which can affect fetal nervous system development and increase the risk of neurodevelopmental disorders after birth. However, the mechanism of the effect of maternal subclinical hypothyroidism on fetal brain development and behavioral phenotypes is still unclear and requires further study. In this study, we constructed a mouse model of maternal subclinical hypothyroidism by exposing dams to drinking water containing 50 ppm propylthiouracil (PTU) during pregnancy and found that its offspring were accompanied by severe cognitive deficits by behavioral testing. Mechanistically, gestational SCH resulted in the upregulation of protein expression and activity of HDAC1/2/3 in the hippocampus of the offspring. ChIP analysis revealed that H3K9ac on the neurogranin (Ng) promoter was reduced in the hippocampus of the offspring of SCH, with a significant reduction in Ng protein, leading to reduced expression levels of synaptic plasticity markers PSD95 (a membrane-associated protein in the postsynaptic density) and SYN (synaptophysin, a specific marker for presynaptic terminals), and impaired synaptic plasticity. In addition, administration of MS-275 (an HDAC1/2/3-specific inhibitor) to SCH offspring alleviated impaired synaptic plasticity and cognitive dysfunction in offspring. Thus, our study suggests that maternal subclinical hypothyroidism may mediate offspring cognitive dysfunction through the HDAC1/2/3-H3K9ac-Ng pathway. Our study contributes to the understanding of the signaling mechanisms underlying maternal subclinical hypothyroidism-mediated cognitive impairment in the offspring.
Collapse
Affiliation(s)
- Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Guo
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yueyang Song
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zejun Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic, Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
10
|
Roghani AK, Garcia RI, Roghani A, Reddy A, Khemka S, Reddy RP, Pattoor V, Jacob M, Reddy PH, Sehar U. Treating Alzheimer's disease using nanoparticle-mediated drug delivery strategies/systems. Ageing Res Rev 2024; 97:102291. [PMID: 38614367 DOI: 10.1016/j.arr.2024.102291] [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: 10/30/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
The administration of promising medications for the treatment of neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) is significantly hampered by the blood-brain barrier (BBB). Nanotechnology has recently come to light as a viable strategy for overcoming this obstacle and improving drug delivery to the brain. With a focus on current developments and prospects, this review article examines the use of nanoparticles to overcome the BBB constraints to improve drug therapy for AD The potential for several nanoparticle-based approaches, such as those utilizing lipid-based, polymeric, and inorganic nanoparticles, to enhance drug transport across the BBB are highlighted. To shed insight on their involvement in aiding effective drug transport to the brain, methods of nanoparticle-mediated drug delivery, such as surface modifications, functionalization, and particular targeting ligands, are also investigated. The article also discusses the most recent findings on innovative medication formulations encapsulated within nanoparticles and the therapeutic effects they have shown in both preclinical and clinical testing. This sector has difficulties and restrictions, such as the need for increased safety, scalability, and translation to clinical applications. However, the major emphasis of this review aims to provide insight and contribute to the knowledge of how nanotechnology can potentially revolutionize the worldwide treatment of NDDs, particularly AD, to enhance clinical outcomes.
Collapse
Affiliation(s)
- Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Frenship High School, Lubbock, TX 79382, USA.
| | - Ricardo Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ali Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Aananya Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; University of South Florida, Tampa, FL 33620, USA.
| | - Michael Jacob
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Services, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
Tesfaye E, Getnet M, Anmut Bitew D, Adugna DG, Maru L. Brain functional connectivity in hyperthyroid patients: systematic review. Front Neurosci 2024; 18:1383355. [PMID: 38726033 PMCID: PMC11080614 DOI: 10.3389/fnins.2024.1383355] [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: 02/07/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Functional connectivity (FC) is the correlation between brain regions' activities, studied through neuroimaging techniques like fMRI. It helps researchers understand brain function, organization, and dysfunction. Hyperthyroidism, characterized by high serum levels of free thyroxin and suppressed thyroid stimulating hormone, can lead to mood disturbance, cognitive impairment, and psychiatric symptoms. Excessive thyroid hormone exposure can enhance neuronal death and decrease brain volume, affecting memory, attention, emotion, vision, and motor planning. METHODS We conducted thorough searches across Google Scholar, PubMed, Hinari, and Science Direct to locate pertinent articles containing original data investigating FC measures in individuals diagnosed with hyperthyroidism. RESULTS The systematic review identified 762 articles, excluding duplicates and non-matching titles and abstracts. Four full-text articles were included in this review. In conclusion, a strong bilateral hippocampal connection in hyperthyroid individuals suggests a possible neurobiological influence on brain networks that may affect cognitive and emotional processing. SYSTEMATIC REVIEW REGISTRATION PROSPERO, CRD42024516216.
Collapse
Affiliation(s)
- Ephrem Tesfaye
- Department of Biomedical Sciences, Madda Walabu University Goba Referral Hospital, Bale-Robe, Ethiopia
| | - Mihret Getnet
- Department of Human Physiology, School of Medicine, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
- Department of Epidemiology and Biostatistics, Institute of Public Health, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Desalegn Anmut Bitew
- Department of Reproductive Health, Institute of Public Health, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Dagnew Getnet Adugna
- Department of Anatomy, School of Medicine, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Lemlemu Maru
- Department of Human Physiology, School of Medicine, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
12
|
Fang Y, Wan JP, Wang Z, Song SY, Zhang CX, Yang L, Zhang QY, Yan CY, Wu FY, Lu SY, Sun F, Han B, Zhao SX, Dong M, Song HD. Deficiency of the HGF/Met pathway leads to thyroid dysgenesis by impeding late thyroid expansion. Nat Commun 2024; 15:3165. [PMID: 38605010 PMCID: PMC11009301 DOI: 10.1038/s41467-024-47363-9] [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: 06/17/2023] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
The mechanisms of bifurcation, a key step in thyroid development, are largely unknown. Here we find three zebrafish lines from a forward genetic screening with similar thyroid dysgenesis phenotypes and identify a stop-gain mutation in hgfa and two missense mutations in met by positional cloning from these zebrafish lines. The elongation of the thyroid primordium along the pharyngeal midline was dramatically disrupted in these zebrafish lines carrying a mutation in hgfa or met. Further studies show that MAPK inhibitor U0126 could mimic thyroid dysgenesis in zebrafish, and the phenotypes are rescued by overexpression of constitutively active MEK or Snail, downstream molecules of the HGF/Met pathway, in thyrocytes. Moreover, HGF promotes thyrocyte migration, which is probably mediated by downregulation of E-cadherin expression. The delayed bifurcation of the thyroid primordium is also observed in thyroid-specific Met knockout mice. Together, our findings reveal that HGF/Met is indispensable for the bifurcation of the thyroid primordium during thyroid development mediated by downregulation of E-cadherin in thyrocytes via MAPK-snail pathway.
Collapse
Affiliation(s)
- Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Jia-Ping Wan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cao-Xu Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qian-Yue Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chen-Yan Yan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Sang-Yu Lu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng Sun
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bing Han
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Mei Dong
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
13
|
Tani N, Ikeda T, Ishikawa T. Effects of Prolactin on Brain Neurons under Hypoxia. Life (Basel) 2024; 14:152. [PMID: 38276281 PMCID: PMC10817236 DOI: 10.3390/life14010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
The levels and potential role of prolactin (PRL) in the brain under conditions of acute systemic hypoxia were examined, focusing on the accumulation of PRL in cerebrospinal fluid (CSF) and its effects on neuronal activity and injury. The amount of PRL in the brain was investigated using brain tissues from forensic autopsy cases. We counted the number of neurites that formed in human primary neurons (HNs) after the addition of PRL. Furthermore, HNs supplemented with PRL or triiodothyronine (T3) were exposed to hypoxic conditions, and the dead cells were counted. The results showed correlations between brain PRL and CSF PRL levels. Additionally, PRL accumulation in the brain was observed in cases of asphyxia. In vitro experimental findings indicated increased neurite formation in the HNs treated with PRL. Moreover, both PRL and T3 demonstrated neuroprotective effects against hypoxia-induced neuronal cell death, with PRL showing stronger neuroprotective potential than T3. These results suggest that PRL accumulates in the brain during hypoxia, potentially influences neuronal activity, and exhibits neuroprotective properties against hypoxia-induced neuronal injury.
Collapse
Affiliation(s)
- Naoto Tani
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
- Forensic Autopsy Section, Medico-Legal Consultation and Postmortem Investigation Support Center, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan
| | - Tomoya Ikeda
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
| | - Takaki Ishikawa
- Department of Legal Medicine, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan; (T.I.); (T.I.)
- Forensic Autopsy Section, Medico-Legal Consultation and Postmortem Investigation Support Center, 1-4-3 Asahi-machi, Abeno, Osaka 545-8585, Japan
| |
Collapse
|
14
|
Yatoo MI, Bahader GA, Beigh SA, Khan AM, James AW, Asmi MR, Shah ZA. Neuroprotection or Sex Bias: A Protective Response to Traumatic Brain Injury in the Females. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:906-916. [PMID: 37592792 DOI: 10.2174/1871527323666230817102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023]
Abstract
Traumatic brain injury (TBI) is a major healthcare problem and a common cause of mortality and morbidity. Clinical and preclinical research suggests sex-related differences in short- and longterm outcomes following TBI; however, males have been the main focus of TBI research. Females show a protective response against TBI. Female animals in preclinical studies and women in clinical trials have shown comparatively better outcomes against mild, moderate, or severe TBI. This reflects a favorable protective nature of the females compared to the males, primarily attributed to various protective mechanisms that provide better prognosis and recovery in the females after TBI. Understanding the sex difference in the TBI pathophysiology and the underlying mechanisms remains an elusive goal. In this review, we provide insights into various mechanisms related to the anatomical, physiological, hormonal, enzymatic, inflammatory, oxidative, genetic, or mitochondrial basis that support the protective nature of females compared to males. Furthermore, we sought to outline the evidence of multiple biomarkers that are highly potential in the investigation of TBI's prognosis, pathophysiology, and treatment and which can serve as objective measures and novel targets for individualized therapeutic interventions in TBI treatment. Implementations from this review are important for the understanding of the effect of sex on TBI outcomes and possible mechanisms behind the favorable response in females. It also emphasizes the critical need to include females as a biological variable and in sufficient numbers in future TBI studies.
Collapse
Affiliation(s)
- Mohammad I Yatoo
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Shafayat A Beigh
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Adil M Khan
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Maleha R Asmi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
15
|
Mahajan C, Prabhakar H, Bilotta F. Endocrine Dysfunction After Traumatic Brain Injury: An Ignored Clinical Syndrome? Neurocrit Care 2023; 39:714-723. [PMID: 36788181 PMCID: PMC10689524 DOI: 10.1007/s12028-022-01672-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/28/2022] [Indexed: 02/16/2023]
Abstract
Traumatic brain injury (TBI) incurs substantial health and economic burden, as it is the leading reason for death and disability globally. Endocrine abnormalities are no longer considered a rare complication of TBI. The reported prevalence is variable across studies, depending on the time frame of injury, time and type of testing, and variability in hormonal values considered normal across different studies. The present review reports evidence on the endocrine dysfunction that can occur after TBI. Several aspects, including the pathophysiological mechanisms, clinical consequences/challenges (in the acute and chronic phases), screening and diagnostic workup, principles of therapeutic management, and insights on future directions/research agenda, are presented. The management of hypopituitarism following TBI involves hormonal replacement therapy. It is essential for health care providers to be aware of this complication because at times, symptoms may be subtle and may be mistaken to be caused by brain injury itself. There is a need for stronger evidence for establishing recommendations for optimum management so that they can be incorporated as standard of care in TBI management.
Collapse
Affiliation(s)
- Charu Mahajan
- Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Hemanshu Prabhakar
- Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Federico Bilotta
- Department of Anesthesiology, Policlinico UmbertoI Hospital, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
16
|
Abo-Zaid OA, Moawed FS, Taha EF, Ahmed ESA, Kawara RS. Melissa officinalis extract suppresses endoplasmic reticulum stress-induced apoptosis in the brain of hypothyroidism-induced rats exposed to γ-radiation. Cell Stress Chaperones 2023; 28:709-720. [PMID: 37368180 PMCID: PMC10746611 DOI: 10.1007/s12192-023-01363-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
The purpose of this study was to demonstrate the neuroprotective effect of Melissa officinalis extract (MEE) against brain damage associated with hypothyroidism induced by propylthiouracil (PTU) and/or γ-radiation (IR) in rats. Hypothyroidism induction and/or exposure to IR resulted in a significant decrease in the serum levels of T3 and T4 associated with increased levels of lipid peroxidation end product, malondialdehyde (MDA), and nitrites (NO) in the brain tissue homogenate. Also, hypothyroidism and /or exposure to IR markedly enhance the endoplasmic reticulum stress by upregulating the gene expressions of the protein kinase RNA-like endoplasmic reticulum kinase (PERK), activated transcription factor 6 (ATF6), endoplasmic reticulum-associated degradation (ERAD), and CCAAT/enhancer-binding protein homologous protein (CHOP) in the brain tissue homogenate associated with a proapoptotic state which indicated by the overexpression of Bax, BCl2, and caspase-12 that culminates in brain damage. Meanwhile, the PTU and /or IR-exposed rats treated with MEE reduced oxidative stress and ERAD through ATF6. Also, the MEE treatment prevented the Bax and caspase-12 gene expression from increasing. This treatment in hypothyroid animals was associated with neuronal protection as indicated by the downregulation in the gene expressions of the microtubule-associated protein tau (MAPT) and amyloid precursor protein (APP) in the brain tissue. Furthermore, the administration of MEE ameliorates the histological structure of brain tissue. In conclusion, MEE might prevent hypothyroidism-induced brain damage associated with oxidative stress and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Omayma Ar Abo-Zaid
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med, Benha University, Moshtohor, Banha, Egypt
| | - Fatma Sm Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
- Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787, Egypt.
| | - Eman Fs Taha
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Esraa S A Ahmed
- Radiation Biology Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ragaa Sm Kawara
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med, Benha University, Moshtohor, Banha, Egypt
| |
Collapse
|
17
|
Shapira G, Israel-Elgali I, Grad M, Avnat E, Rachmany L, Sarne Y, Shomron N. Hippocampal differential expression underlying the neuroprotective effect of delta-9-tetrahydrocannabinol microdose on old mice. Front Neurosci 2023; 17:1182932. [PMID: 37534036 PMCID: PMC10393280 DOI: 10.3389/fnins.2023.1182932] [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: 03/12/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Delta-9-tetrahydrocannabinol (THC) is the primary psychoactive compound of the cannabis plant and an exogenous ligand of the endocannabinoid system. In previous studies, we demonstrated that a single microdose of THC (0.002 mg/kg, 3-4 orders of magnitude lower than the standard dose for rodents) exerts distinct, long-term neuroprotection in model mice subjected to acute neurological insults. When administered to old, healthy mice, the THC microdose induced remarkable long-lasting (weeks) improvement in a wide range of cognitive functions, including significant morphological and biochemical brain alterations. To elucidate the mechanisms underlying these effects, we analyzed the gene expression of hippocampal samples from the model mice. Samples taken 5 days after THC treatment showed significant differential expression of genes associated with neurogenesis and brain development. In samples taken 5 weeks after treatment, the transcriptional signature was shifted to that of neuronal differentiation and survival. This study demonstrated the use of hippocampal transcriptome profiling in uncovering the molecular basis of the atypical, anti-aging effects of THC microdose treatment in old mice.
Collapse
Affiliation(s)
- Guy Shapira
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Ifat Israel-Elgali
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Meitar Grad
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eden Avnat
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lital Rachmany
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yosef Sarne
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
18
|
Rastoldo G, Tighilet B. Thyroid Axis and Vestibular Physiopathology: From Animal Model to Pathology. Int J Mol Sci 2023; 24:9826. [PMID: 37372973 DOI: 10.3390/ijms24129826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/02/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
A recent work of our group has shown the significant effects of thyroxine treatment on the restoration of postural balance function in a rodent model of acute peripheral vestibulopathy. Based on these findings, we attempt to shed light in this review on the interaction between the hypothalamic-pituitary-thyroid axis and the vestibular system in normal and pathological situations. Pubmed database and relevant websites were searched from inception through to 4 February 2023. All studies relevant to each subsection of this review have been included. After describing the role of thyroid hormones in the development of the inner ear, we investigated the possible link between the thyroid axis and the vestibular system in normal and pathological conditions. The mechanisms and cellular sites of action of thyroid hormones on animal models of vestibulopathy are postulated and therapeutic options are proposed. In view of their pleiotropic action, thyroid hormones represent a target of choice to promote vestibular compensation at different levels. However, very few studies have investigated the relationship between thyroid hormones and the vestibular system. It seems then important to more extensively investigate the link between the endocrine system and the vestibule in order to better understand the vestibular physiopathology and to find new therapeutic leads.
Collapse
Affiliation(s)
- Guillaume Rastoldo
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
| | - Brahim Tighilet
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
- GDR Vertige CNRS Unité GDR2074, 13331 Marseille, France
| |
Collapse
|
19
|
Thomas J, Sairoz, Jose A, Poojari VG, Shetty S, K SP, Prabhu R V K, Rao M. Role and Clinical Significance of Monocarboxylate Transporter 8 (MCT8) During Pregnancy. Reprod Sci 2023; 30:1758-1769. [PMID: 36595209 PMCID: PMC10229697 DOI: 10.1007/s43032-022-01162-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023]
Abstract
The review aims to summarize the available research focusing on the importance of monocarboxylate transporter (MCT8) in thyroid hormone trafficking across the placenta and fetal development. A systematic search was carried out in PubMed; studies available in English related to "monocarboxylate transporter", "adverse pregnancy", "fetal development," and "thyroid hormone" were identified and assessed. The references within the resulting articles were manually searched. MCT8 is a highly active and selective thyroid hormone transporter that facilitates the cellular uptake of triiodothyronine (T3), thyroxine (T4), reverse triiodothyronine (rT3), and diiodothyronine (T2) in different tissues. MCT8 is expressed in the placenta from the first trimester onwards, allowing the transport of thyroid hormone from mother to fetus. Mutations in MCT8 cause an X-linked disorder known as Allan-Herndon-Dudley syndrome (AHDS), characterized by severe psychomotor impairment and peripheral thyrotoxicosis. Hence, any maternal thyroid dysfunction may cause severe consequences for the fetus and newborn. Further research regarding MCT8 gene expression, polymorphic variation, and adverse pregnancy outcomes must be done to establish that MCT8 is a novel prognostic marker for the early detection of pregnancy-related complications.
Collapse
Affiliation(s)
- Jinsu Thomas
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sairoz
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anmi Jose
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vidyashree G Poojari
- Department of Reproductive Medicine and Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sahana Shetty
- Department of Endocrinology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shama Prasada K
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Krishnananda Prabhu R V
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
20
|
Zhang F, Li X, Wei Y. Selenium and Selenoproteins in Health. Biomolecules 2023; 13:biom13050799. [PMID: 37238669 DOI: 10.3390/biom13050799] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Selenium is a trace mineral that is essential for health. After being obtained from food and taken up by the liver, selenium performs various physiological functions in the body in the form of selenoproteins, which are best known for their redox activity and anti-inflammatory properties. Selenium stimulates the activation of immune cells and is important for the activation of the immune system. Selenium is also essential for the maintenance of brain function. Selenium supplements can regulate lipid metabolism, cell apoptosis, and autophagy, and have displayed significant alleviating effects in most cardiovascular diseases. However, the effect of increased selenium intake on the risk of cancer remains unclear. Elevated serum selenium levels are associated with an increased risk of type 2 diabetes, and this relationship is complex and nonlinear. Selenium supplementation seems beneficial to some extent; however, existing studies have not fully explained the influence of selenium on various diseases. Further, more intervention trials are needed to verify the beneficial or harmful effects of selenium supplementation in various diseases.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuelian Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
21
|
Identification of adolescent patients with depression via assessment of the niacin skin flushing response. J Affect Disord 2023; 324:69-76. [PMID: 36521667 DOI: 10.1016/j.jad.2022.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Depressive disorder (DD) affects approximately 20 % of adolescents worldwide, but it is underdiagnosed due to the lack of objective biomarkers. Niacin skin flushing response (NSFR) is an objective and noninvasive biomarker of adult depression; however, its effectiveness has not been assessed in adolescents. METHODS This study included 198 adolescents with 50 % healthy controls (HC). Linear mixed-effects model and multiple linear regression analyses were performed to assess differences in NSFR between the DD and HC groups. Logistic regression models based on NSFR were constructed, and the area under curve (AUC) was calculated to evaluate the performance of models. Spearman correlations were calculated to assess the relationships between NSFR and disease duration and hormone levels associated with puberty. RESULTS Adolescents with DD displayed significantly attenuated and delayed NSFR compared to HC. NSFR effectively distinguished DD patients from HC with AUC values of 0.719 (sensitivity = 0.844) and 0.721 (sensitivity = 0.829) determined in the discovery and validation sets, respectively. Within the DD group, the maximum degree of NSFR was negatively correlated with the disease duration (r = -0.28, p = 0.011), and the overall degree of NSFR was positively associated with prolactin (r = 0.29, p = 0.039) and thyroxine (r = 0.29, p = 0.027) levels. LIMITATIONS Future investigations will be necessary to confirm our results in an independent sample set. CONCLUSIONS This study provides the first evidence of the utility of NSFR as an objective auxiliary diagnostic biomarker for adolescent depression. It provides new clues to understand the pathophysiology of the disease, and helps promote precise diagnosis, treatment, and prognostic evaluation of adolescent depression.
Collapse
|
22
|
Thyroid Hormone Transporters MCT8 and OATP1C1 Are Expressed in Pyramidal Neurons and Interneurons in the Adult Motor Cortex of Human and Macaque Brain. Int J Mol Sci 2023; 24:ijms24043207. [PMID: 36834621 PMCID: PMC9965431 DOI: 10.3390/ijms24043207] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Monocarboxylate transporter 8 (MCT8) and organic anion transporter polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters that play an important role in the availability of TH for neural cells, allowing their proper development and function. It is important to define which cortical cellular subpopulations express those transporters to explain why MCT8 and OATP1C1 deficiency in humans leads to dramatic alterations in the motor system. By means of immunohistochemistry and double/multiple labeling immunofluorescence in adult human and monkey motor cortices, we demonstrate the presence of both transporters in long-projection pyramidal neurons and in several types of short-projection GABAergic interneurons in both species, suggesting a critical position of these transporters for modulating the efferent motor system. MCT8 is present at the neurovascular unit, but OATP1C1 is only present in some of the large vessels. Both transporters are expressed in astrocytes. OATP1C1 was unexpectedly found, only in the human motor cortex, inside the Corpora amylacea complexes, aggregates linked to substance evacuation towards the subpial system. On the basis of our findings, we propose an etiopathogenic model that emphasizes these transporters' role in controlling excitatory/inhibitory motor cortex circuits in order to understand some of the severe motor disturbances observed in TH transporter deficiency syndromes.
Collapse
|
23
|
Dong X, Deng L, Yao S, Wu W, Cao J, Sun L, Bai Y, Li H, Weng X, Ren H, Ren W. Protective effects of curcumin against thyroid hormone imbalance after gas explosion-induced traumatic brain injury via activation of the hypothalamic-pituitary-thyroid axis in male rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:74619-74631. [PMID: 35641736 DOI: 10.1007/s11356-022-20943-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
Gas explosion (GE)-induced traumatic brain injury (TBI) can affect thyroid hormone (TH) homeostasis in miners. This study evaluated the effects of hepatic transthyretin and hypothalamic-pituitary-thyroid (HPT) axis on thyroids and explored the protective effect and mechanism of curcumin on GE-induced TBI. Thirty rats were randomly divided into three groups (10 per group): first group (control group)-rats received GE treatment once; second group (GE group)-rats received GE treatment (200 m from the source of the explosion once); third group (GE + Cur group)-rats received curcumin (Cur) by lavage at a dose of 100 mg/kg/day once every other day for 7 days after receiving GE. After GE, the pathological changes were analyzed by hemotoxylin and eosin staining, and the levels of serum reactive oxygen species (ROS), urine iodine (UI), THs, nuclear factor-kappa B (NF-κB), superoxide dismutase (SOD), glutathione peroxidase (Gpx), and malondialdehyde (MDA) were analyzed using ELISA. Expression of proteins in the HPT axis of rats was examined by immunohistochemistry and Western blotting. We found that GE could induce pathologic changes in rat thyroid and liver. Serum levels of THs, NF-κB and serum redox state became unbalanced in rats after GE. GE could inhibit the biosynthesis and biotransformation of THs by affecting key HPT axis proteins. Additionally, GE reduced the level of hepatic transthyretin. Serum THs levels and thyroid sections were almost recovered to normal after curcumin treatment. The aforementioned key HPT axis proteins in the curcumin group showed opposite expression trends. In summary, GE affected THs balance while curcumin can protect against these injury effects by affecting TH biosynthesis, biotransformation, and transport, and inducing oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Xinwen Dong
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Lvfei Deng
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Sanqiao Yao
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Weidong Wu
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Lei Sun
- Institute of Toxicology, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yichun Bai
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Haibin Li
- Department of Environmental and Occupational Health, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiaogang Weng
- Institute of Trauma and Orthopedics, Xinxiang Medical University, Xinxiang, 453000, Henan, China
| | - Houcheng Ren
- Department of Human Resources, Sanquan College, Xinxiang Medical University, Xinxiang, 453000, Henan, China
| | - Wenjie Ren
- Institutes of Health Central Plains, Xinxiang Medical University, 601 Jinsui Street , Xinxiang, 453003, Henan, China.
| |
Collapse
|
24
|
Zhang NN, Zhang Y, Wang ZZ, Chen NH. Connexin 43: insights into candidate pathological mechanisms of depression and its implications in antidepressant therapy. Acta Pharmacol Sin 2022; 43:2448-2461. [PMID: 35145238 PMCID: PMC9525669 DOI: 10.1038/s41401-022-00861-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/06/2022] [Indexed: 11/09/2022]
Abstract
Major depressive disorder (MDD), a chronic and recurrent disease characterized by anhedonia, pessimism or even suicidal thought, remains a major chronic mental concern worldwide. Connexin 43 (Cx43) is the most abundant connexin expressed in astrocytes and forms the gap junction channels (GJCs) between astrocytes, the most abundant and functional glial cells in the brain. Astrocytes regulate neurons' synaptic strength and function by expressing receptors and regulating various neurotransmitters. Astrocyte dysfunction causes synaptic abnormalities, which are related to various mood disorders, e.g., depression. Increasing evidence suggests a crucial role of Cx43 in the pathogenesis of depression. Depression down-regulates Cx43 expression in humans and rats, and dysfunction of Cx43 also induces depressive behaviors in rats and mice. Recently Cx43 has received considerable critical attention and is highly implicated in the onset of depression. However, the pathological mechanisms of depression-like behavior associated with Cx43 still remain ambiguous. In this review we summarize the recent progress regarding the underlying mechanisms of Cx43 in the etiology of depression-like behaviors including gliotransmission, metabolic disorders, and neuroinflammation. We also discuss the effects of antidepressants (monoamine antidepressants and ketamine) on Cx43. The clarity of the candidate pathological mechanisms of depression-like behaviors associated with Cx43 and its potential pharmacological roles for antidepressants will benefit the exploration of a novel antidepressant target.
Collapse
Affiliation(s)
- Ning-Ning Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
25
|
Abe K, Li J, Liu YY, Brent GA. Thyroid Hormone-mediated Histone Modification Protects Cortical Neurons From the Toxic Effects of Hypoxic Injury. J Endocr Soc 2022; 6:bvac139. [PMID: 36817622 PMCID: PMC9562813 DOI: 10.1210/jendso/bvac139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Indexed: 11/19/2022] Open
Abstract
Context Thyroid hormone has been shown to have a protective role in neuronal injury, although the mechanisms have not been established. The cellular response to stress that promotes adaptation and survival has been shown to involve epigenetic modifications. Objective We hypothesized that the neuroprotective role of thyroid hormone was associated with epigenetic modifications of histone proteins. We used hypoxic neurons as a model system for hypoxia-induced brain injury. Methods Mouse primary cortical neurons were exposed to 0.2% oxygen for 7 hours, with or without, treatment with triiodothyronine (T3). We analyzed the expression of histone-modifying enzymes by RNA-seq and the post-translationally modified histone 3 proteins by enzyme-linked immunosorbent assay (ELISA) and Western blot. Results We found that methylation of H3K27, associated with inactive promoters, was highly induced in hypoxic neurons, and this histone methylation was reduced by T3 treatment. H3K4 methylation is the hallmark of active promoters. The expression of 3 (Set1db, Kmta2c, and Kmt2e) out of 6 H3K4 methyltransferases was downregulated by hypoxia and expression was restored by T3 treatment. H3K4me3 protein, measured by ELISA, was increased 76% in T3-treated hypoxic neurons compared with the levels without T3 treatment. H3K56ac plays a critical role in transcription initiation and was markedly increased in T3-treated hypoxic neurons compared with those without T3 treatment, indicating stimulation of gene transcription. Additionally, T3 treatment restored hypoxia-induced downregulation of histone acetyltransferase, Kat6a, Kat6b, and Crebbp, which function as transcription factors. Conclusion These findings indicate that T3 treatment mitigates hypoxia-induced histone modifications and protects neurons from hypoxia-induced injury.
Collapse
Affiliation(s)
- Kiyomi Abe
- Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jianrong Li
- Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Yan Yun Liu
- Correspondence: Yan-Yun Liu, PhD, Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA. ; or Gregory A. Brent, MD, Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| | - Gregory A Brent
- Correspondence: Yan-Yun Liu, PhD, Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA. ; or Gregory A. Brent, MD, Division of Endocrinology, Diabetes and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| |
Collapse
|
26
|
Qin Q, Wang T, Xu Z, Liu S, Zhang H, Du Z, Wang J, Wang Y, Wang Z, Yuan S, Wu J, He W, Wang C, Yan X, Wang Y, Jiang X. Ectoderm-derived frontal bone mesenchymal stem cells promote traumatic brain injury recovery by alleviating neuroinflammation and glutamate excitotoxicity partially via FGF1. Stem Cell Res Ther 2022; 13:341. [PMID: 35883153 PMCID: PMC9327213 DOI: 10.1186/s13287-022-03032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. Methods A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. Results Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2β. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. Conclusions Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03032-6.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China.,Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Ting Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhangzhen Du
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jianing Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Yadi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenning Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shanshan Yuan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jiamei Wu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Changzhen Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China.
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
27
|
Pan H, Wang Y, Wang X, Yan C. Dimethyl fumarate improves cognitive impairment by enhancing hippocampal brain-derived neurotrophic factor levels in hypothyroid rats. BMC Endocr Disord 2022; 22:188. [PMID: 35869475 PMCID: PMC9306081 DOI: 10.1186/s12902-022-01086-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dimethyl fumarate (DMF) is an effective drug for multiple sclerosis and can improve the cognitive dysfunction caused by streptozotocin, but the effect on cognitive dysfunction caused by hypothyroidism is unclear. METHODS After the hypothyroidism rat model induced by propylthiouracil, we gave rats 25 mg/kg DMF by gavage. The body weight during model building and administration was recorded. The levels of T4 and T3 in serum were detected by an automatic biochemical analyzer. Morris water maze test was used to detect the effect of DMF on cognitive learning ability. The effect of DMF on Nissl bodies in the brain tissue was evaluated by Nissl staining. The mRNA and protein levels of BDNF in brain tissue were detected by quantitative reverse transcription-polymerase chain reaction and Western blot. The degrees of p-AKT/AKT and p-CREB/CREB in brain tissue were detected by Western blot. RESULTS After DMF treatment, the body weight of hypothyroid rats recovered, and the levels of T3 and T4 in the serum were ameliorated. DMF also reduced the escape latency and distance traveled, and increased the swim speed. The number of Nissl bodies and expression of BDNF, p-AKT/AKT, and p-CREB/CREB in the brain tissue were increased after DMF treatment. CONCLUSION DMF improved the cognitive dysfunction of hypothyroid rats by increasing the level of BDNF in the brain tissue of hypothyroid rats.
Collapse
Affiliation(s)
- Haiyan Pan
- Department of Endocrinology, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310000, China
| | - Yanbo Wang
- Department of Endocrinology, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310000, China
| | - Xiaowei Wang
- Department of Endocrinology, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310000, China
| | - Ci Yan
- Departments of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, 310000, China.
| |
Collapse
|
28
|
Abstract
Iodine is a micronutrient needed for the production of thyroid hormones, which regulate metabolism, growth, and development. Iodine deficiency or excess may alter the thyroid hormone synthesis. The potential effects on infant development depend on the degree, timing, and duration of exposure. The iodine requirement is particularly high during infancy because of elevated thyroid hormone turnover. Breastfed infants rely on iodine provided by human milk, but the iodine concentration in breast milk is determined by the maternal iodine intake. Diets in many countries cannot provide sufficient iodine, and deficiency is prevented by iodine fortification of salt. However, the coverage of iodized salt varies between countries. Epidemiological data suggest large differences in the iodine intake in lactating women, infants, and toddlers worldwide, ranging from deficient to excessive intake. In this review, we provide an overview of the current knowledge and recent advances in the understanding of iodine nutrition and its association with thyroid function in lactating women, infants, and toddlers. We discuss risk factors for iodine malnutrition and the impact of targeted intervention strategies on these vulnerable population groups. We highlight the importance of appropriate definitions of optimal iodine nutrition and the need for more data assessing the risk of mild iodine deficiency for thyroid disorders during the first 2 years in life.
Collapse
Affiliation(s)
- Maria Andersson
- Nutrition Research Unit, University Children’s Hospital Zurich, CH-8032 Zürich, Switzerland
| | - Christian P Braegger
- Nutrition Research Unit, University Children’s Hospital Zurich, CH-8032 Zürich, Switzerland
| |
Collapse
|
29
|
Fang Y, Dang P, Liang Y, Zhao D, Wang R, Xi Y, Zhang D, Wang W, Shan Z, Teng W, Teng X. Histological, functional and transcriptomic alterations in the juvenile hippocampus in a mouse model of thyroid hormone resistance. Eur Thyroid J 2022; 11:e210097. [PMID: 35262510 PMCID: PMC9066571 DOI: 10.1530/etj-21-0097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Proper thyroid hormone signaling via the TRα1 nuclear receptor is required for normal neurodevelopmental processes. The specific downstream mechanisms mediated by TRα1 that impact brain development remain to be investigated. METHODS In this study, the structure, function and transcriptome of hippocampal tissue in a mouse model expressing the first RTHα mutation discovered in a patient, THRA E403X, were analyzed. RNAscope was used to visualize the spatial and temporal expression of Thra1 mRNA in the hippocampus of WT mice, which is corresponding to THRA1 mRNA in humans. The morphological structure was analyzed by Nissl staining, and the synaptic transmission was analyzed on the basis of long-term potentiation. The Morris water maze test and the zero maze test were used to evaluate the behavior. RNA-seq and quantitative real-time PCR were used to analyze the differentially expressed genes (DEGs) of the hippocampal tissues in the mouse model expressing the Thra E403X mutation. RESULTS The juvenile mutant Thra E403X mice presented with delayed neuronal migration, disordered neuronal distribution, and decreased synaptic plasticity. A total of 754 DEGs, including 361 upregulated genes and 393 downregulated genes, were identified by RNA-seq. DEG-enriched Gene Ontology (GO) and KEGG pathways were associated with PI3K-Akt signaling, ECM-receptor interaction, neuroactive ligand-receptor interaction, and a range of immune-related pathways. 25 DEGs were validated by qPCR. CONCLUSIONS The ThraE403X mutation results in histological and functional abnormalities, as well as transcriptomic alterations in the juvenile mouse hippocampus. This study of the ThraE403X mutant offers new insights into the biological cause of RTHα-associated neurological diseases.
Collapse
Affiliation(s)
- Yingxin Fang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Pingping Dang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yue Liang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Defa Zhao
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Ranran Wang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yue Xi
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Dan Zhang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Wei Wang
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Correspondence should be addressed to X Teng:
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrine, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, People’s Republic of China
- Correspondence should be addressed to X Teng:
| |
Collapse
|
30
|
Takagishi M, Aleogho BM, Okumura M, Ushida K, Yamada Y, Seino Y, Fujimura S, Nakashima K, Shindo A. Nutritional control of thyroid morphogenesis through gastrointestinal hormones. Curr Biol 2022; 32:1485-1496.e4. [PMID: 35196509 DOI: 10.1016/j.cub.2022.01.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 01/27/2023]
Abstract
Developing animals absorb nutrients either through the placenta or from ingested food; however, the mechanisms by which embryos use external nutrients for individual organ morphogenesis remain to be elucidated. In this study, we assessed nutrient-dependent thyroid follicle morphogenesis in Xenopus laevis and investigated the role of secreted gastrointestinal (GI) hormones post-feeding. We found that feeding triggers thyroid follicle formation, and the thyroid cells showed transient inactivation of cell proliferation after feeding. In addition, the thyroid cells with multi-lumina were frequently observed in the fed tadpoles. The expression of the particular GI hormone incretin, glucose-dependent insulinotropic polypeptide (GIP), responded to feeding in the intestines of Xenopus tadpoles. Inhibition of dipeptidyl peptidase 4 (Dpp4), a degradative enzyme of incretin, increased the size of the thyroid follicles by facilitating follicular lumina connection, whereas inhibition of the sodium-glucose cotransporter (SGLT) reversed the effects of Dpp4 inhibition. Furthermore, injection of GIP peptide in unfed tadpoles initiated thyroid follicle formation-without requiring feeding-and injection of an incretin receptor antagonist suppressed follicle enlargement in the fed tadpoles. Lastly, GIP receptor knockout in neonatal mice showed smaller follicles in the thyroid, suggesting that the GI hormone-dependent thyroid morphogenesis is conserved in mammals. In conclusion, our study links external nutrients to thyroid morphogenesis and provides new insights into the function of GI hormone as a regulator of organ morphology in developing animals.
Collapse
Affiliation(s)
- Maki Takagishi
- Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Binta Maria Aleogho
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Masako Okumura
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Kaori Ushida
- Division for Medical Research Engineering, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yuichiro Yamada
- Kansai Electric Power Medical Research Institute, 2-1-7 Fukushima, Fukushima-ku, Osaka 553-0003, Japan
| | - Yusuke Seino
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Sayoko Fujimura
- Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Kaoru Nakashima
- Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Asako Shindo
- Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan; Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
31
|
Lv J, Li Y, Chen J, Li R, Bao C, Ding Z, Ren W, Du Z, Wang S, Huang Y, Wang QN. Maternal exposure to bis(2-ethylhexyl) phthalate during the thyroid hormone-dependent stage induces persistent emotional and cognitive impairment in middle-aged offspring mice. Food Chem Toxicol 2022; 163:112967. [PMID: 35354077 DOI: 10.1016/j.fct.2022.112967] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/10/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
Abstract
Prenatal DEHP exposure can cause offspring neurodevelopmental toxicity, but the persistent effects of such exposure window are unclear. This study aimed to investigate the lasting neurobehavioral impact of DEHP on offspring following early exposure from GD9.5 (fetal neural tube closure) to GD16.5 (fetal thyroxin, TH, synthesis). Data showed maternal exposure to DEHP during the thyroid hormone-dependent stage induced a range of neurobehavioral phenotypic changes in adult and middle-aged mice, including anxiety, depression and cognitive impairment. Significant reductions in free TH, TH transporters, and TH metabolic enzyme deiodinase II (D2) were observed in the fetal brain, whereas D3 was elevated, indicating that TH signaling disruption was caused by in utero exposure. Gene expression analyses suggested the expression levels of the TH receptors Trα1, Trβ1 and their downstream target, brain-derived neurotrophic factor, were significantly attenuated, which may partially explain the mechanisms of neurodevelopmental impairment. This study provides new evidence of the persistent effects of sex-specific neurodevelopmental impairment due to in utero DEHP exposure, possibly through damage to the fetal brain TH signaling systems that causes lifelong brain damage. These results further suggest a profound neurobehavioral toxicity of DEHP that may be programmed during early developmental stage exposure and manifested later in life.
Collapse
Affiliation(s)
- Jia Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Yanling Li
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Jianrong Chen
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Rong Li
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Chao Bao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Zheng Ding
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Wenqiang Ren
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Zhiping Du
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; Jinhua Center for Disease Control and Prevention, Jinhua, Zhejiang, China
| | - Sheng Wang
- Center for Scientific Research of Anhui Medical University, Hefei, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| | - Qu-Nan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China.
| |
Collapse
|
32
|
L-Thyroxine Improves Vestibular Compensation in a Rat Model of Acute Peripheral Vestibulopathy: Cellular and Behavioral Aspects. Cells 2022; 11:cells11040684. [PMID: 35203333 PMCID: PMC8869897 DOI: 10.3390/cells11040684] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
Unilateral vestibular lesions induce a vestibular syndrome, which recovers over time due to vestibular compensation. The therapeutic effect of L-Thyroxine (L-T4) on vestibular compensation was investigated by behavioral testing and immunohistochemical analysis in a rat model of unilateral vestibular neurectomy (UVN). We demonstrated that a short-term L-T4 treatment reduced the vestibular syndrome and significantly promoted vestibular compensation. Thyroid hormone receptors (TRα and TRβ) and type II iodothyronine deiodinase (DIO2) were present in the vestibular nuclei (VN), supporting a local action of L-T4. We confirmed the T4-induced metabolic effects by demonstrating an increase in the number of cytochrome oxidase-labeled neurons in the VN three days after the lesion. L-T4 treatment modulated glial reaction by decreasing both microglia and oligodendrocytes in the deafferented VN three days after UVN and increased cell proliferation. Survival of newly generated cells in the deafferented vestibular nuclei was not affected, but microglial rather than neuronal differentiation was favored by L-T4 treatment.
Collapse
|
33
|
Luo W, Yang Z, Zhang W, Zhou D, Guo X, Wang S, He F, Wang Y. Quantitative Proteomics Reveals the Dynamic Pathophysiology Across Different Stages in a Rat Model of Severe Traumatic Brain Injury. Front Mol Neurosci 2022; 14:785938. [PMID: 35145378 PMCID: PMC8821658 DOI: 10.3389/fnmol.2021.785938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/30/2021] [Indexed: 11/30/2022] Open
Abstract
Background Severe traumatic brain injury (TBI) has become a global health problem and causes a vast worldwide societal burden. However, distinct mechanisms between acute and subacute stages have not been systemically revealed. The present study aimed to identify differentially expressed proteins in severe TBI from the acute to subacute phase. Methods Sixty Sprague Dawley (SD) rats were randomly divided into sham surgery and model groups. The severe TBI models were induced by the controlled cortical impact (CCI) method. We evaluated the neurological deficits through the modified neurological severity score (NSS). Meanwhile, H&E staining and immunofluorescence were performed to assess the injured brain tissues. The protein expressions of the hippocampus on the wounded side of CCI groups and the same side of Sham groups were analyzed by the tandem mass tag-based (TMT) quantitative proteomics on the third and fourteenth days. Then, using the gene ontology (GO), Kyoto encyclopedia of genes and genomes (KEGG), and protein–protein interaction (PPI), the shared and stage-specific differentially expressed proteins (DEPs) were screened, analyzed, and visualized. Eventually, target proteins were further verified by Western blotting (WB). Results In the severe TBI, the neurological deficits always exist from the acute stage to the subacute stage, and brain parenchyma was dramatically impaired in either period. Of the significant DEPs identified, 312 were unique to the acute phase, 76 were specific to the subacute phase, and 63 were shared in both. Of the 375 DEPs between Sham-a and CCI-a, 240 and 135 proteins were up-regulated and down-regulated, respectively. Of 139 DEPs, 84 proteins were upregulated, and 55 were downregulated in the Sham-s and CCI-s. Bioinformatics analysis revealed that the differential pathophysiology across both stages. One of the most critical shared pathways is the complement and coagulation cascades. Notably, three pathways associated with gastric acid secretion, insulin secretion, and thyroid hormone synthesis were only enriched in the acute phase. Amyotrophic lateral sclerosis (ALS) was significantly enriched in the subacute stage. WB experiments confirmed the reliability of the TMT quantitative proteomics results. Conclusion Our findings highlight the same and different pathological processes in the acute and subacute phases of severe TBI at the proteomic level. The results of potential protein biomarkers might facilitate the design of novel strategies to treat TBI.
Collapse
Affiliation(s)
- Weikang Luo
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaoyu Yang
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Dan Zhou
- Periodical Office, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaohang Guo
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Shunshun Wang
- Postpartum Health Care Department, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Feng He
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yang Wang,
| |
Collapse
|
34
|
Ariyani W, Miyazaki W, Amano I, Koibuchi N. Involvement of integrin αvβ3 in thyroid hormone-induced dendritogenesis. Front Endocrinol (Lausanne) 2022; 13:938596. [PMID: 36072926 PMCID: PMC9441609 DOI: 10.3389/fendo.2022.938596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/29/2022] [Indexed: 11/15/2022] Open
Abstract
Activation and/or modulation of the membrane-associated receptors plays a critical role in brain development. Thyroid hormone (TH) acts on both nuclear receptors (thyroid hormone receptor, TR) and membrane-associated receptors, particularly integrin αvβ3 in neurons and glia. Integrin αvβ3-mediated signal transduction mediates various cellular events during development including morphogenesis, migration, synaptogenesis, and intracellular metabolism. However, the involvement of integrin αvβ3-mediated TH action during brain development remains poorly understood. Thus, we examined the integrin αvβ3-mediated effects of TH (T3, T4, and rT3) in the neurons and astrocytes using primary cerebellar culture, astrocyte-enriched culture, Neuro-2A clonal cells, and co-culture of neurons and astrocytes. We found that TH augments dendrite arborization of cerebellar Purkinje cells. This augmentation was suppressed by knockdown of integrin αvβ3, as well as TRα and TRβ. A selective integrin αvβ3 antagonist, LM609, was also found to suppress TH-induced arborization. However, whether this effect was a direct action of TH on Purkinje cells or due to indirect actions of other cells subset such as astrocytes was not clarified. To further study neuron-specific molecular mechanisms, we used Neuro-2A clonal cells and found TH also induces neurite growth. TH-induced neurite growth was reduced by co-exposure with LM609 or knockdown of TRα, but not TRβ. Moreover, co-culture of Neuro-2A and astrocytes also increased TH-induced neurite growth, indicating astrocytes may be involved in neuritogenesis. TH increased the localization of synapsin-1 and F-actin in filopodia tips. TH exposure also increased phosphorylation of FAK, Akt, and ERK1/2. Phosphorylation was suppressed by co-exposure with LM609 and TRα knockdown. These results indicate that TRs and integrin αvβ3 play essential roles in TH-induced dendritogenesis and neuritogenesis. Furthermore, astrocytes-neuron communication via TR-dependent and TR-independent signaling through membrane receptors and F-actin are required for TH-induced neuritogenesis.
Collapse
Affiliation(s)
- Winda Ariyani
- International Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- *Correspondence: Winda Ariyani, ; Noriyuki Koibuchi,
| | - Wataru Miyazaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Science, Hirosaki, Aomori, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- *Correspondence: Winda Ariyani, ; Noriyuki Koibuchi,
| |
Collapse
|
35
|
Sabatino L, Federighi G, Del Seppia C, Lapi D, Costagli C, Scuri R, Iervasi G. Thyroid hormone deiodinases response in brain of spontaneausly hypertensive rats after hypotensive effects induced by mandibular extension. Endocrine 2021; 74:100-107. [PMID: 33761105 DOI: 10.1007/s12020-021-02684-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/05/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE The deiodinases activate or inactivate the thyroid hormones (TH) in virtually all tissues in both physiological and pathological conditions. The three deiodinases, DIO1, DIO2, and DIO3, have different catalytic functions and regulate TH tissue distribution. The aim of the present study was to evaluate the modulation of gene expression of the deiodinases and TH transporters and protein levels of DIO1 in parietal and frontal areas of cerebral cortex of spontaneously hypertensive rats (SHRs), after two successive mandibular extensions (ME). METHODS ME was performed on anesthetized rats by a dilatator appropriately designed and real-time PCR and western blotting techniques were employed for gene expression and protein level study. RESULTS Mean blood pressure (MBP) significantly decreased in 2ME-treated rats when compared to sham-operated rats (p < 0.001) and this decrease lasted for the entire observation period. In gene expression analysis, in 2ME-treated rats we did not observe any significant variation of DIO1 and DIO3 with respect to the sham-operated rats. Differently, DIO2 gene expression significantly increased in frontal area of 2ME-treated rats, with respect to sham-operated rats (p < 0.01). Furthermore, in parietal area, protein levels of DIO1 in 2ME-treated rats were significantly higher than in sham-operated rats (p < 0.01). Moreover MCT8 and OATP1C1 both resulted significantly higher (p < 0.05 and p < 0.001) in sham frontal cortex. CONCLUSION In summary, our data on SHRs, while confirming the hypotensive effect of two MEs, show that the treatment also solicits the three deiodinases production in the cerebral cortex.
Collapse
Affiliation(s)
| | - Giuseppe Federighi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Dominga Lapi
- Department of Biology, University of Pisa, Pisa, Italy
| | - Chiara Costagli
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rossana Scuri
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | |
Collapse
|
36
|
Transcriptome and Methylome Analysis Reveal Complex Cross-Talks between Thyroid Hormone and Glucocorticoid Signaling at Xenopus Metamorphosis. Cells 2021; 10:cells10092375. [PMID: 34572025 PMCID: PMC8468809 DOI: 10.3390/cells10092375] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Most work in endocrinology focus on the action of a single hormone, and very little on the cross-talks between two hormones. Here we characterize the nature of interactions between thyroid hormone and glucocorticoid signaling during Xenopus tropicalis metamorphosis. Methods: We used functional genomics to derive genome wide profiles of methylated DNA and measured changes of gene expression after hormonal treatments of a highly responsive tissue, tailfin. Clustering classified the data into four types of biological responses, and biological networks were modeled by system biology. Results: We found that gene expression is mostly regulated by either T3 or CORT, or their additive effect when they both regulate the same genes. A small but non-negligible fraction of genes (12%) displayed non-trivial regulations indicative of complex interactions between the signaling pathways. Strikingly, DNA methylation changes display the opposite and are dominated by cross-talks. Conclusion: Cross-talks between thyroid hormones and glucocorticoids are more complex than initially envisioned and are not limited to the simple addition of their individual effects, a statement that can be summarized with the pseudo-equation: TH ∙ GC > TH + GC. DNA methylation changes are highly dynamic and buffered from genome expression.
Collapse
|
37
|
Congenital hypothyroidism impairs spine growth of dentate granule cells by downregulation of CaMKIV. Cell Death Discov 2021; 7:143. [PMID: 34127648 PMCID: PMC8203692 DOI: 10.1038/s41420-021-00530-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022] Open
Abstract
Congenital hypothyroidism (CH), a common neonatal endocrine disorder, can result in cognitive deficits if delay in diagnose and treatment. Dentate gyrus (DG) is the severely affected subregion of the hippocampus by the CH, where the dentate granule cells (DGCs) reside in. However, how CH impairs the cognitive function via affecting DGCs and the underlying mechanisms are not fully elucidated. In the present study, the CH model of rat pups was successfully established, and the aberrant dendrite growth of the DGCs and the impaired cognitive behaviors were observed in the offspring. Transcriptome analysis of hippocampal tissues following rat CH successfully identified that calcium/calmodulin-dependent protein kinase IV (CaMKIV) was the prominent regulator involved in mediating deficient growth of DGC dendrites. CaMKIV was shown to be dynamically regulated in the DG subregion of the rats following drug-induced CH. Interference of CaMKIV expression in the primary DGCs significantly reduced the spine density of dendrites, while addition of T3 to the primary DGCs isolated from CH pups could facilitate the spine growth of dendrites. Insights into relevant mechanisms revealed that CH-mediated CaMKIV deficiency resulted in the significant decrease of phosphorylated CREB in DGCs, in association with the abnormality of dendrites. Our results have provided a distinct cell type in hippocampus that is affected by CH, which would be beneficial for the treatment of CH-induced cognitive deficiency.
Collapse
|
38
|
Abstract
Thyroid hormone is essential for brain development and brain function in the adult. During development, thyroid hormone acts in a spatial and temporal-specific manner to regulate the expression of genes essential for normal neural cell differentiation, migration, and myelination. In the adult brain, thyroid hormone is important for maintaining normal brain function. Thyroid hormone excess, hyperthyroidism, and thyroid hormone deficiency, hypothyroidism, are associated with disordered brain function, including depression, memory loss, impaired cognitive function, irritability, and anxiety. Adequate thyroid hormone levels are required for normal brain function. Thyroid hormone acts through a cascade of signaling components: activation and inactivation by deiodinase enzymes, thyroid hormone membrane transporters, and nuclear thyroid hormone receptors. Additionally, the hypothalamic-pituitary-thyroid axis, with negative feedback of thyroid hormone on thyrotropin-releasing hormone (TRH) and thyroid-stimulating hormone (TSH) secretion, regulates serum thyroid hormone levels in a narrow range. Animal and human studies have shown both systemic and local reduction in thyroid hormone availability in neurologic disease and after brain trauma. Treatment with thyroid hormone and selective thyroid hormone analogs has resulted in a reduction in injury and improved recovery. This article will describe the thyroid hormone signal transduction pathway in the brain and the role of thyroid hormone in the aging brain, neurologic diseases, and the protective role when administered after traumatic brain injury. © 2021 American Physiological Society. Compr Physiol 11:1-21, 2021.
Collapse
Affiliation(s)
- Yan-Yun Liu
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gregory A Brent
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
39
|
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
|
40
|
Wang S, Zhang J, Zhang S, Shi F, Feng D, Feng X. Exposure to Melamine cyanuric acid in adolescent mice caused emotional disorder and behavioral disorder. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 211:111938. [PMID: 33476844 DOI: 10.1016/j.ecoenv.2021.111938] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Melamine cyanuric acid (MCA) is a flame retardant linked by hydrogen bonds between melamine and cyanuric acid. MCA is used in an excellent series of phosphorus and nitrogen flame retardants. MCA can harm the kidney, liver, testis, and spleen cells. However, the effects of MCA on the emotions and behaviour of adolescent mice have not yet been investigated. In this article, male mice were exposed to MCA at 10, 20, and 40 mg/kg for four weeks. MCA exposure resulted in enhanced mouse locomotor and nocturnal activity. We also observed anxiety-like and depression-like behaviours. Moreover, after MCA exposure, the serum concentrations of thyroid-related hormones were changed, and the mRNA levels were affected. In short, MCA exposure can cause behavioural and emotion disorders.
Collapse
Affiliation(s)
- Sijie Wang
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Jingwen Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Shaozhi Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Feifei Shi
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Daofu Feng
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China.
| | - Xizeng Feng
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin 300071, China.
| |
Collapse
|
41
|
Zhang Z, Yu J, Wang P, Lin L, Liu R, Zeng R, Ma H, Zhao Y. iTRAQ-based proteomic profiling reveals protein alterations after traumatic brain injury and supports thyroxine as a potential treatment. Mol Brain 2021; 14:25. [PMID: 33504361 PMCID: PMC7839205 DOI: 10.1186/s13041-021-00739-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/16/2021] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury (TBI) is a primary cause of disability and death across the world. Previously, RNA analysis was widely used to study the pathophysiological mechanisms underlying TBI; however, the relatively low correlation between the transcriptome and proteome revealed that RNA transcription abundance does not reliably predict protein abundance, which led to the emergence of proteomic research. In this study, an iTRAQ proteomics approach was applied to detect protein alterations after TBI on a large scale. A total of 3937 proteins were identified, and 146 proteins were significantly changed after TBI. Moreover, 23 upregulated proteins were verified by parallel reaction monitoring (PRM), and fold changes in 16 proteins were consistent with iTRAQ outcomes. Transthyretin (Ttr) upregulation has been demonstrated at the transcriptional level, and this study further confirmed this at the protein level. After treatment with thyroxine (T4), which is transported by Ttr, the effects of T4 on neuronal histopathology and behavioral performance were determined in vivo (TBI + T4 group). Brain edema was alleviated, and the integrity of the blood brain barrier (BBB) improved. Escape latency in the Morris water maze (MWM) declined significantly compared with the group without T4 treatment. Modified neurological severity scores (mNSS) of the TBI + T4 group decreased from day 1 to day 7 post-TBI compared with the TBI + saline group. These results indicate that T4 treatment has potential to alleviate pathologic and behavioral abnormalities post-TBI. Protein alterations after T4 treatment were also detected by iTRAQ proteomics. Upregulation of proteins like Lgals3, Gfap and Apoe after TBI were reversed by T4 treatment. GO enrichment showed T4 mainly affected intermediate filament organization, cholesterol transportation and axonal regeneration. In summary, iTRAQ proteomics provides information about the impact of TBI on protein alterations and yields insight into underlying mechanisms and pathways involved in TBI and T4 treatment. Finally, Ttr and other proteins identified by iTRAQ may become potential novel treatment targets post-TBI.
Collapse
Affiliation(s)
- Zhongxiang Zhang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Pengcheng Wang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Lian Lin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Ruining Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Rong Zeng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Haoli Ma
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| |
Collapse
|
42
|
Doshi M, Watanabe S, Natori Y, Hosoyamada M, Hirashima-Akae Y. Triiodothyronine Aggravates Global Cerebral Ischemia-Reperfusion Injury in Mice. Biol Pharm Bull 2021; 44:1824-1831. [PMID: 34853265 DOI: 10.1248/bpb.b21-00424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Thyroid hormones (THs) have been suggested to play an important role in both physiological and pathological events in the central nervous system. Hypothyroidism, which is characterized by low levels of serum THs, has been associated with aggravation of ischemic neuronal injuries in stroke patients. We hypothesized that administration of T3, the main active form of THs, may attenuate the ischemic neuronal injuries. In mice, global cerebral ischemia (GCI), which is induced by transient occlusion of the bilateral common carotid artery, causes neuronal injuries by inducing neuronal death and activating inflammatory responses after reperfusion in the hippocampus. In this study, we examined the effect of T3 administration on DNA fragmentation induced by neuronal death and the activation of inflammatory cells such as astrocytes and microglia in the hippocampus following GCI. The content of nucleosomes generated by DNA fragmentation in the hippocampus was increased by GCI and further increased by T3 administration. The protein expression levels of glial fibrillary acidic protein (GFAP), an astrocytic marker, and Ionized calcium binding adaptor protein 1 (Iba1), a microglial marker, in the hippocampus were also increased by GCI and further increased by T3 administration. The levels of T3 in both the serum and hippocampus were elevated by T3 administration. Our results indicate that T3 administration aggravates GCI-reperfusion injury in mice. There may be an increased risk of aggravation of ischemic stroke by the excessive elevation of T3 levels during the drug treatment of hypothyroidism.
Collapse
Affiliation(s)
- Masaru Doshi
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University
| | - Shiro Watanabe
- Division of Nutritional Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Yujin Natori
- Department of Legal Medicine and Bioethics, Nagoya University Graduate School of Medicine
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University
| | | |
Collapse
|
43
|
Peltier MR, Fassett MJ, Chiu VY, Getahun D. Maternal Hypothyroidism Increases the Risk of Attention-Deficit Hyperactivity Disorder in the Offspring. Am J Perinatol 2021; 38:191-201. [PMID: 33086392 DOI: 10.1055/s-0040-1717073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE This study aimed to determine if hypothyroidism prior to, or during, pregnancy increases the risk of attention-deficit hyperactivity disorder (ADHD) in the child and how the association may be modified by preterm birth, sex of the child, and race-ethnicity. STUDY DESIGN Data were abstracted from linked maternal-child medical records. Incidence rate differences (IRDs), adjusted hazard ratios (aHRs), and their 95% confidence intervals (CIs) were estimated to evaluate the association of maternal hypothyroidism with childhood ADHD risk. Stratified analyses were used to evaluate whether the association is affected by timing of first diagnosis, gestational age at birth (term vs. preterm), sex, and race-ethnicity. RESULTS Hypothyroidism diagnosed prior to (IRD = 1.30), or during (IRD = 0.59) pregnancy increases the risk of ADHD in the children (aHR = 1.27; 95% CI: 1.15, 1.41, and 1.17; 95% CI: 1.00, 1.38). The association was strongest when diagnosed during the first trimester (IRD = 0.97 and aHR = 1.28; 95% CI: 1.04, 1.58). For children born preterm, there was significantly increased risk of ADHD if their mothers were diagnosed prior to (IRD = 3.06 and aHR = 1.43; 95% CI: 1.09, 1.88), but not during pregnancy. The effect of maternal hypothyroidism on increased risk of ADHD was stronger for boys (IRD = 1.84 and aHR = 1.26; 95% CI: 1.14, 1.40) than it was for girls (IRD = 0.48 and aHR = 1.19; 95% CI: 1.01, 1.40) and for Hispanic children (IRD = 1.60 and aHR = 1.45; 95% CI: 1.25, 1.68) compared with other race ethnicities. CONCLUSION Exposure to maternal hypothyroidism during the periconceptual period significantly increases the risk of ADHD and that the association varies with gestational age at delivery, child sex, and race-ethnicity. KEY POINTS · Maternal hypothyroidism increases the risk of ADHD diagnosis in the offspring.. · The association of maternal hypothyroidism with childhood ADHD was influenced by timing of diagnosis.. · Strength of the association was strongest in preterm born infants, boys, and Hispanic children..
Collapse
Affiliation(s)
- Morgan R Peltier
- Department of Foundations of Medicine, NYU-Long Island School of Medicine, Mineola, New York.,Department of Obstetrics and Gynecology, NYU-Winthrop Hospital, Mineola, New York
| | - Michael J Fassett
- Department of Obstetrics and Gynecology, Kaiser Permanente West Los Angeles Medical Center, Los Angeles, California
| | - Vicki Y Chiu
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | - Darios Getahun
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California.,Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| |
Collapse
|
44
|
Marcelino CP, McAninch EA, Fernandes GW, Bocco BMLC, Ribeiro MO, Bianco AC. Temporal Pole Responds to Subtle Changes in Local Thyroid Hormone Signaling. J Endocr Soc 2020; 4:bvaa136. [PMID: 33123655 PMCID: PMC7575126 DOI: 10.1210/jendso/bvaa136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
To study thyroid hormone (TH) signaling in the human brain, we analyzed published microarray data sets of the temporal pole (Brodmann area 38) of 19 deceased donors. An index of TH signaling built on the expression of 19 well known TH-responsive genes in mouse brains (T3S+) varied from 0.92 to 1.1. After Factor analysis, T3S+ correlated independently with the expression of TH transporters (MCT8, LAT2), TH receptor (TR) beta and TR coregulators (CARM1, MED1, KAT2B, SRC2, SRC3, NCOR2a). Unexpectedly, no correlation was found between T3S+ vs DIO2, DIO3, SRC1, or TRα. An unbiased systematic analysis of the entire transcriptome identified a set of 1649 genes (set #1) with strong positive correlation with T3S+ (r > 0.75). Factor analysis of set #1 identified 2 sets of genes that correlated independently with T3S+, sets #2 (329 genes) and #3 (191 genes). When processed through the Molecular Signatures Data Base (MSigDB), both sets #2 and #3 were enriched with Gene Ontology (GO)-sets related to synaptic transmission and metabolic processes. Ranking individual human brain donors according to their T3S+ led us to identify 1262 genes (set #4) with >1.3-fold higher expression in the top half. The analysis of the overlapped genes between sets #1 and #4 resulted in 769 genes (set #5), which have a very similar MSigDB signature as sets #2 and #3. In conclusion, gene expression in the human temporal pole can be assessed through T3S+ and fluctuates with subtle variations in local TH signaling.
Collapse
Affiliation(s)
- Cícera P Marcelino
- Department of Health and Biological Sciences - CCBS, Mackenzie Presbyterian University, Sao Paulo, Sao Paulo, Brazil
- Department of Translational Medicine, Federal University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois
| | - Gustavo W Fernandes
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| | - Barbara M L C Bocco
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| | - Miriam O Ribeiro
- Department of Health and Biological Sciences - CCBS, Mackenzie Presbyterian University, Sao Paulo, Sao Paulo, Brazil
- Department of Translational Medicine, Federal University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Antonio C Bianco
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| |
Collapse
|
45
|
Hamuro J, Deguchi H, Fujita T, Ueda K, Tokuda Y, Hiramoto N, Numa K, Nakano M, Bush J, Ueno M, Sotozono C, Kinoshita S. Polarized Expression of Ion Channels and Solute Carrier Family Transporters on Heterogeneous Cultured Human Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2020; 61:47. [PMID: 32455435 PMCID: PMC7405722 DOI: 10.1167/iovs.61.5.47] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To clarify the expression profiles of ion channels and transporters of metabolic substrates among heterogeneous cultured human corneal endothelial cells (cHCECs) distinct in their effectiveness in reconstituting the corneal endothelium. Methods Integrated proteomics for cell lysates by liquid chromatography–tandem mass spectrometry was carried out from three aliquots of cHCECs enriched in either cluster of definition (CD)44−/+ (mature) cHCECs or CD44++/+++ cell-state transition (CST) cHCECs. The expression profiles of cations/anions, monocarboxylic acid transporters (MCTs), and solute carrier (SLC) family proteins, as well as carbonic anhydrases (CAs), were investigated. Results The polarized expression of cations/anions, MCTs, and SLC family proteins, as well as CAs, was clarified for mature and CST cHCECs. Most SLC4 family members, including SLC4A11 and SLC4A4 (NBCe1), were upregulated in the CST cHCECs, whereas SLC9A1 (Na+/H+ exchanger isoform one [NHE1]) and CA5B were detected only in the mature cHCECs. In addition, SLC25A42, catalyzing the entry of coenzyme A into the mitochondria, and SLC25A18, functioning as a mitochondrial glutamate carrier 2 (both relevant for providing the substrates for mitochondrial bioenergetics), were selectively expressed in the mature cHCECs. Conclusions Our findings may suggest the relevance of qualifying the polarized expression of these ion channels and transporter-like proteins to ensure not only the suitability but also the in vivo biological functionality of cHCECs selected for use in a cell-injection therapy.
Collapse
|
46
|
Lin C, Li N, Chang H, Shen Y, Li Z, Wei W, Chen H, Lu H, Ji J, Liu N. Dual effects of thyroid hormone on neurons and neurogenesis in traumatic brain injury. Cell Death Dis 2020; 11:671. [PMID: 32826870 PMCID: PMC7442821 DOI: 10.1038/s41419-020-02836-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 11/30/2022]
Abstract
Thyroid hormone (TH) plays a crucial role in neurodevelopment, but its function and specific mechanisms remain unclear after traumatic brain injury (TBI). Here we found that treatment with triiodothyronine (T3) ameliorated the progression of neurological deficits in mice subjected to TBI. The data showed that T3 reduced neural death and promoted the elimination of damaged mitochondria via mitophagy. However, T3 did not prevent TBI-induced cell death in phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (Pink1) knockout mice suggesting the involvement of mitophagy. Moreover, we also found that T3 promoted neurogenesis via crosstalk between mature neurons and neural stem cells (NSCs) after TBI. In neuron cultures undergoing oxygen and glucose deprivation (OGD), conditioned neuron culture medium collected after T3 treatment enhanced the in vitro differentiation of NSCs into mature neurons, a process in which mitophagy was required. Taken together, these data suggested that T3 treatment could provide a therapeutic approach for TBI by preventing neuronal death via mitophagy and promoting neurogenesis via neuron–NSC crosstalk.
Collapse
Affiliation(s)
- Chao Lin
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China.
| | - Nan Li
- Department of Nephrology, Drum Tower Hospital, Nanjing, 210029, China
| | - Hanxiao Chang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Yuqi Shen
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Zheng Li
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Wu Wei
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Hua Chen
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Hua Lu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jing Ji
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China.
| | - Ning Liu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
47
|
Deisenroth C, Soldatow VY, Ford J, Stewart W, Brinkman C, LeCluyse EL, MacMillan DK, Thomas RS. Development of an In Vitro Human Thyroid Microtissue Model for Chemical Screening. Toxicol Sci 2020; 174:63-78. [PMID: 31808822 PMCID: PMC8061085 DOI: 10.1093/toxsci/kfz238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Thyroid hormones (TH) are essential for regulating a number of diverse physiological processes required for normal growth, development, and metabolism. The US EPA Endocrine Disruptor Screening Program (EDSP) has identified several molecular thyroid targets relevant to hormone synthesis dynamics that have been adapted to high-throughput screening (HTS) assays to rapidly evaluate the ToxCast/Tox21 chemical inventories for potential thyroid disrupting chemicals (TDCs). The uncertainty surrounding the specificity of active chemicals identified in these screens and the relevance to phenotypic effects on in vivo human TH synthesis are notable data gaps for hazard identification of TDCs. The objective of this study was to develop a medium-throughput organotypic screening assay comprised of reconstructed human thyroid microtissues to quantitatively evaluate the disruptive effects of chemicals on TH production and secretion. Primary human thyroid cells procured from qualified euthyroid donors were analyzed for retention of NK2 homeobox 1 (NKX2-1), Keratin 7 (KRT7), and Thyroglobulin (TG) protein expression by high-content image analysis to verify enrichment of follicular epithelial cells. A direct comparison of 2-dimensional (2D) and 3-dimensional (3D) 96-well culture formats was employed to characterize the morphology, differential gene expression, TG production, and TH synthesis over the course of 20 days. The results indicate that modeling human thyroid cells in the 3D format was sufficient to restore TH synthesis not observed in the 2D culture format. Inhibition of TH synthesis in an optimized 3D culture format was demonstrated with reference chemicals for key molecular targets within the thyroid gland. Implementation of the assay may prove useful for interpreting phenotypic effects of candidate TDCs identified by HTS efforts currently underway in the EDSP.
Collapse
Affiliation(s)
- Chad Deisenroth
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | | - Jermaine Ford
- Research Cores Unit, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina 27711
| | - Wendy Stewart
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Cassandra Brinkman
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | | - Denise K. MacMillan
- Research Cores Unit, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina 27711
| | - Russell S. Thomas
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
48
|
Zhang J, Liu H, Li J, Lou L, Zhang S, Feng D, Feng X. Exposure to deltamethrin in adolescent mice induced thyroid dysfunction and behavioral disorders. CHEMOSPHERE 2020; 241:125118. [PMID: 31683416 DOI: 10.1016/j.chemosphere.2019.125118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023]
Abstract
Deltamethrin (DM) has become one of the most widely used insecticides in the world due to its low toxicity, high efficiency and low persistence in soil. However, it is still unknown whether DM exposure has any effects on the Hypothalamic-Pituitary-Thyroid (HPT) axis in adolescent mice. In this study, the open field test and circadian activity test showed that DM exposure increased activity. There was no significant difference between the groups in the light/dark box test and nest building test. Forced swimming test showed that after 6 and 12 mg kg-1 DM exposure 28 days, the immobility time was increased and the swimming time was reduced. After 6 mg kg-1 DM treatment, the thyroid stimulating hormone (TSH) content increased, and thyrotropin releasing hormone (TRH), triiodothyronine (T3) and thyroxine (T4) decreased. After exposure to 6 and 12 mg kg-1 DM, mRNA levels of HPT axis-related genes were destroyed. The histological examination showed that, the DM groups mice thyroid tissues appeared expanded thyroid follicles, scanty colloid and hyperplastic thyroid cells. Western blot results showed that the expression level of tyrosine hydroxylase (TH) protein decreased and the content of dopamine transporter (DAT) protein increased in DM treated mice striatum. Collectively, our results indicated that DM exposure could induce thyroid dysfunction and behavioral disorders in adolescent mice.
Collapse
Affiliation(s)
- Jingwen Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China; Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, 300350, China
| | - Haoyue Liu
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Jiangning Li
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Lixiang Lou
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Shaozhi Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Daofu Feng
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Xizeng Feng
- The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China; Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, 300350, China.
| |
Collapse
|
49
|
Talhada D, Feiteiro J, Costa AR, Talhada T, Cairrão E, Wieloch T, Englund E, Santos CR, Gonçalves I, Ruscher K. Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke. Acta Neuropathol Commun 2019; 7:216. [PMID: 31864415 PMCID: PMC6925884 DOI: 10.1186/s40478-019-0866-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023] Open
Abstract
The development of new therapeutic approaches for stroke patients requires a detailed understanding of the mechanisms that enhance recovery of lost neurological functions. The efficacy to enhance homeostatic mechanisms during the first weeks after stroke will influence functional outcome. Thyroid hormones (TH) are essential regulators of neuronal plasticity, however, their role in recovery related mechanisms of neuronal plasticity after stroke remains unknown. This study addresses important findings of 3,5,3′-triiodo-L-thyronine (T3) in the regulation of homeostatic mechanisms that adjust excitability – inhibition ratio in the post-ischemic brain. This is valid during the first 2 weeks after experimental stroke induced by photothrombosis (PT) and in cultured neurons subjected to an in vitro model of acute cerebral ischemia. In the human post-stroke brain, we assessed the expression pattern of TH receptors (TR) protein levels, important for mediating T3 actions. Our results show that T3 modulates several plasticity mechanisms that may operate on different temporal and spatial scales as compensatory mechanisms to assure appropriate synaptic neurotransmission. We have shown in vivo that long-term administration of T3 after PT significantly (1) enhances lost sensorimotor function; (2) increases levels of synaptotagmin 1&2 and levels of the post-synaptic GluR2 subunit in AMPA receptors in the peri-infarct area; (3) increases dendritic spine density in the peri-infarct and contralateral region and (4) decreases tonic GABAergic signaling in the peri-infarct area by a reduced number of parvalbumin+ / c-fos+ neurons and glutamic acid decarboxylase 65/67 levels. In addition, we have shown that T3 modulates in vitro neuron membrane properties with the balance of inward glutamate ligand-gated channels currents and decreases synaptotagmin levels in conditions of deprived oxygen and glucose. Interestingly, we found increased levels of TRβ1 in the infarct core of post-mortem human stroke patients, which mediate T3 actions. Summarizing, our data identify T3 as a potential key therapeutic agent to enhance recovery of lost neurological functions after ischemic stroke.
Collapse
|
50
|
He Q, Li L, Li Y, Lu Y, Wu K, Zhang R, Teng J, Zhao J, Jia Y. Free thyroxine level is associated with both relapse rate and poor neurofunction in first-attack Neuromyelitis Optica Spectrum Disorder (NMOSD) patients. BMC Neurol 2019; 19:329. [PMID: 31852443 PMCID: PMC6921452 DOI: 10.1186/s12883-019-1560-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 12/08/2019] [Indexed: 12/11/2022] Open
Abstract
Background To investigate whether the serum free thyroxine (FT4) level is a prognostic factor for the first-attack neuromyelitis optica spectrum disorders (NMOSD). Methods This retrospective study enrolled 109 patients with first-attack NMOSD. The Expanded Disability Status Scale (EDSS) and the relapse rate were used to evaluate the outcomes. The logistic regression model was used to analyze the independent effects of FT4 on relapse and final EDSS. Kaplan-Meier analysis, scatter plot smoothing method, and two-phase piecewise linear regression model were used to investigate the relationship between the FT4 level and the relapse rate. Results Multivariate analysis revealed that serum FT4 level might be a risk factor for both final EDSS (β = 0.17; 95% confidence interval: 0.03–0.32) and the relapse rate (HR = 1.18; 95% confidence interval: 1.05–1.32). Furthermore, 1400 days after the onset, nearly 100% of patients in the high-FT4 group relapsed, while only 40% of the patients in the low-FT4 group relapsed. Finally, we found that the relationship between the FT4 level and the NMOSD relapse rate was nonlinear. The risk of NMOSD relapse increased with the FT4 level up to the inflection point of 12.01 pmol/L (HR = 1.45; 95% confidence interval: 1.06–1.98). When the FT4 level was > 12.01 pmol/L, there was no correlation between the FT4 level and the risk of NMOSD relapse (HR = 1.05; 95% confidence interval: 0.78–1.41). Conclusion Serum FT4 level may be a prognostic indicator for the first-attack in patients with NMOSD. High FT4 levels are associated with poor neurofunctions and a high relapse rate in patients with the first-attack in patients with NMOSD.
Collapse
Affiliation(s)
- Qianyi He
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lifeng Li
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanfei Li
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yanhui Lu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Kaimin Wu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruiyi Zhang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Junfang Teng
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jie Zhao
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,National Telemedicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, Henan, China
| | - Yanjie Jia
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|