1
|
Huang Y, Su T. Dysregulation of LINC01094 is involved in the pathogenesis of pulpitis by regulating the miR-340-5p expression. Odontology 2025:10.1007/s10266-024-01046-5. [PMID: 39786709 DOI: 10.1007/s10266-024-01046-5] [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: 09/30/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Pulpitis seriously affects people's living standards and dental health, so identifying effective therapeutic targets is crucial for pulpitis. The research aimed to explore the underlying regulatory mechanism of LINC01094 and miR-340-5p in pulpitis. The study involved a total of 173 subjects (97 pulpitis and 76 healthy individuals). The expression of LINC01094 and miR-340-5p were evaluated through the polymerase chain reaction (PCR). The association linking LINC01094 and miR-340-5p expression was assessed by Pearson correlation analysis. The Human dental pulp cells (HDPCs) injury model was conducted by lipopolysaccharide (LPS). Cell proliferation was examined through the Cell Counting Kit-8 assay and flow cytometry. Cell apoptosis was also evaluated by flow cytometry. The caspase-3 levels and inflammatory cytokines were quantified using an enzyme-linked immunosorbent assay (ELISA). Upregulated LINC01094 and downregulated miR-340-5p expression were observed in pulpitis and LPS-induced HDPC injury models. A negative correlation was observed between miR-340-5p and LINC01094 expression in pulpitis. LPS could suppress proliferation and promote apoptosis of HDPCs. The TNF-α, IL-6, and IL-1β levels in LPS-induced HDPCs were also elevated. The HDPC injury induced by LPS could be aggravated by the LINC01094 overexpression. MiR-340-5p showed a relieved effect on HDPC injury and could alleviate the HDPC injury aggravated by LINC01094 overexpression. In summary, upregulated LINC01094 and downregulated miR-340-5p expression was observed in pulpitis. LINC01094 could accelerate the pulpitis progression via targeting miR-340-5p.
Collapse
Affiliation(s)
- Yuao Huang
- Jinzhou Medical University Graduate Training Base (Central Hospital of Fengxian District, Shanghai), Shanghai, 201499, China
- Department of Stomatology, Shanghai Fengxian District Traditional Chinese Medicine Hospital, Shanghai, 201499, China
| | - Tao Su
- Department of Stomatology, Shanghai Fengxian District Central Hospital, No.6600 Nanfeng Highway, Shanghai, 201400, China.
| |
Collapse
|
2
|
Bastos CR, Bevilacqua LM, Mendes LFB, Xavier J, Gruhn K, Kaster MP, Ghisleni G. Amygdala-specific changes in Cacna1c, Nfat5, and Bdnf expression are associated with stress responsivity in mice: A possible mechanism for psychiatric disorders. J Psychiatr Res 2024; 175:259-270. [PMID: 38754148 DOI: 10.1016/j.jpsychires.2024.05.019] [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: 09/23/2023] [Revised: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The CACNA1C gene encodes the alpha-1c subunit of the Cav1.2 calcium channel, a regulator of neuronal calcium influx involved in neurotransmitter release and synaptic plasticity. Genetic data show a role for CACNA1C in depressive symptoms underlying different psychiatric diagnoses. However, the mechanisms involved still require further exploration. This study aimed to investigate sex and region-specific changes in the Cacna1c gene and behavioral outcomes in mice exposed to chronic stress. Moreover, we evaluated the Nuclear factor of activated T-cells 5 (Nfat5) and the Brain-derived neurotrophic factor (Bdnf) as potential upstream and downstream Cacna1c targets and their correlation in stressed mice and humans with depression. Male and female Swiss mice were exposed to chronic unpredictable stress (CUS) for 21 days. Animal-integrated emotionality was assessed using the sucrose splash test, the tail suspension, the open-field test, and the elevated-plus-maze. Gene expression analysis was performed in the amygdala, prefrontal cortex, and hippocampus. Human data for in silico analysis was obtained from the Gene Expression Omnibus. CUS-induced impairment in integrated emotional regulation was observed in males. Gene expression analysis showed decreased levels of Cacna1c and Nfat5 and increased levels of Bdnf transcripts in the amygdala of stressed male mice. In contrast, there were no major changes in behavioral responses or gene expression in female mice after stress. The expression of the three genes was significantly correlated in the amygdala of mice and humans. The strong and positive correlation between Canac1c and Nfat5 suggests a potential role for this transcription factor in Canac1c expression. These changes could impact amygdala reactivity and emotional responses, making them a potential target for psychiatric intervention.
Collapse
Affiliation(s)
- Clarissa Ribeiro Bastos
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil; Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Laura Menegatti Bevilacqua
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Luiz Filipe Bastos Mendes
- Center of Oxidative Stress Research, Department of Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Janaina Xavier
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Karen Gruhn
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Manuella Pinto Kaster
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil.
| | - Gabriele Ghisleni
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
3
|
Xu F, Bian N, Li X. SNHG14 Elevates NFAT5 Expression Through Sequestering miR-375-3p to Promote MPP + -Induced Neuronal Apoptosis, Inflammation, and Oxidative Stress in Parkinson's Disease. Neurochem Res 2024; 49:1212-1225. [PMID: 38381247 DOI: 10.1007/s11064-024-04106-y] [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: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons. LncRNA small nucleolar RNA host gene 14 (SNHG14) was found to promote neuron injury in PD. Here, we investigated the mechanisms of SNHG14 in PD process. In vivo or in vitro PD model was established by using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice or 1-methyl-4-phenylpyridinium (MPP +)-stimulated SK-N-SH cells. The expression of genes and proteins was measured by qRT-PCR and Western blot. In vitro assays were conducted using ELISA, CCK-8, colony formation, EdU, flow cytometry, and Western blot assays, respectively. The oxidative stress was evaluated by determining the production of superoxide dismutase (SOD) and malondialdehyde (MDA). The direct interactions between miR-375-3p and NFAT5 (Nuclear factor of activated T-cells 5) or SNHG14 was verified using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. SNHG14 and NFAT5 were elevated, while miR-375-3p was decreased in MPTP-mediated PD mouse model and MPP + -induced SK-N-SH cells. Knockdown of SNHG14 or NFAT5, or overexpression of miR-375-3p reversed MPP + -induced neuronal apoptosis, inflammation, and oxidative stress. Mechanistically, SNHG14 directly bound to miR-375, which targeted NFAT5. Inhibition of miR-375-3p abolished the inhibitory activity of SNHG14 knockdown on MPP + -evoked neuronal damage. Besides that, NFAT5 up-regulation counteracted the effects of miR-375-3p on MPP + -mediated neuronal damage. SNHG14 contributed to MPP + -induced neuronal injury by miR-375/NFAT5 axis, suggesting a new insight into the pathogenesis of PD.
Collapse
Affiliation(s)
- Furong Xu
- Department of Neurology, Chengdu Seventh People's Hospital, Chengdu, 610000, Sichuan, China
| | - Na Bian
- Department of Neurology, Baoji City People's Hospital, Baoji, 721000, Shaanxi, China
| | - Xuewen Li
- Department of Neurosurgery, People's Hospital of Dingxi City, 22 Anding Road, Anding District, Dingxi, 743000, Gansu, China.
| |
Collapse
|
4
|
He L, Ma S, Ding Z, Huang Z, Zhang Y, Xi C, Zou K, Deng Q, Huang WJM, Guo Q, Huang C. Inhibition of NFAT5-Dependent Astrocyte Swelling Alleviates Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302916. [PMID: 38195869 PMCID: PMC10953562 DOI: 10.1002/advs.202302916] [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: 05/08/2023] [Revised: 12/03/2023] [Indexed: 01/11/2024]
Abstract
Astrocyte swelling is implicated in various neurological disorders. However, whether astrocyte swelling contributes to neuropathic pain remains elusive. This study elucidates the pivotal role of the nuclear factor of activated T-cells 5 (NFAT5) emerges as a master regulator of astrocyte swelling in the spinal dorsal horn (SDH) during neuropathic pain. Despite the ubiquitous expression of NFAT5 protein in SDH cell types, it selectively induces swelling specifically in astrocytes, not in microglia. Mechanistically, NFAT5 directly controls the expression of the water channel aquaporin-4 (AQP4), a key regulator exclusive to astrocytes. Additionally, aurora kinase B (AURKB) orchestrates NFAT5 phosphorylation, enhancing its protein stability and nuclear translocation, thereby regulating AQP4 expression. The findings establish NFAT5 as a crucial regulator for neuropathic pain through the modulation of astrocyte swelling. The AURKB-NFAT5-AQP4 pathway in astrocytes emerges as a potential therapeutic target to combat neuropathic pain.
Collapse
Affiliation(s)
- Liqiong He
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Shengyun Ma
- Department of Cellular and Molecular MedicineUniversity of California San DiegoSan DiegoCA92093USA
| | - Zijin Ding
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Zhifeng Huang
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Yu Zhang
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Caiyun Xi
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Kailu Zou
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Qingwei Deng
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Wendy Jia Men Huang
- Department of Cellular and Molecular MedicineUniversity of California San DiegoSan DiegoCA92093USA
| | - Qulian Guo
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Changsheng Huang
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
5
|
Massri AJ, Fitzpatrick M, Cunny H, Li JL, Harry GJ. Differential gene expression profiling implicates altered network development in rat postnatal day 4 cortex following 4-Methylimidazole (4-MeI) induced maternal seizures. Neurotoxicol Teratol 2023; 100:107301. [PMID: 37783441 PMCID: PMC10843020 DOI: 10.1016/j.ntt.2023.107301] [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: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
Compromised maternal health leading to maternal seizures can have adverse effects on the healthy development of offspring. This may be the result of inflammation, hypoxia-ischemia, and altered GABA signaling. The current study examined cortical tissue from F2b (2nd litter of the 2nd generation) postnatal day 4 (PND4) offspring of female Harlan SD rats chronically exposed to the seizuregenic compound, 4-Methylimidazole (0, 750, or 2500 ppm 4-MeI). Maternal seizures were evident only at 2500 ppm 4-MeI. GABA related gene expression as examined by qRT-PCR and whole genome microarray showed no indication of disrupted GABA or glutamatergic signaling. Canonical pathway hierarchical clustering and multi-omics combinatory genomic (CNet) plots of differentially expressed genes (DEG) showed alterations in genes associated with regulatory processes of cell development including neuronal differentiation and synaptogenesis. Functional enrichment analysis showed a similarity of cellular processes across the two exposure groups however, the genes comprising each cluster were primarily unique rather than shared and often showed different directionality. A dose-related induction of cytokine signaling was indicated however, pathways associated with individual cytokine signaling were not elevated, suggesting an alternative involvement of cytokine signaling. Pathways related to growth process and cell signaling showed a negative activation supporting an interpretation of disruption or delay in developmental processes at the 2500 ppm 4-MeI exposure level with maternal seizures. Thus, while GABA signaling was not altered as has been observed with maternal seizures, the pattern of DEG suggested a potential for alteration in neuronal network formation.
Collapse
Affiliation(s)
- Abdull J Massri
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Mackenzie Fitzpatrick
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Helen Cunny
- Office of the Scientific Director, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - G Jean Harry
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
6
|
Ambati R, Kho LK, Prentice D, Thompson A. Osmotic demyelination syndrome: novel risk factors and proposed pathophysiology. Intern Med J 2023; 53:1154-1162. [PMID: 35717664 DOI: 10.1111/imj.15855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/30/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Osmotic demyelination syndrome (ODS) is non-inflammatory demyelination in response to an osmotic challenge. It can be pontine or extrapontine in presentation. AIMS To retrospectively review cases involving ODS and define the spectrum of causes, risk factors, clinical and radiological presentations, and functional outcomes. RESULTS The study utilised data from 15 patients with a mean age of 53.6 years. Malnutrition (9; 60%) and chronic alcoholism (10; 66.7%) were the most common associated disorders. Two (13.3%) patients had severe hyponatraemia (<120 mmol/L). The average highest single-day change was 5.1 mmol/L. Radiologically, 14 (93.3%) had pontine and 6 (40%) had extra-pontine lesions. Hypokalaemia (14; 93.3%) and hypophosphataemia (9; 60%) were commonly associated. Common clinical manifestations include altered consciousness/encephalopathy (9; 60%), dysphagia (4; 26.7%) and limb weakness (4; 26.7%). At 3 months, two (14.3%) had died and six (40%) were functionally independent (modified Rankin scale 0-2). CONCLUSION We found that ODS occurred despite appropriate correction rates of hyponatraemia. Factors such as malnutrition, chronic alcoholism, hypokalaemia and hypophosphataemia are thought to play a role in its pathogenesis. Approximately half of the patients survived and became functionally independent.
Collapse
Affiliation(s)
- Ravi Ambati
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - Lay K Kho
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - David Prentice
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - Andrew Thompson
- Neurological Intervention and Imaging Service of WA (NIISwa), Royal Perth and Sir Charles Gairdner Hospitals, Perth, Western Australia, Australia
| |
Collapse
|
7
|
Jansen MI, Castorina A. Identification of Key Genes and Regulatory Pathways in Multiple Sclerosis Brain Samples: A Meta-Analysis of Micro-Array Datasets. Int J Mol Sci 2023; 24:ijms24119361. [PMID: 37298310 DOI: 10.3390/ijms24119361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS) whose aetiology is only partly understood. Investigating the intricate transcriptional changes occurring in MS brains is critical to unravel novel pathogenic mechanisms and therapeutic targets. Unfortunately, this process is often hindered by the difficulty in retrieving an adequate number of samples. However, by merging data from publicly available datasets, it is possible to identify alterations in gene expression profiles and regulatory pathways that were previously overlooked. Here, we merged microarray gene expression profiles obtained from CNS white matter samples taken from MS donors to identify novel differentially expressed genes (DEGs) linked with MS. Data from three independent datasets (GSE38010, GSE32915, and GSE108000) were combined and used to detect novel DEGs using the Stouffer's Z-score method. Corresponding regulatory pathways were analysed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. Finally, top up- and down-regulated transcripts were validated by real-time quantitative PCR (qPCR) using an independent set of white matter tissue samples obtained from MS donors with different disease subtypes. There were a total of 1446 DEGs, of which 742 were up-regulated and 704 genes were down-regulated. DEGs were associated with several myelin-related pathways and protein metabolism pathways. Validation studies of selected top up- or down-regulated genes highlighted MS subtype-specific differences in the expression of some of the identified genes, underlining a more complex scenario of white matter pathology amongst people afflicted by this devastating disease.
Collapse
Affiliation(s)
- Margo I Jansen
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
| |
Collapse
|
8
|
Zeng M, Zhang T, Lin Y, Lin Y, Wu Z. The Common LncRNAs of Neuroinflammation-Related Diseases. Mol Pharmacol 2023; 103:113-131. [PMID: 36456192 DOI: 10.1124/molpharm.122.000530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 12/04/2022] Open
Abstract
Spatio-temporal specific long noncoding RNAs (lncRNAs) play important regulatory roles not only in the growth and development of the brain but also in the occurrence and development of neurologic diseases. Generally, the occurrence of neurologic diseases is accompanied by neuroinflammation. Elucidation of the regulatory mechanisms of lncRNAs on neuroinflammation is helpful for the clinical treatment of neurologic diseases. This paper focuses on recent findings on the regulatory effect of lncRNAs on neuroinflammatory diseases and selects 10 lncRNAs that have been intensively studied to analyze their mechanism action. The clinical treatment status of lncRNAs as drug targets is also reviewed. SIGNIFICANCE STATEMENT: Gene therapies such as clustered regularly interspaced short palindrome repeats technology, antisense RNA technology, and RNAi technology are gradually applied in clinical treatment, and the development of technology is based on a large number of basic research investigations. This paper focuses on the mechanisms of lncRNAs regulation of neuroinflammation, elucidates the beneficial or harmful effects of lncRNAs in neurosystemic diseases, and provides theoretical bases for lncRNAs as drug targets.
Collapse
Affiliation(s)
- Meixing Zeng
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Ting Zhang
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Yan Lin
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Yongluan Lin
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Zhuomin Wu
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| |
Collapse
|
9
|
Packialakshmi B, Hira S, Lund K, Zhang AH, Halterman J, Feng Y, Scott DW, Lees JR, Zhou X. NFAT5 contributes to the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and decrease of T regulatory cells in female mice. Cell Immunol 2022; 375:104515. [DOI: 10.1016/j.cellimm.2022.104515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/03/2022]
|
10
|
Yuan M, Zhang J, He Y, Yi G, Rong L, Zheng L, Zhan T, Zhou C. Circ_0062558 promotes growth, migration, and glutamine metabolism in triple-negative breast cancer by targeting the miR-876-3p/SLC1A5 axis. Arch Gynecol Obstet 2022; 306:1643-1655. [PMID: 35284960 DOI: 10.1007/s00404-022-06481-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to function as vital regulators in cancers, including triple-negative breast cancer (TNBC). This study aimed to explore the role of circ_0062558 in TNBC. METHODS The real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to quantify the expressions of circ_0062558, microRNA-876-3p (miR-876-3p), and solute carrier family 1 (neutral amino acid transporter), member 5 (SLC1A5) in TNBC tissues and cells. 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT), thymidine analog 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, wound healing, and Transwell assays were employed for cell phenotype analyses. Protein expression was tested by western blot analysis. Dual-luciferase reporter was used to confirm the association among circ_0062558, miR-876-3p, and SLC1A5 in TNBC. Xenograft experiments were performed to elucidate the function of circ_0062558 in vivo. RESULTS TNBC tissues and cells showed the higher level of circ_0062558 when compared with control samples. Downregulation of circ_0062558 inhibited proliferation, migration, invasion, and glutamine metabolism, while enhanced apoptosis of TNBC cells, and silencing of circ_0062558 also inhibited the growth of tumor in vivo. MiR-876-3p was confirmed as a target of circ_0062558, and circ_0062558 knockdown repressed TNBC cell malignant behaviors by increasing miR-876-3p. Furthermore, miR-876-3p inhibited malignant behaviors of TNBC cells by down-regulating SLC1A5, a newly identified target of miR-876-3p. CONCLUSION Circ_0062558 promoted TNBC progression by enhancing proliferation, survival, migration, invasion, and glutamine metabolism via miR-876-3p/SLC1A5 axis, which was helpful for understanding the carcinogenic roles of circ_0062558.
Collapse
Affiliation(s)
- Mengzhen Yuan
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Jun Zhang
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Yuxin He
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Guangming Yi
- The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, China
| | - Liwen Rong
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Liangjian Zheng
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Tingting Zhan
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China
| | - Congming Zhou
- Department of Oncology, The Third People's Hospital of Chengdu, No.82, Qinglong Street, Qingyang District, Chengdu, 610031, Sichuan, China.
| |
Collapse
|
11
|
Su L, Li R, Zhang Z, Liu J, Du J, Wei H. Identification of altered exosomal microRNAs and mRNAs in Alzheimer's disease. Ageing Res Rev 2022; 73:101497. [PMID: 34710587 DOI: 10.1016/j.arr.2021.101497] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by decreased memory and cognitive functions. Exosomes carry a variety of important information such as proteins, lipids, DNA and RNA of mother cells. It is reported that exosomes play critical roles in nervous system physiology and neurodegenerative diseases. However, the functions of exosomes in AD progression are not fully elucidated. In this study, we detected the expression pattern of mRNAs and miRNAs in exosomes derived from the AD and health mice. A total of 1320 mRNAs and 29 miRNAs were differentially expressed in exosomes between the two groups. Subsequently, the downregulation of Chi3l1 and upregulation of Rhog in AD mice were verified by qRT-PCR. Meanwhile, the downregulation of miR-148a-5p and upregulation of miR-27a-5p in AD group were also tested by qRT-PCR. The functions of differentially expressed mRNAs and potential target genes of miRNAs were determined by GO and KEGG analysis. According to the ceRNA hypothesis, we established an integrated ceRNA network of circRNA-lncRNA-miRNA-mRNA. In conclusion, exosomal lncRNAs, mRNAs, circRNAs and miRNAs were identified to participate in the progression of AD which might be possible biomarkers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Lining Su
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| | - Renqing Li
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| | - Zhiqing Zhang
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| | - Jijia Liu
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| | - Jingkao Du
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| | - Huiping Wei
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China.
| |
Collapse
|
12
|
Jesus JBD, Sena CBCD, Macchi BDM, do Nascimento JLM. Cyclosporin A as an Alternative Neuroimmune Strategy to Control Neurites and Recover Neuronal Tissues in Leprosy. Neuroimmunomodulation 2022; 29:15-20. [PMID: 34350891 DOI: 10.1159/000517993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/21/2020] [Indexed: 11/19/2022] Open
Abstract
Leprosy, also known as Hansen's disease, continues to have a substantial impact on infectious diseases throughout the world. Leprosy is a chronic granulomatous infection caused by Mycobacterium leprae and shows a wide clinical and immunopathological spectrum related to the immune response of the host. This disease affects the skin and other internal organs with a predilection to infect Schwann cells, which play an active role during axonal degeneration, affecting peripheral nerves and promoting neurological damage. This chronic inflammation influences immune function, leading to neuroimmune disorders. Leprosy is also associated with neuroimmune reactions, including type 1 (reverse) and type 2 (erythema nodosum leprosum) reactions, which are immune-mediated inflammatory complications that can occur during the disease and appear to worsen dramatically; these complications are the main concerns of patients. The reactions may induce neuritis and neuropathic pain that progressively worsen with irreversible deformity and disabilities responsible for the immunopathological damage and glial/neuronal death. However, the neuronal damage is not always associated with the reactional episode. Also, the efficacy in the treatment of reactions remains low because of the nonexistence of a specific treatment and missing informations about the immunopathogenesis of the reactional episode. There is increasing evidence that peripheral neuron dysfunction strongly depends on the activity of neurotrophins. The most important neurotrophin in leprosy is nerve growth factor (NGF), which is decreased in the course of leprosy, as well as the presence of autoantibodies against NGF in all clinical forms of leprosy and neuroimmune reactions. The levels of autoantibodies against NGF are decreased by the immunomodulatory activity of cyclosporin A, which mainly controls pain and improves motor function and sensitivity. Therefore, the suppression of anti-NGF and the regulation of NGF levels can be attractive targets for immunomodulatory treatment and for controlling the neuroimmune reactions of leprosy, although further studies are needed to clarify this point.
Collapse
Affiliation(s)
- Jessica Batista de Jesus
- Laboratory of Molecular and Cellular Neurochemistry, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Chubert Bernardo Castro de Sena
- Laboratory of Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- National Institute of Science and Technology in Neuroimmunomodulation (INCT - NIM), Rio de Janeiro, Brazil
| | - Barbarella de Matos Macchi
- Laboratory of Molecular and Cellular Neurochemistry, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- National Institute of Science and Technology in Neuroimmunomodulation (INCT - NIM), Rio de Janeiro, Brazil
| | - José Luiz Martins do Nascimento
- Laboratory of Molecular and Cellular Neurochemistry, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- National Institute of Science and Technology in Neuroimmunomodulation (INCT - NIM), Rio de Janeiro, Brazil
- Graduation Program in Pharmaceutical Science, Federal University of Amapá, Macapá, Brazil
| |
Collapse
|
13
|
Exposure to hypertonic solutions during pregnancy induces autism-like behaviors via the NFAT-5 pathway in offspring in a rat model. Physiol Behav 2021; 240:113545. [PMID: 34363817 DOI: 10.1016/j.physbeh.2021.113545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVES to investigate the effects of hyperosmolar state (HS) on immune response and inflammation via the NFAT5 pathway and examine whether immune-mediated conditions trigger autism-like behavior in offspring. METHODS a pregnant rat model was performed by administering hyperosmotic solutions. Pregnant rats were divided into 2 main groups; control (group I) and hyperosmolar groups (group II). Control group rats were given % 0.25 NaCI (tap water) (n = 6), the Hyperosmolar (HO) group was further subdivided into 3 groups as; Group II a rats which were given % 3 hypertonic NaCl (n = 6), Group II b rats were given mineral water (% 3 NaHCO3+magnesium+calcium content) (n = 6), and Group II c rats were given Ayran (% 0.8 NaCl content) (n = 6). Their offspring were examined for behaviors, biochemical and histological abnormality. RESULTS in offspring, TNF- α, IL-17, NFAT-5, and NGF levels in the brain were significantly higher in hyperosmotic solution groups than in control rats. Exposure of pregnant rats to hyperosmotic solution resulted in autism-like behaviors in their offspring. Through immunohistochemical methods, we found that CA1 and CA2 of the hippocampus indicated decreased number of neurons in hyperosmotic solution groups compared with the control group. CONCLUSIONS our findings once again emphasized that the immune-mediated conditions involved in the pathophysiology of autism. NFAT5 pathway may be a key factor in the development of neuroinflammation by hyperosmotic solutions.
Collapse
|
14
|
Zhang M, Yang H, Chen Z, Hu X, Wu T, Liu W. Long Noncoding RNA X-Inactive-Specific Transcript Promotes the Secretion of Inflammatory Cytokines in LPS Stimulated Astrocyte Cell Via Sponging miR-29c-3p and Regulating Nuclear Factor of Activated T cell 5 Expression. Front Endocrinol (Lausanne) 2021; 12:573143. [PMID: 33776905 PMCID: PMC7995889 DOI: 10.3389/fendo.2021.573143] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Astrocyte activation promotes glutamate accumulation and secretion of inflammatory factors, mainly responsible for epilepsy. Long noncoding RNA (lncRNA) X-inactive-specific transcript (XIST) regulates inflammation; however, the biological role and regulatory mechanism of XIST during astrocyte activation remain unclear. METHODS In the present study, rat epilepsy model and lipopolysaccharide (LPS)-treated CTX-TNA2 were established. XIST and miR-29c-3p expression were evaluated using quantitative real-time polymerase chain reaction. Nuclear factor of activated T cells 5 (NFAT5) was measured using western blot analysis. Interleukin (IL)-1β, IL-6, tumor necrosis factor-α, and L-glutamate levels in the culture supernatants were assessed using enzyme-linked immunosorbent assay. The binding between XIST and miR-29c-3p and between miR-29c-3p and the 3'-UTR of NFAT5 was analyzed using dual-luciferase reporter, RNA-binding protein immunoprecipitation (RIP), and Biotin pull-down assay. The proliferation and apoptosis were evaluated using CCK8 and flow cytometry, respectively. RESULTS XIST expression and NFAT5 protein level was increased, whereas miR-29c-3p expression was decreased in the epilepsy rat model and LPS-treated CTX-TNA2 cells. Silenced XIST expression, miR-29c-3p overexpression, or silenced NFAT5 expression inhibited the secretion of IL-1β, IL-6, and TNF-α and promoted glutamate transport in LPS-treated CTX-TNA2 cells. miR-29c-3p was the potential miRNA sponged by XIST. NFAT5 acted as a direct binding target of miR-29c-3p. Silenced miR-29c-3p expression or NFAT5 overexpression reversed the effect of silenced XIST expression on LPS-treated CTX-TNA2.XIST and miR-29c-3p treatment does not affect NFAT5 mRNA expression, but affects NFAT5 protein level. Furthermore, underexpressed XIST or overexpressed miR-29c-3p in LPS-stimulated CTX-TNA2 can attenuate neuronal apoptosis induced by LPS-stimulated CTX-TNA2. CONCLUSION LncRNA XIST promotes the secretion of inflammatory cytokines in LPS- treated CTX-TNA2 via sponging miR-29c-3p and regulating NFAT5 expression.
Collapse
|
15
|
O'Sullivan MP, Casey S, Finder M, Ahearne C, Clarke G, Hallberg B, Boylan GB, Murray DM. Up-Regulation of Nfat5 mRNA and Fzd4 mRNA as a Marker of Poor Outcome in Neonatal Hypoxic-Ischemic Encephalopathy. J Pediatr 2021; 228:74-81.e2. [PMID: 32828883 DOI: 10.1016/j.jpeds.2020.08.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate umbilical cord messenger RNA (mRNA) expression as biomarkers for the grade of hypoxic-ischemic encephalopathy (HIE) and long-term neurodevelopment outcome. STUDY DESIGN Infants were recruited from the BiHiVE1 study, Ireland (2009-2011), and the BiHiVE2 study, Ireland, and Sweden (2013-2015). Infants with HIE were assigned modified Sarnat scores at 24 hours and followed at 18-36 months. mRNA expression from cord blood was measured using quantitative real-time polymerase chain reaction. RESULTS We studied 124 infants (controls, n = 37; perinatal asphyxia, n = 43; and HIE, n = 44). Fzd4 mRNA increased in severe HIE (median relative quantification, 2.98; IQR, 2.23-3.68) vs mild HIE (0.88; IQR, 0.46-1.37; P = .004), and in severe HIE vs moderate HIE (1.06; IQR, 0.81-1.20; P = .003). Fzd4 mRNA also increased in infants eligible for therapeutic hypothermia (1.20; IQR, 0.92-2.37) vs those who were ineligible for therapeutic hypothermia group (0.81; IQR, 0.46-1.53; P = .017). Neurodevelopmental outcome was analyzed for 56 infants. Nfat5 mRNA increased in infants with severely abnormal (1.26; IQR, 1.17-1.39) vs normal outcomes (0.97; IQR, 0.83-1.24; P = .036), and also in infants with severely abnormal vs mildly abnormal outcomes (0.96; IQR, 0.80-1.06; P = .013). Fzd4 mRNA increased in infants with severely abnormal (2.51; IQR, 1.60-3.56) vs normal outcomes (0.74; IQR, 0.48-1.49; P = .004) and in infants with severely abnormal vs mildly abnormal outcomes (0.97; IQR, 0.75-1.34; P = .026). CONCLUSIONS Increased Fzd4 mRNA expression was observed in cord blood of infants with severe HIE; Nfat5 mRNA and Fzd4 mRNA expression were increased in infants with severely abnormal long-term outcomes. These mRNA may augment current measures as early objective markers of HIE severity at delivery.
Collapse
Affiliation(s)
- Marc Paul O'Sullivan
- INFANT Research Centre, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork, Ireland; National Children's Research Centre, Crumlin, Dublin, Ireland.
| | - Sophie Casey
- INFANT Research Centre, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Mikael Finder
- Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Neonatal Department, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Ahearne
- INFANT Research Centre, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Gerard Clarke
- INFANT Research Centre, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome, Ireland, University College Cork, Cork, Ireland
| | - Boubou Hallberg
- Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Neonatal Department, Karolinska University Hospital, Stockholm, Sweden
| | - Geraldine B Boylan
- INFANT Research Centre, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Deirdre M Murray
- INFANT Research Centre, Ireland; Department of Paediatrics and Child Health, University College Cork, Cork, Ireland; National Children's Research Centre, Crumlin, Dublin, Ireland
| |
Collapse
|
16
|
Duan S, Wang F, Cao J, Wang C. Exosomes Derived from MicroRNA-146a-5p-Enriched Bone Marrow Mesenchymal Stem Cells Alleviate Intracerebral Hemorrhage by Inhibiting Neuronal Apoptosis and Microglial M1 Polarization. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3143-3158. [PMID: 32821084 PMCID: PMC7425091 DOI: 10.2147/dddt.s255828] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/31/2022]
Abstract
Introduction Intracerebral hemorrhage (ICH) is a devastating type of stroke with high mortality, and the effective therapies for ICH remain to be explored. Exosomes (Exos) have been found to play important roles in cell communication by transferring molecules, including microRNAs (miRNAs/miRs). MiRNAs are critical regulators of genes involved in many various biological processes and have been demonstrated to aggravate or alleviate brain damages induced by ICH. The aim of the present study was to investigate the effect of Exos derived from miR-146a-5p-enriched bone marrow mesenchymal stem cells (BMSCs-miR-146a-5p-Exos) on experimental ICH. Methods ICH was induced in adult male Sprague-Dawley rats by an intrastriatal injection of collagenase type IV. At 24 h after surgery, Exos were administrated. For detecting apoptotic cells, TUNEL staining was performed using an in situ Cell Death Detection Kit. Fluoro-Jade B staining was performed to detect degenerating neurons. Immunofluorescence assay was performed to detect the expression of myeloperoxidase (MPO) and OX-42. The binding of miR-146a-5p and its target genes was confirmed by luciferase reporter assay. Results At 24 h after surgery, BMSCs-miR-146a-5p-Exos administration significantly improved neurological function, reduced apoptotic and degenerative neurons, and inhibited inflammatory response. Furthermore, miR-146a-5p-enriched Exos obviously inhibited the M1 polarization of microglia after ICH in rats, accompanied by the reduced expression of pro-inflammatory mediators releasing by M1 microglia including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and monocyte chemoattractant protein-1 (MCP-1). Finally, we observed that miR-146a-5p directly targeted interleukin-1 receptor-associated kinase1 (IRAK1) and nuclear factor of activated T cells 5 (NFAT5), which contributed to the inflammation response and the polarization of M1 microglia/macrophages. Conclusion We demonstrated that miR-146a-5p-riched BMSCs-Exos could offer neuroprotection and functional improvements after ICH through reducing neuronal apoptosis, and inflammation associated with the inhibition of microglial M1 polarization by downregulating the expression of IRAK1 and NFAT5.
Collapse
Affiliation(s)
- Shurong Duan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Fei Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Jingwei Cao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Chunyan Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| |
Collapse
|
17
|
DDC expression is not regulated by NFAT5 (TonEBP) in dopaminergic neural cell lines. Gene 2020; 742:144569. [PMID: 32165301 DOI: 10.1016/j.gene.2020.144569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/21/2020] [Accepted: 03/08/2020] [Indexed: 11/30/2022]
Abstract
The nuclear factor of activated T-cells 5 (NFAT5), also known as tonicity-responsive enhancer-binding protein (TonEBP), is a transcription factor that regulates osmoadaptive response in multiple tissues and is highly expressed in the developing central nervous system. A former study reported that NFAT5 activation through hypertonic stress increases the expression of the dopa decarboxylase enzyme (DDC), also known as aromatic-l-amino-acid decarboxylase (AADC), in human renal proximal tubule cells, leading to an increase of dopamine synthesis. In a previous study, we identified NFAT5 as a candidate gene for cocaine dependence, a complex psychiatric disorder in which dopaminergic neurotransmission plays an important role. Therefore, to test the hypothesis that NFAT5 may also affect dopamine levels in the nervous system through the regulation of DDC expression, we examined this regulation using two neural dopaminergic cell lines, SH-SY5Y and PC12. The effect of NFAT5 on the expression of the neuronal isoform of DDC was evaluated by qRT-PCR. Upon hypertonic stress, NFAT5 was activated and accumulated into the nuclei and, subsequently, the expression of NFAT5 and its known targets sodium/myo-inositol cotransporter 1 (SMIT) and sodium chloride/taurine cotransporter (TAUT) increased, as expected. However, the expression of DDC decreased. When silencing the expression of NFAT5 with a specific shRNA we observed that the downregulation of DDC is independent from NFAT5 in both cell lines and is due to hypertonic stress. In conclusion, NFAT5 does not regulate the expression of the neuronal isoform of DDC in neural dopaminergic cell lines and, consequently, it does not modulate dopamine synthesis through DDC.
Collapse
|
18
|
Wang R, Li Q, He Y, Yang Y, Ma Q, Li C. miR-29c-3p inhibits microglial NLRP3 inflammasome activation by targeting NFAT5 in Parkinson's disease. Genes Cells 2020; 25:364-374. [PMID: 32160394 DOI: 10.1111/gtc.12764] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
Microglial inflammation is identified as a key process associated with Parkinson's disease (PD) pathogenesis. Our previous study showed that miR-29c-3p (miR-29c) exhibited anti-inflammatory properties in PD animal and neuronal models. However, the specific role and regulatory mechanism of miR-29c played in microglia are still unclear. In this study, lipopolysaccharide (LPS)-stimulated BV-2 cells were used to establish a cellular model of microglial activation for investigating PD. The results showed a decreased expression of miR-29c in LPS-induced BV-2 cells. Over-expression of miR-29c suppressed LPS-triggered Iba-1 increment, pro-inflammatory cytokine release, and NF-кB and TXNIP/NLRP3 inflammasome activation. Silence of miR-29c induced similar effects with LPS on microglial inflammation. In addition, we found that NFAT5 was negatively correlated with miR-29c. Knockdown of NFAT5 blocked the aggravated inflammation in microglia treated by miR-29c inhibitor. Thus, these findings suggest that miR-29c modulates NLRP3 inflammasome to impair microglial inflammatory responses by targeting NFAT5, which represents a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Ruili Wang
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qing Li
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ya He
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Yang
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiaoya Ma
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Li
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
19
|
Johansen ML, Stetson LC, Vadmal V, Waite K, Berens ME, Connor JR, Lathia J, Rubin JB, Barnholtz-Sloan JS. Gliomas display distinct sex-based differential methylation patterns based on molecular subtype. Neurooncol Adv 2020; 2:vdaa002. [PMID: 32642674 PMCID: PMC7212920 DOI: 10.1093/noajnl/vdaa002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Gliomas are the most common type of primary brain tumor and one of many cancers where males are diagnosed with greater frequency than females. However, little is known about the sex-based molecular differences in glioblastomas (GBMs) or lower grade glioma (non-GBM) subtypes. DNA methylation is an epigenetic mechanism involved in regulating gene transcription. In glioma and other cancers, hypermethylation of specific gene promoters downregulates transcription and may have a profound effect on patient outcome. The purpose of this study was to determine if sex-based methylation differences exist in different glioma subtypes. Methods Molecular and clinical data from glioma patients were obtained from The Cancer Genome Atlas and grouped according to tumor grade and molecular subtype (IDH1/2 mutation and 1p/19q chromosomal deletion). Sex-specific differentially methylated probes (DMPs) were identified in each subtype and further analyzed to determine if they were part of differentially methylated regions (DMRs) or associated with differentially methylated DNA transcription regulatory binding motifs. Results Analysis of methylation data in 4 glioma subtypes revealed unique sets of both sex-specific DMPs and DMRs in each subtype. Motif analysis based on DMP position also identified distinct sex-based sets of DNA-binding motifs that varied according to glioma subtype. Downstream targets of 2 of the GBM-specific transcription binding sites, NFAT5 and KLF6, showed differential gene expression consistent with increased methylation mediating downregulation. Conclusion DNA methylation differences between males and females in 4 glioma molecular subtypes suggest an important, sex-specific role for DNA methylation in epigenetic regulation of gliomagenesis.
Collapse
Affiliation(s)
- Mette L Johansen
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - L C Stetson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Vachan Vadmal
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Kristin Waite
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Cleveland Center for Health Outcomes Research, Cleveland, Ohio, USA
| | - Michael E Berens
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Justin Lathia
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joshua B Rubin
- Departments of Pediatrics and Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Cleveland Center for Health Outcomes Research, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Cen L, Xing F, Xu L, Cao Y. Potential Role of Gene Regulator NFAT5 in the Pathogenesis of Diabetes Mellitus. J Diabetes Res 2020; 2020:6927429. [PMID: 33015193 PMCID: PMC7512074 DOI: 10.1155/2020/6927429] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Nuclear factor of activated T cells 5 (NFAT5), a Rel/nuclear factor- (NF-) κB family member, is the only known gene regulator of the mammalian adaptive response to osmotic stress. Exposure to elevated glucose increases the expression and nuclear translocation of NFAT5, as well as NFAT5-driven transcriptional activity in vivo and in vitro. Increased expression of NFAT5 is closely correlated with the progression of diabetes in patients. The distinct structure of NFAT5 governs its physiological and pathogenic roles, indicating its opposing functions. The ability of NFAT5 to maintain cell homeostasis and proliferation is impaired in patients with diabetes. NFAT5 promotes the formation of aldose reductase, pathogenesis of diabetic vascular complications, and insulin resistance. Additionally, NFAT5 activates inflammation at a very early stage of diabetes and induces persistent inflammation. Recent studies revealed that NFAT5 is an effective therapeutic target for diabetes. Here, we describe the current knowledge about NFAT5 and its relationship with diabetes, focusing on its diverse regulatory functions, and highlight the importance of this protein as a potential therapeutic target in patients with diabetes.
Collapse
Affiliation(s)
- Lusha Cen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengling Xing
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Liying Xu
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Youdian Rd. 54th, Hangzhou 310006, China
| |
Collapse
|
21
|
Yang XL, Zeng ML, Shao L, Jiang GT, Cheng JJ, Chen TX, Han S, Yin J, Liu WH, He XH, Peng BW. NFAT5 and HIF-1α Coordinate to Regulate NKCC1 Expression in Hippocampal Neurons After Hypoxia-Ischemia. Front Cell Dev Biol 2019; 7:339. [PMID: 31921851 PMCID: PMC6923656 DOI: 10.3389/fcell.2019.00339] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/29/2019] [Indexed: 12/22/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious birth complication with severe long-term sequelae such as cerebral palsy, epilepsy and cognitive disabilities. Na+-K+-2Cl– cotransporters 1 (NKCC1) is dramatically upregulated after hypoxia-ischemia (HI), which aggravates brain edema and brain damage. Clinically, an NKCC1-specific inhibitor, bumetanide, is used to treat diseases related to aberrant NKCC1 expression, but the underlying mechanism of aberrant NKCC1 expression has rarely been studied in HIE. In this study, the cooperative effect of hypoxia-inducible factor-1α (HIF-1α) and nuclear factor of activated T cells 5 (NFAT5) on NKCC1 expression was explored in hippocampal neurons under hypoxic conditions. HI increased HIF-1α nuclear localization and transcriptional activity, and pharmacological inhibition of the HIF-1α transcription activity or mutation of hypoxia responsive element (HRE) motifs recovered the hypoxia-induced aberrant expression and promoter activity of NKCC1. In contrast, oxygen–glucose deprivation (OGD)-induced downregulation of NFAT5 expression was reversed by treating with hypertonic saline, which ameliorated aberrant NKCC1 expression. More importantly, knocking down NFAT5 or mutation of the tonicity enhancer element (TonE) stimulated NKCC1 expression and promoter activity under normal physiological conditions. The positive regulation of NKCC1 by HIF-1α and the negative regulation of NKCC1 by NFAT5 may serve to maintain NKCC1 expression levels, which may shed light on the transcription regulation of NKCC1 in hippocampal neurons after hypoxia.
Collapse
Affiliation(s)
- Xing-Liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Lin Shao
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Guang-Tong Jiang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jing-Jing Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Hua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Andres-Hernando A, Johnson RJ, Lanaspa MA. Endogenous fructose production: what do we know and how relevant is it? Curr Opin Clin Nutr Metab Care 2019; 22:289-294. [PMID: 31166222 PMCID: PMC6684314 DOI: 10.1097/mco.0000000000000573] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Excessive sugar and particularly fructose consumption has been proposed to be a key player in the pathogenesis of metabolic syndrome and kidney disease in humans and animal models. However, besides its dietary source, fructose can be endogenously produced in the body from glucose via the activation of the polyol pathway. In this review, we aim to describe the most recent findings and current knowledge on the potential role of endogenous fructose production and metabolism in disease. RECENT FINDINGS Over the recent years, the activation of the polyol pathway and endogenous fructose production has been observed in multiple tissues including the liver, renal cortex, and hypothalamic areas of the brain. The activation occurs during the development and progression of metabolic syndrome and kidney disease and results from different stimuli including osmotic effects, diabetes, and ischemia. Even though the potential toxicity of the activation of the polyol pathway can be attributed to several intermediate products, the blockade of endogenous fructose metabolism either by using fructokinase deficient mice or specific inhibitors resulted in marked amelioration of multiple metabolic diseases. SUMMARY New findings suggest that fructose can be produced in the body and that the blockade of tis metabolism could be clinically relevant for the prevention and treatment of metabolic syndrome and kidney disease.
Collapse
|