1
|
Lu L, Shi Y, Wei B, Li W, Yu X, Zhao Y, Yu D, Sun M. YTHDF3 modulates the Cbln1 level by recruiting BTG2 and is implicated in the impaired cognition of prenatal hypoxia offspring. iScience 2024; 27:108703. [PMID: 38205248 PMCID: PMC10776956 DOI: 10.1016/j.isci.2023.108703] [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: 04/13/2023] [Revised: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
The "Fetal Origins of Adult Disease (FOAD)" hypothesis holds that adverse factors during pregnancy can increase the risk of chronic diseases in offspring. Here, we investigated the effects of prenatal hypoxia (PH) on brain structure and function in adult offspring and explored the role of the N6-methyladenosine (m6A) pathway. The results suggest that abnormal cognition in PH offspring may be related to the dysregulation of the m6A pathway, specifically increased levels of YTHDF3 in the hippocampus. YTHDF3 interacts with BTG2 and is involved in the decay of Cbln1 mRNA, leading to the down-regulation of Cbln1 expression. Deficiency of Cbln1 may contribute to abnormal synaptic function, which in turn causes cognitive impairment in PH offspring. This study provides a scientific clues for understanding the mechanisms of impaired cognition in PH offspring and provides a theoretical basis for the treatment of cognitive impairment in offspring exposed to PH.
Collapse
Affiliation(s)
- Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Weisheng Li
- Department of Gynaecology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Dongyi Yu
- 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
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Islam M, Behura SK. Role of caveolin-1 in metabolic programming of fetal brain. iScience 2023; 26:107710. [PMID: 37720105 PMCID: PMC10500482 DOI: 10.1016/j.isci.2023.107710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/10/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023] Open
Abstract
Mice lacking caveolin-1 (Cav1), a key protein of plasma membrane, exhibit brain aging at an early adult stage. Here, integrative analyses of metabolomics, transcriptomics, epigenetics, and single-cell data were performed to test the hypothesis that metabolic deregulation of fetal brain due to the ablation of Cav1 is linked to brain aging in these mice. The results of this study show that lack of Cav1 caused deregulation in the lipid and amino acid metabolism in the fetal brain, and genes associated with these deregulated metabolites were significantly altered in the brain upon aging. Moreover, ablation of Cav1 deregulated several metabolic genes in specific cell types of the fetal brain and impacted DNA methylation of those genes in coordination with mouse epigenetic clock. The findings of this study suggest that the aging program of brain is confounded by metabolic abnormalities in the fetal stage due to the absence of Cav1.
Collapse
Affiliation(s)
- Maliha Islam
- Division of Animal Sciences, 920 East Campus Drive, University of Missouri, Columbia, MO 65211, USA
| | - Susanta K. Behura
- Division of Animal Sciences, 920 East Campus Drive, University of Missouri, Columbia, MO 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
- Interdisciplinary Reproduction and Health Group, University of Missouri, Columbia, MO, USA
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA
| |
Collapse
|
3
|
Li W, Zhang Y, Lv J, Zhang Y, Bai J, Zhen L, He X. MicroRNA-137-mediated lysine demethylase 4A regulates the recovery of spinal cord injury via the SFRP4-Wnt/β-Catenin axis. Int J Neurosci 2023; 133:37-50. [PMID: 33499717 DOI: 10.1080/00207454.2021.1881093] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Spinal cord injury (SCI) causes great harm to the normal life of patients. Histone demethylase is involved in many biological processes, including SCI. Hence, this study explored the role and mechanism of histone lysine demethylase 4A (KDM4A) in SCI. METHODS The acute SCI (ASCI) rat model was established after spinal compression and the SCI neuronal model was induced via treating PC12 cells with lipopolysaccharide (LPS). KDM4A expression during SCI was detected. The microRNA (miRNA) targeting KDM4A was predicted and verified. The miRNA and KDM4A expression patterns were intervened in LPS-stimulated PC12 cells to evaluate their combined effects on neuronal cells in SCI. The downstream pathways of KDM4A were predicted, and SFRP4 and H3K9me3 expressions were determined. After the intervention of SFRP4 in LPS-treated cells, β-Catenin expression and the effect of SFRP4 on neuronal cells in SCI were detected. Finally, the effectiveness of the miR-137/KDM4A/SFRP4/Wnt/β-Catenin axis was verified in vivo. RESULTS KDM4A was abnormally elevated in SCI. miR-137 targeted KDM4A. miR-137 effectively inhibited the apoptosis of LPS-challenged PC12 cells, which could be reversed after overexpressing KDM4A. KDM4A promoted SFRP4 expression through demethylation of H3K9me3. Overexpression of SFRP4 blocked the Wnt/β-Catenin pathway and promoted apoptosis of LPS-stimulated cells. In vivo, miR-137 overexpression remarkably improved SCI symptoms, accompanied by obviously increased β-Catenin expression and notably decreased KDM4A and SFRP4 expressions, while overexpressed KDM4A treatment showed the opposite trend in the presence of miR-137. CONCLUSION We demonstrated that miR-137 targeted KDM4A and then downregulated SFRP4 to ameliorate SCI in a Wnt/β-Catenin-dependent manner.
Collapse
Affiliation(s)
- Wei Li
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Ying Zhang
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Jianrui Lv
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Yong Zhang
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Jie Bai
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Luming Zhen
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Xijing He
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
4
|
Hif-1α regulates Tet1-c-Myc binding involved in depression-like behavior in prenatal hypoxia offspring. Neuroscience 2022; 502:41-51. [PMID: 36041588 DOI: 10.1016/j.neuroscience.2022.08.014] [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: 06/21/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022]
Abstract
Prenatal hypoxia (PH) is one of the most common adverse stimulation during pregnancy. The brain is fragile in the fetal period and sensitive to hypoxia. The offspring who have experienced PH may be at increased risk of developing neurodevelopmental disorders after birth and various neuropsychiatric diseases after adulthood. In this study, pregnant mice used to generate PH offspring were treated with hypoxia (10.5% oxygen) from gestational day 12.5 to 17.5. Compared with control mice, the birth weight of offspring in the PH group was significantly lower and the male adult offspring exhibited significant depression-like behavior. The expression of the oxygen-sensitive subunit of hypoxia-inducible factor (Hif-1α) was significantly elevated, whereas Ten-eleven translocated methylcytosine dioxygenase 1 (Tet1) and c-Myc, which is closely related to cell proliferation, was significantly decreased in the hippocampus of the male offspring in the PH group. In addition, the PH group showed increased binding of Hif-1α to Tet1, and decreased binding of Tet1 to c-Myc, resulting in increased ubiquitinated degradation of c-Myc and decreased neurogenesis in the hippocampus of the male offspring. These findings suggest that Hif-1α regulates Tet1-c-Myc binding involved in depression-like behavior in PH offspring and Hif-1α can be used as a detection index of stress-related diseases.
Collapse
|
5
|
Yao C, Lu L, Ji Y, Zhang Y, Li W, Shi Y, Liu J, Sun M, Xia F. Hypo-Hydroxymethylation of Nobox is Associated with Ovarian Dysfunction in Rat Offspring Exposed to Prenatal Hypoxia. Reprod Sci 2022; 29:1424-1436. [PMID: 35257353 PMCID: PMC9005429 DOI: 10.1007/s43032-022-00866-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/23/2022] [Indexed: 11/23/2022]
Abstract
Prenatal hypoxia (PH) is a common feature of a suboptimal intrauterine environment affecting the development of fetuses. Whether PH leads to abnormal ovary development is not yet clear. This study investigated ovarian function in offspring exposed to PH and the potential underlying molecular mechanisms. SD female rats (n = 12 per group) at 9 weeks of age were housed in individual cages (21% O2). After the pregnant rats were exposed to hypoxia (10.5% oxygen) from embryonic day (E) 5 to E21, PH offspring were generated. All animals maintained normoxia during lactation. The number of follicles was counted in female offspring at 3 months under an optical microscope. The expression of Nobox, Gdf9, and Tets was detected by quantitative real-time polymerase chain reaction (PCR) and Western blot. Global DNA hydroxymethylation was measured by dot blot. The hydroxymethylation level of the Nobox gene was evaluated with an NGS-based multiple targeted CpG hydroxymethylation analysis method. Body weight and ovary weight were significantly decreased in the PH group compared with the control group. PH offspring have abnormal estrous cycle, decreased serum anti-Mullerian hormone (AMH), and increased serum follicle-stimulating hormone (FSH), and follicular atresia, which are consistent with the clinical manifestations in patients with ovarian dysfunction. In terms of mechanism, the expression of Nobox was significantly decreased in the PH group. Subsequent high-throughput sequencing results showed that the level of hydroxymethylation in the candidate region of the Nobox gene was reduced. Cultured cells treated with hypoxia exhibited lower levels of both 5hmC and Nobox, while vitamin C, a coactivator of Tets, rescued hypo-hydroxymethylation and increased the expression level of Nobox. This study indicated that PH could cause hypo-hydroxymethylation of Nobox through epigenetic regulation and may consequently contribute to ovarian dysfunction in adult rat offspring.
Collapse
Affiliation(s)
- Changfang Yao
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
- Obstetrics of the Third Affiliated Hospital of Soochow University, Changzhou, 213000 Jiangsu China
| | - Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Yiting Ji
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
| | - Yingying Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Weisheng Li
- 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
| | - Jinliu Liu
- 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
| | - Fei Xia
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
| |
Collapse
|
6
|
Wang B, Xia L, Zhu D, Zeng H, Wei B, Lu L, Li W, Shi Y, Liu J, Zhang Y, Sun M. Paternal High-Fat Diet Altered Sperm 5'tsRNA-Gly-GCC Is Associated With Enhanced Gluconeogenesis in the Offspring. Front Mol Biosci 2022; 9:857875. [PMID: 35480893 PMCID: PMC9035875 DOI: 10.3389/fmolb.2022.857875] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
Background: Paternal lifestyle, stress and environmental exposures play a crucial role in the health of offspring and are associated with non-genetic inheritance of acquired traits, however the underlying mechanisms are unclear. In this study, we aimed to find out how the sperm tsRNA involved in paternal high-fat diet induced abnormal gluconeogenesis of F1 offspring, and explore the underlying molecular mechanism of its regulation. Method: We generated a paternal high fat diet (42% kcal fat) model to investigate the mechanism by which paternal diet affects offspring metabolism. Four-week-old C57BL/6J male mice were randomly assigned into two groups to receive either a control diet (CD; 10% kcal fat) or a high-fat (HFD; 42% kcal fat) diet for 10 weeks, and mice from each group were then mated with 8-week-old females with control diet in a 1:2 ratio to generate F1. F0 sperms were isolated and small RNAs was sequenced by high-throughput sequencing. Metabolic phenotypes were examined with both F0 and F1. Results: A significant increase in body weight was observed with HFD-F0 mice at 8 weeks of age as compared to CD mice at the same age. F0 mice showed impaired glucose tolerance (GTT), resistance to insulin tolerance (ITT) and increased pyruvate tolerance (PTT) at 14 weeks. HFD-F1 male mice showed no significant difference in body weight. An increase in PTT was found at 13 weeks of age and no significant changes in GTT and ITT. PEPCK and G6Pase that related to gluconeogenesis increased significantly in the liver of HFD-F1 male mice. Sperm sequencing results showed that 5′tsRNA-Gly-GCC derived from tRNA-Gly-GCC-2 specifically was remarkably upregulated in sperm of HFD F0 mice. Q-PCR further showed that this tsRNA was also increased in the liver of HFD-F1 comparison with CD-F1 mice. In addition, we found that 5′tsRNA-Gly-GCC can regulate Sirt6-FoxO1 pathway and be involved in the gluconeogenesis pathway in liver. Conclusion: 5′tsRNA-Gly-GCC that increased in HFD mice mature sperms can promote gluconeogenesis in liver by regulating Sirt6-FoxO1 pathway, which might represent a potential paternal epigenetic factor mediating the intergenerational inheritance of diet-induced metabolic alteration.
Collapse
Affiliation(s)
- Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Lin Xia
- Medical Center of Hematology, The Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Dan Zhu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Weisheng Li
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Jingliu Liu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
| | - Yunfang Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- *Correspondence: Yunfang Zhang, ; Miao Sun,
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, China
- *Correspondence: Yunfang Zhang, ; Miao Sun,
| |
Collapse
|
7
|
Guo R, Wang X, Fang Y, Chen X, Chen K, Huang W, Chen J, Hu J, Liang F, Du J, Dordoe C, Tian X, Lin L. rhFGF20 promotes angiogenesis and vascular repair following traumatic brain injury by regulating Wnt/β-catenin pathway. Biomed Pharmacother 2021; 143:112200. [PMID: 34649342 DOI: 10.1016/j.biopha.2021.112200] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
The pathology of cerebrovascular disorders takes an important role in traumatic brain injury (TBI) by increasing intracranial pressure. Fibroblast growth factor 20 (FGF20) is a brain-derived neurotrophic factor, that has been shown to play an important role in the survival of dopaminergic neurons and the treatment of Parkinson's disease (PD). However, little is known about the role of FGF20 in the treatment of TBI and its underlying mechanism. The purpose of this study was to evaluate the protective effect of recombinant human FGF20 (rhFGF20) on protecting cerebral blood vessels after TBI. In this study, we indicated that rhFGF20 could reduce brain edema, Evans blue penetration and upregulated the expression of blood-brain barrier (BBB)-related tight junction (TJ) proteins, exerting a protective effect on the BBB in vivo after TBI. In the TBI repair phase, rhFGF20 promoted angiogenesis, neurological and cognitive function recovery. In tumor necrosis factor-α (TNF-α)-induced human brain microvascular endothelial cells (hCMEC/D3), an in vitro BBB disruption model, rhFGF20 reversed the impairment in cell migration and tube formation induced by TNF-α. Moreover, in both the TBI mouse model and the in vitro model, rhFGF20 increased the expression of β-catenin and GSK3β, which are the two key regulators in the Wnt/β-catenin signaling pathway. In addition, the Wnt/β-catenin inhibitor IWR-1-endo significantly reversed the effects of rhFGF20. These results indicate that rhFGF20 may prevent vascular repair and angiogenesis through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiongjian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Kun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xianxi Tian
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China.
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Beijing 100730, China.
| |
Collapse
|
8
|
Zhang M, Liu J, Zhang R, Liang Z, Ding S, Yu H, Shan Y. Nobiletin, a hexamethoxyflavonoid from citrus pomace, attenuates G1 cell cycle arrest and apoptosis in hypoxia-induced human trophoblast cells of JEG-3 and BeWo via regulating the p53 signaling pathway. Food Nutr Res 2021; 65:5649. [PMID: 34650395 PMCID: PMC8494266 DOI: 10.29219/fnr.v65.5649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/11/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022] Open
Abstract
Background Hypoxia is associated with abnormal cell apoptosis in trophoblast cells, which causes fetal growth restriction and related placental pathologies. Few effective methods for the prevention and treatment of placenta-related diseases exist. Natural products and functional foods have always been a rich source of potential anti-apoptotic drugs. Nobiletin (NOB), a hexamethoxyflavonoid derived from the citrus pomace, shows an anti-apoptotic activity, which is a non-toxic constituent of dietary phytochemicals approved by the Food and Drug Administration. However, their effects on hypoxia-induced human trophoblast cells have not been fully studied. Objective The aim of this study was to investigate the protective effects of NOB on hypoxia-induced apoptosis of human trophoblast JEG-3 and BeWo cells, and their underlying mechanisms. Design First, the protective effect of NOB on hypoxia-induced apoptosis of JEG-3 and BeWo cells was studied. Cell viability and membrane integrity were determined by CCK-8 assay and lactate dehydrogenase activity, respectively. Real Time Quantitative PCR (RT-qPCR) and Western blot analysis were used to detect the mRNA and protein levels of HIF1α. Propidium iodide (PI)-labeled flow cytometry was used to detect cell cycle distribution. Cell apoptosis was detected by flow cytometry with Annexin V-FITC and PI double staining, and the expression of apoptosis marker protein cl-PARP was detected by Western blot analysis. Then, the molecular mechanism of NOB against apoptosis was investigated. Computer molecular docking and dynamics were used to simulate the interaction between NOB and p53 protein, and this interaction was verified in vitro by Ultraviolet and visible spectrum (UV-visible spectroscopy), fluorescence spectroscopy and circular dichroism. Furthermore, the changes in the expression of p53 signaling pathway genes and proteins were detected by RT-qPCR and Western blot analysis, respectively. Results Hypoxia treatment resulted in a decreased cell viability and cell membrane integrity in JEG-3 and BeWo cell lines, and an increased expression of HIF1α, cell cycle arrest in the G1 phase, and massive cell apoptosis, which were alleviated after NOB treatment. Molecular docking and dynamics simulations found that NOB spontaneously bonded to human p53 protein, leading to the change of protein conformation. The intermolecular interaction between NOB and human p53 protein was further confirmed by UV-visible spectroscopy, fluorescence spectroscopy and circular dichroism. After the treatment of 100 μM NOB, a down-regulation of mRNA and protein levels of p53 and p21 and an up-regulation of BCL2/BAX mRNA and protein ratio were observed in JEG-3 cells; however, there was also a down-regulation of mRNA and protein levels observed for p53 and p21 in BeWo cells after the treatment of NOB. The BCL2/BAX ratio of BeWo cells did not change after the treatment of 100 μM NOB. Conclusion NOB attenuated hypoxia-induced apoptosis in JEG-3 and BeWo cell lines and might be a potential functional ingredient to prevent pregnancy-related diseases caused by hypoxia-induced apoptosis. These findings would also suggest the exploration and utilization of citrus resources, and the development of citrus industry.
Collapse
Affiliation(s)
- Mengling Zhang
- Longping Branch Graduate School, Hunan University, Changsha, Hunan Province, China.,School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China.,Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan Province, China.,Hunan Key Lab of Fruits & Vegetables Storage, Processing, Quality and Safety, Hunan Agricultural Products Processing Institute, Changsha, Hunan Province, China
| | - Jian Liu
- Longping Branch Graduate School, Hunan University, Changsha, Hunan Province, China.,Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan Province, China.,Hunan Key Lab of Fruits & Vegetables Storage, Processing, Quality and Safety, Hunan Agricultural Products Processing Institute, Changsha, Hunan Province, China
| | - Rui Zhang
- School of Medical Humanity, Peking University, Beijing, China
| | - Zengenni Liang
- Longping Branch Graduate School, Hunan University, Changsha, Hunan Province, China.,Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan Province, China.,Hunan Key Lab of Fruits & Vegetables Storage, Processing, Quality and Safety, Hunan Agricultural Products Processing Institute, Changsha, Hunan Province, China
| | - Shenghua Ding
- Longping Branch Graduate School, Hunan University, Changsha, Hunan Province, China.,Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan Province, China.,Hunan Key Lab of Fruits & Vegetables Storage, Processing, Quality and Safety, Hunan Agricultural Products Processing Institute, Changsha, Hunan Province, China
| | - Huanling Yu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yang Shan
- Longping Branch Graduate School, Hunan University, Changsha, Hunan Province, China.,Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan Province, China.,Hunan Key Lab of Fruits & Vegetables Storage, Processing, Quality and Safety, Hunan Agricultural Products Processing Institute, Changsha, Hunan Province, China
| |
Collapse
|
9
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
10
|
Manukjan N, Ahmed Z, Fulton D, Blankesteijn WM, Foulquier S. A Systematic Review of WNT Signaling in Endothelial Cell Oligodendrocyte Interactions: Potential Relevance to Cerebral Small Vessel Disease. Cells 2020; 9:cells9061545. [PMID: 32630426 PMCID: PMC7349551 DOI: 10.3390/cells9061545] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/β-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/β-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, MHeNs—School for Mental Health and Neuroscience, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881409
| |
Collapse
|
11
|
Chronic disturbance in the thalamus following cranial irradiation to the developing mouse brain. Sci Rep 2019; 9:9588. [PMID: 31270437 PMCID: PMC6610082 DOI: 10.1038/s41598-019-45973-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
Better survival rates among pediatric brain tumor patients have resulted in an increased awareness of late side effects that commonly appear following cancer treatment. Radiation-induced changes in hippocampus and white matter are well described, but do not explain the full range of neurological late effects in childhood cancer survivors. The aim of this study was to investigate thalamus following cranial irradiation (CIR) to the developing brain. At postnatal day 14, male mice pups received a single dose of 8 Gy CIR. Cellular effects in thalamus were assessed using immunohistochemistry 4 months after CIR. Interestingly, the density of neurons decreased with 35% (p = 0.0431) and the density of astrocytes increased with 44% (p = 0.011). To investigate thalamic astrocytes, S100β+ cells were isolated by fluorescence-activated cell sorting and genetically profiled using next-generation sequencing. The phenotypical characterization indicated a disrupted function, such as downregulated microtubules’ function, higher metabolic activity, immature phenotype and degraded ECM. The current study provides novel insight into that thalamus, just like hippocampus and white matter, is severely affected by CIR. This knowledge is of importance to understand the late effects seen in pediatric brain tumor survivors and can be used to give them the best suitable care.
Collapse
|
12
|
Zhang Y, Zhang M, Li L, Wei B, He A, Lu L, Li X, Zhang L, Xu Z, Sun M. Methylation-reprogrammed Wnt/β-catenin signalling mediated prenatal hypoxia-induced brain injury in foetal and offspring rats. J Cell Mol Med 2018; 22:3866-3874. [PMID: 29808608 PMCID: PMC6050486 DOI: 10.1111/jcmm.13660] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/27/2018] [Indexed: 12/13/2022] Open
Abstract
Prenatal hypoxia (PH) is a common pregnancy complication, harmful to brain development. This study investigated whether and how PH affected Wnt pathway in the brain. Pregnant rats were exposed to hypoxia (10.5% O2) or normoxia (21% O2; Control). Foetal brain weight and body weight were decreased in the PH group, the ratio of brain weight to body weight was increased significantly. Prenatal hypoxia increased mRNA expression of Wnt3a, Wnt7a, Wnt7b and Fzd4, but not Lrp6. Activated β‐catenin protein and Fosl1 expression were also significantly up‐regulated. Increased Hif1a expression was found in the PH group associated with the higher Wnt signalling. Among 5 members of the Sfrp family, Sfrp4 was down‐regulated. In the methylation‐regulating genes, higher mRNA expressions of Dnmt1 and Dnmt3b were found in the PH group. Sodium bisulphite and sequencing revealed hyper‐methylation in the promoter region of Sfrp4 gene in the foetal brain, accounting for its decreased expression and contributing to the activation of the Wnt‐Catenin signalling. The study of PC12 cells treated with 5‐aza further approved that decreased methylation could result in the higher Sfrp4 expression. In the offspring hippocampus, protein levels of Hif1a and mRNA expression of Sfrp4 were unchanged, whereas Wnt signal pathway was inhibited. The data demonstrated that PH activated the Wnt pathway in the foetal brain, related to the hyper‐methylation of Sfrp4 as well as Hif1a signalling. Activated Wnt signalling might play acute protective roles to the foetal brain in response to hypoxia, also would result in disadvantageous influence on the offspring in long‐term.
Collapse
Affiliation(s)
- Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Mengshu Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Bin Wei
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Axin He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Likui Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lubo Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China.,Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China.,Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| |
Collapse
|