1
|
Fu DX, Lei YT, Guo HB, Chen T, Gao XY, Wang XL, Huang X, Song LL, Wang SY, Dai QX. PRDX1 affects acrylamide-induced neural damage through the PTEN/AKT signaling pathway. Neurotoxicology 2025; 108:150-158. [PMID: 40189060 DOI: 10.1016/j.neuro.2025.04.003] [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: 01/16/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Peroxiredoxin 1 (PRDX1) is a member of the peroxidase family of antioxidant enzymes. However, the role and mechanism of PRDX1 in acrylamide (ACR)-induced nerve damage have not been reported. We used SD rats and well-differentiated rat pheochromocytoma cells (PC-12 cells) to established in vivo and in vitro models of ACR. Immunohistochemistry, immunofluorescence and RT-qPCR experiments were used to detect the expression of PRDX1 in neurons of rat hippocampal tissue. The ultrastructural changes of neurons and PC-12 cells in rat hippocampal tissue were observed under transmission electron microscope. Western blot detected the protein expression levels of PRDX1, PTEN, AKT and p-AKT. In vivo and in vitro experimental results showed that PRDX1 showed a significant up-regulation trend after ACR exposure (p < 0.05). In vitro experiments showed that after inhibiting PRDX1 expression with PRDX1 siRNA, the survival rate of PC-12 cells significantly increased, and the damage to cell morphology and organelles was markedly improved. Western blot analysis revealed that ACR exposure can cause a significant increase in PTEN protein expression level and p-AKT/AKT protein ratio (p < 0.05). After inhibiting the expression of PRDX1, the protein expression level of PTEN and the protein ratio of p-AKT/AKT were significantly reduced, while the protein levels of SYN1 and BDNF were significantly increased (p < 0.05). This study, for the first time, demonstrates that PRDX1 affects ACR-induced neurotoxicity by regulating the PTEN/AKT signaling pathway. And, provides novel insights into the prevention and treatment of neurotoxicity in populations exposed to ACR.
Collapse
Affiliation(s)
- Dong-Xue Fu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Ya-Ting Lei
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China; Department of Preventive Medicine, Department of medicine, Qiannan medical college for nationalities, Guizhou Province, PR China
| | - Hai-Bo Guo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Ting Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Xiang-Ying Gao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Xiao-Li Wang
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China
| | | | - Ling-Ling Song
- Department of Neurology, The Second Hospital of Chaoyang (Women's and Children's Hospital of Chaoyang), PR China.
| | - Sheng-Yuan Wang
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China.
| | - Qin-Xue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China.
| |
Collapse
|
2
|
Zhong W, Xu L, Jiang G, Gao Y, Song J, Ma Y, Wang G, Fan J, Li W, Zhou S, Yang Y, Yu X. Sodium tanshinone IIA sulfonate promotes proliferation and differentiation of endogenous neural stem cells to repair rat spinal cord injury via the Notch pathway. J Transl Med 2025; 23:367. [PMID: 40128847 PMCID: PMC11934593 DOI: 10.1186/s12967-025-06331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Interventions that promote the proliferation of endogenous neural stem cells (ENSCs) and induce their differentiation into neurons after spinal cord injury (SCI) hold significant potential for SCI repair. Tanshinone IIA (TIIA) exhibits extensive neuroprotective effects, and its derivative, sodium tanshinone IIA sulfonate (STS), has enhanced water solubility, making it easier to prepare injectable formulations and increasing bioavailability. STS injections have been extensively utilized in the treatment of cardiovascular and cerebrovascular diseases, and their clinical application in SCI shows promising potential. However, it remains unclear whether STS can promote spinal cord injury repair in rats by modulating the proliferation and differentiation of ENSCs, and the underlying regulatory mechanisms are yet to be elucidated. METHODS In this study, an incomplete spinal cord injury model was established in rats using the NYU spinal cord impactor. The regulatory effects of STS on ENSCs in rats post-SCI were observed by detecting the NSC marker Nestin, the neuronal marker NeuN, and the astrocyte marker GFAP. Additionally, rat behavioral assessments, histopathology, serum inflammation indices, and Notch signaling pathway activation were evaluated. In vitro experiments utilized an lipopolysaccharide (LPS)-induced rats spinal cord NSCs inflammation model. The effects of STS on the proliferation and viability of rats spinal cord NSCs were assessed using the CCK-8 assay and immunofluorescence cell counting. The mechanisms by which STS regulates NSC proliferation and differentiation via the Notch pathway were verified using immunofluorescence, Western blot, and RT-PCR techniques. RESULTS In vitro, STS significantly reduced the levels of inflammatory indices in the LPS-induced rats NSCs inflammation model and improved the viability of rats NSCs following inflammatory injury. STS also significantly increased the proliferation of NSCs and their differentiation into neurons while reducing their differentiation into astrocytes. Moreover, LPS significantly activated the Notch pathway, similar to the effects of the Notch pathway agonist valproic acid (VPA), whereas STS intervention could inhibit the LPS- or VPA-induced activation of the Notch pathway. In vivo, STS markedly improved the hindlimb motor function of rats with SCI, decreased the levels of pro-inflammatory factors IL-6 and TNF-α, and increased the level of the anti-inflammatory factor IL-10, thereby improving the pathological morphology of the injured spinal cord in rats post-SCI. STS effectively promoted the proliferation of ENSCs post-SCI, facilitated their differentiation into neurons, and inhibited their differentiation into astrocytes. Additionally, STS suppressed the excessive activation of the Notch signaling pathway following SCI. CONCLUSION STS promotes the proliferation of ENSCs post-SCI in rats, induces their differentiation into neurons, and inhibits their differentiation into astrocytes, thereby improving the pathological morphology of the injured spinal cord and promoting the recovery of hindlimb motor function in rats post-SCI. Furthermore, the regulatory effects of STS on the proliferation and differentiation of ENSCs post-SCI in rats may be related to its inhibition of the excessive activation of the Notch signaling pathway.
Collapse
Affiliation(s)
- Wenqing Zhong
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Luchun Xu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Guozheng Jiang
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Yushan Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiawei Song
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Yukun Ma
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Guanlong Wang
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Jiaojiao Fan
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Wenhao Li
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Shibo Zhou
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China
| | - Yongdong Yang
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China.
| | - Xing Yu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, PR China.
| |
Collapse
|
3
|
Wang W, Li Y, Zhu M, Xu Q, Cui J, Liu Y, Liu Y. Danlian-Tongmai formula improves diabetic vascular calcification by regulating CCN3/NOTCH signal axis to inhibit inflammatory reaction. Front Pharmacol 2025; 15:1510030. [PMID: 39834821 PMCID: PMC11743396 DOI: 10.3389/fphar.2024.1510030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Background Vascular calcification (VC) commonly occurs in diabetes and is associated with cardiovascular disease incidence and mortality. Currently, there is no drug treatment for VC. The Danlian-Tongmai formula (DLTM) is a traditional Chinese medicine (TCM) prescription used for diabetic VC (DVC), but its mechanisms of action remain unclear. This study aims to elucidate the effects of DLTM on DVC and explore the underlying mechanisms of action. Methods Ultra-high-performance liquid chromatography-mass spectrometry (UHPLC-MS) was used to identify the metabolites of DLTM. A DVC rat model was established using streptozotocin (STZ) combined with vitamin D3 (VitD3). The effects of DLTM on DVC were evaluated through alizarin red staining, calcium deposition, and changes in osteogenic and contractile markers. The specific molecular mechanism of DLTM in treating diabetic VC was comprehensively analyzed by transcriptomics, molecular docking and in vivo experimental verification. Results We identified 108 major metabolites of DLTM. In vivo, high-dose DLTM significantly alleviated VC in diabetic rats. Transcriptomic analysis showed that DLTM treatment markedly altered the transcriptomic profile of rat aortas, which was associated with regulating the CCN3/NOTCH signaling pathway, promoting vascular smooth muscle contraction, and inhibiting the inflammatory responses. Molecular docking and molecular dynamics simulation demonstrated strong binding interactions between DLTM metabolites and key molecules within the CCN3/NOTCH pathway, including NOTCH1, DLL1, DLL4, hes1, and hey1. In vivo experiments confirmed that DLTM could upregulate CCN3, inhibit the activation of NOTCH signaling ligands DLL1 and downstream transcription factors hes1 and hey1, and reduce the release of inflammatory cytokines IL6, IL1β, and TNFα. Conclusion DLTM alleviates DVC by regulating the CCN3/NOTCH signaling axis to inhibit inflammatory responses. Our research provides experimental basis for clinical treatment and drug transformation of diabetic VC.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiwen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Xu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Cui
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Wang K, Mitoh Y, Horie K, Yoshida R. Exploring the Role of Ccn3 in Type III Cell of Mice Taste Buds. J Neurochem 2025; 169:e16291. [PMID: 39709613 DOI: 10.1111/jnc.16291] [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: 09/03/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Different taste cells express unique cell-type markers, enabling researchers to distinguish them and study their functional differentiation. Using single-cell RNA-Seq of taste cells in mouse fungiform papillae, we found that Cellular Communication Network Factor 3 (Ccn3) was highly expressed in Type III taste cells but not in Type II taste cells. Ccn3 is a protein-coding gene involved in various biological processes, such as cell proliferation, angiogenesis, tumorigenesis, and wound healing. Therefore, in this study, we aimed to explore the expression and function of Ccn3 in mouse taste bud cells. Using reverse transcription polymerase chain reaction (RT-PCR), in situ hybridization, and immunohistochemistry (IHC), we confirmed that Ccn3 was predominantly expressed in Type III taste cells. Through IHC, quantitative real-time RT-PCR, gustatory nerve recordings, and short-term lick tests, we observed that Ccn3 knockout (Ccn3-KO) mice did not exhibit any significant differences in the expression of taste cell markers and taste responses compared to wild-type controls. To explore the function of Ccn3 in taste cells, bioinformatics analyses were conducted and predicted possible roles of Ccn3 in tissue regeneration, perception of pain, protein secretion, and immune response. Among them, an immune function is the most plausible based on our experimental results. In summary, our study indicates that although Ccn3 is strongly expressed in Type III taste cells, its knockout did not influence the basic taste response, but bioinformatics provided valuable insights into the possible role of Ccn3 in taste buds and shed light on future research directions.
Collapse
Affiliation(s)
- Kuanyu Wang
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshihiro Mitoh
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kengo Horie
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
5
|
Liu J, Gao J, Wang H, Fan X, Li L, Wang X, Wang X, Lu J, Shi X, Yang P. Acute Neurobehavioral and Glial Responses to Explosion Gas Inhalation in Rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:5099-5111. [PMID: 39092980 DOI: 10.1002/tox.24389] [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: 02/22/2024] [Revised: 04/27/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Military personnel, firefighters, and fire survivors exhibit a higher prevalence of mental health conditions such as depression and post-traumatic stress disorder (PTSD) compared to the general population. While numerous studies have examined the neurological impacts of physical trauma and psychological stress, research on acute neurobehavioral effects of gas inhalation from explosions or fires is limited. This study investigates the early-stage neurobehavioral and neuronal consequences of acute explosion gas inhalation in Sprague-Dawley rats. Rats were exposed to simulated explosive gas and subsequently assessed using behavioral tests and neurobiological analyses. The high-dose exposure group demonstrated significant depression-like behaviors, including reduced mobility and exploration. However, neuronal damage was not evident in histological analyses. Immunofluorescence revealed increased density of radial glia and oligodendrocytes in specific brain regions, suggesting hypoxia and axon damage induced by gas inhalation as a potential mechanism for the observed neurobehavioral changes. These findings underscore the acute impact of explosion gas inhalation on mental health, highlighting the habenula and dentate gyrus of hippocampus as the possible target regions. The findings are expected to support early diagnosis and treatment strategies for brain injuries caused by explosion gas, offering insights into early intervention for depression and PTSD in affected populations.
Collapse
Affiliation(s)
- Jinren Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Junhong Gao
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Hong Wang
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Xiaolin Fan
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Liang Li
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Xiangni Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiying Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jiajia Lu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xingmin Shi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Pinglin Yang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
Chen H, Zhang X, Zhang Z, Li G, Li X, Yang S, Liu Y, Yang M. Identification and validation of CCN family genes to predict the prognosis in gastric cancer. Discov Oncol 2024; 15:610. [PMID: 39485579 PMCID: PMC11530581 DOI: 10.1007/s12672-024-01459-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a deadly malignancy with an ever-increasing incidence worldwide. The cellular communication network (CCN) family serves as matricellular proteins and exerts their various functions via regulating cell proliferation and differentiation. This study aimed to perform an integrated analysis of CCNs to predict the prognosis in GC. METHODS The microarray datasets were obtained from Gene Expression Omnibus database to identify the differentially expressed genes between GC and non-tumor tissues. Functional enrichment and genetic alteration analysis revealed the biological functions and alteration status associated with CCNs. We analyzed the mRNA and protein expressions of CCN family in GC patients. Furthermore, the prognostic value of distinct CCN family members were analyzed using the Kaplan-Meier plotter database. Finally, the human gastric cancer cell lines were used for in vitro experiments to further validate the role of WISP1. RESULTS 26 genes were firstly identified to be significantly highly expressed in gastric tumor tissues. CCN family genes were identified to predict the prognosis in GC. Among the six CCNs, WISP1 is upregulated in GC tissues and its highly expression is associated with poor survival in GC patients. Moreover, a significant correlation is found between the expression of WISP1 and the pathological stage of patients with GC. Additionally, in vitro experiments demonstrated that WISP1 promotes the proliferation and invasive potential of GC cells, suggesting it may be a potential therapeutic target for GC. CONCLUSIONS A comprehensive bioinformatic analysis of CCN genes provides new insights into the potential roles of this family in GC. Importantly, WISP1 may be a good prognostic predictor and a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Huanting Chen
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Department of General Surgery, People's Hospital of Shenzhen Baoan District, Affiliated Shenzhen Baoan Hospital of Southern Medical University, Shenzhen, 518101, China
| | - Xiaomin Zhang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Zhe Zhang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Guoqiang Li
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Xin Li
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Siran Yang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Yajie Liu
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China.
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China.
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China.
| | - Mengqi Yang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, Guangdong, China.
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Shenzhen, 518036, China.
- Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China.
| |
Collapse
|
7
|
Li M, Yang J, Gao L. YTHDF1 gene inhibits epilepsy progression by epigenetic activation of PTEN gene. Heliyon 2024; 10:e39481. [PMID: 39497959 PMCID: PMC11533598 DOI: 10.1016/j.heliyon.2024.e39481] [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: 07/08/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Epilepsy is a common chronic neurological disorder with high prevalence that profoundly affects millions of people worldwide. Inflammatory dysregulation affects central nervous system disorders including epilepsy, and YTHDF1, the most common "reader" of m6A and m6A-binding protein, can attenuate the inflammatory response and activate PTEN, and here we aimed to investigate its effect on epilepsy through epigenetics. All mice were injected intraperitoneally with 12 mg/kg of sea manic acid to establish an epilepsy model, and the epileptic behaviors of the mice were classified into 6 grades; epileptic behaviors of grade 3 or above were defined as seizures, and consecutive epileptic seizures of more than 30 min were considered as successful modeling. Mouse behavior was examined using the Morris Water Maze tracking assay; inflammatory factors IL-6, TNF-α, and IL-1β were detected by qPCR/WB/ELISA; cell activity was analyzed by CCK-8; apoptotic markers were identified by immunofluorescence assay and Western blot analysis. YTHDF1 knockout mice have poor spatial memory capacity and sensitivity to external stimuli. Under the influence of YTHDF1, the neuroinflammation and nseuron death decreased. YTHDF1 works by repressing the production of pro-inflammatory cytokines and the activation of astrocytes. It was found that YTHDF1 epigenetically activates PTEN through m6A modification, activates glial cells and represses pro-inflammatory cytokines production and inhibits the development of epilepsy.
Collapse
Affiliation(s)
- Mingxia Li
- Department of Paediatrics, Yantaishan Hospital, Yantai, Shandong, 264001, China
| | - Junli Yang
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, 264100, China
| | - Lixiang Gao
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, 264100, China
| |
Collapse
|
8
|
Chen M, Guo L, Li Q, Yang S, Li W, Lai Y, Lv Z. Research progress on hippocampal neurogenesis in autism spectrum disorder. Pediatr Investig 2024; 8:215-223. [PMID: 39347523 PMCID: PMC11427895 DOI: 10.1002/ped4.12440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/10/2024] [Indexed: 10/01/2024] Open
Abstract
Autism spectrum disorder (ASD) is a group of severe neurodevelopmental disorders with unclear etiology and significant heterogeneity that is emerging as a global public health concern. Increasing research suggests the involvement of hippocampal neurogenesis defects in the onset and development of ASD, drawing increasing amounts of attention to hippocampal neurogenesis issues in ASD. In this paper, we analyze relevant international studies on hippocampal neurogenesis in ASD, discuss the role of neurobiology in the pathogenesis of ASD, and explore the potential of improving hippocampal neurogenesis as a therapeutic approach for ASD. This review aims to provide new treatment perspectives and theoretical foundations for clinical practice.
Collapse
Affiliation(s)
- Mengxiang Chen
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| | - Lanmin Guo
- Department of Child HealthThe Third Affiliated Hospital of Jiamusi UniversityHeilongjiangChina
| | - Qinghong Li
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| | - Shunbo Yang
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| | - Wei Li
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| | - Yanmei Lai
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| | - Zhihai Lv
- Department of Pediatric RehabilitationLonggang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)GuangdongChina
| |
Collapse
|
9
|
Carter SWD, Fee EL, Usuda H, Oguz G, Ramasamy A, Amin Z, Agnihotri B, Wei Q, Xiawen L, Takahashi T, Takahashi Y, Ikeda H, Kumagai Y, Saito Y, Saito M, Mattar C, Evans MI, Illanes SE, Jobe AH, Choolani M, Kemp MW. Antenatal steroids elicited neurodegenerative-associated transcriptional changes in the hippocampus of preterm fetal sheep independent of lung maturation. BMC Med 2024; 22:338. [PMID: 39183288 PMCID: PMC11346182 DOI: 10.1186/s12916-024-03542-5] [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: 05/12/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Antenatal steroid therapy for fetal lung maturation is routinely administered to women at risk of preterm delivery. There is strong evidence to demonstrate benefit from antenatal steroids in terms of survival and respiratory disease, notably in infants delivered at or below 32 weeks' gestation. However, dosing remains unoptimized and lung benefits are highly variable. Current treatment regimens generate high-concentration, pulsatile fetal steroid exposures now associated with increased risk of childhood neurodevelopmental diseases. We hypothesized that damage-associated changes in the fetal hippocampal transcriptome would be independent of preterm lung function. METHODS Date-mated ewes carrying a single fetus at 122 ± 2dGA (term = 150dGA) were randomized into 4 groups: (i) Saline Control Group, 4×2ml maternal saline intramuscular(IM) injections at 12hr intervals (n = 11); or (ii) Dex High Group, 2×12mg maternal IM dexamethasone phosphate injections at 12hr intervals followed by 2×2ml IM saline injections at 12hr intervals (n = 12; representing a clinical regimen used in Singapore); or (iii) Dex Low Group, 4×1.5mg maternal IM dexamethasone phosphate injections 12hr intervals (n = 12); or (iv) Beta-Acetate Group, 1×0.125mg/kg maternal IM betamethasone acetate injection followed by 3×2ml IM sterile normal saline injections 12hr intervals (n = 8). Lambs were surgically delivered 48hr after first maternal injection at 122-125dGA, ventilated for 30min to establish lung function, and euthanised for necropsy and tissue collection. RESULTS Preterm lambs from the Dex Low and Beta-Acetate Groups had statistically and biologically significant lung function improvements (measured by gas exchange, lung compliance). Compared to the Saline Control Group, hippocampal transcriptomic data identified 879 differentially significant expressed genes (at least 1.5-fold change and FDR < 5%) in the steroid-treated groups. Pulsatile dexamethasone-only exposed groups (Dex High and Dex Low) had three common positively enriched differentially expressed pathways related in part to neurodegeneration ("Prion Disease", "Alzheimer's Disease", "Arachidonic Acid metabolism"). Adverse changes were independent of respiratory function during ventilation. CONCLUSIONS Our data suggests that exposure to antenatal steroid therapy is an independent cause of damage- associated transcriptomic changes in the brain of preterm, fetal sheep. These data highlight an urgent need for careful reconsideration and balancing of how antenatal steroids are used, both for patient selection and dosing regimens.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Zubair Amin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Biswas Agnihotri
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Qin Wei
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Liu Xiawen
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hideyuki Ikeda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Citra Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Mark I Evans
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Fetal Medicine Foundation of America, New York, NY, USA
| | - Sebastián E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Reproductive Biology Program, Center for Biomedical Research and Innovation, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Alan H Jobe
- Centre for Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Women and Infants Research Foundation, Perth, WA, Australia
| |
Collapse
|
10
|
Guo Z, Zhang H, Huang T, Liu C. CCN3/NOV inhibition attenuates oxidative stress-induced apoptosis of mouse neural stem/progenitor cells by blocking the activation of p38 MAPK: An in vitro study. Brain Res 2024; 1827:148756. [PMID: 38199307 DOI: 10.1016/j.brainres.2024.148756] [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: 11/14/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Neural stem/progenitor cells (NSPCs) hold immense promise in clinical applications, yet the harsh conditions resulting from central nervous system (CNS) injuries, particularly oxidative stress, lead to the demise of both native and transplanted NSPCs. Cellular communication network factor 3 (CCN3) exhibits a protective effect against oxidative stress in various cell types. This study investigates the impact of CCN3 on NSPCs apoptosis induced by oxidative stress. To establish models of primary cultured mouse NSPCs under oxidative stress, we exposed them to 50 μM H2O2 for 4 h. Remarkably, pre-exposing CCN3 exacerbated the H2O2-induced decline in cell viability in a concentration-dependent manner. However, employing gene-targeted siRNA to inhibit CCN3 protected NSPCs against H2O2-induced cell death. Conversely, CCN3 replenishment reversed this protective effect, as evidenced by TUNEL staining, the ratio of Cleaved-caspase-3 to Pro-caspase-3, and Bcl-2/Bax. Further investigations revealed that CCN3 pretreatment increased the phosphorylation level of p38 MAPK, while silencing CCN3 diminished p38 MAPK activation. Ultimately, the impact of changes in CCN3 protein expression on H2O2-induced apoptosis was nullified using anisomycin (a p38 activator) and SB 203580 (a p38 inhibitor). Our findings suggest that CCN3 inhibition prevents H2O2-induced cell death in cultured mouse NSPCs via the p38 pathway. These discoveries may contribute to the development of strategies aimed at enhancing the survival of both endogenous and transplanted NSPCs following CNS oxidative stress insults.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanyue Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Tingqin Huang
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chongxiao Liu
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|