1
|
Zhan P, Huang S, Chen D, Li Y, Chen D. Echinatin inhibits osteoarthritis through the NF-κB signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7363-7375. [PMID: 39747466 DOI: 10.1007/s00210-024-03756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Abstract
Osteoarthritis (OA) is currently the most common degenerative joint disease in China and even worldwide and is the leading cause of disability in the elderly population. So far, due to an insufficient understanding of the pathogenesis and etiology of the disease, there is still no effective targeted treatment for early OA. Pro-inflammatory cytokine interleukin-1 is an important inflammatory mediator secreted in early OA, and IL-1β plays a crucial role in the pathogenesis of OA, affecting chondrocytes and the extracellular matrix of CARTILAGE. Echinatin has been used for years as a health supplement, retaining its antioxidant, anti-inflammatory, and autophagy-promoting effects. However, whether echinatin has inhibitory effects on OA is still unknown. In this study, we used an in vitro OA model of chondrocytes induced by IL-1β and an in vivo OA model of rats induced by anterior cruciate ligament transection (ACLT), and through experiments such as western blotting and IHC, we demonstrated that echinatin can be used as a novel drug for treating OA. Mechanistically, we found that echinatin inhibits the activity of chondrocytes induced by IL-1β through the NF-kB signaling pathway. This study can provide more effective treatment options for OA patients and further diagnostic and therapeutic methods for clinical treatment.
Collapse
Affiliation(s)
- Peng Zhan
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Shiming Huang
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Daohua Chen
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Ying Li
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Dongfeng Chen
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China.
| |
Collapse
|
2
|
Dong Z, Dong Z, Xu L, Zhang J, Li L, Wang R, Huang X, Zou Z. Vortioxetine alleviates motor, cognitive and emotional disorders in post-stroke rats by regulating the TLR-2/NF- κB pathway. Front Pharmacol 2025; 16:1555079. [PMID: 40144659 PMCID: PMC11937001 DOI: 10.3389/fphar.2025.1555079] [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: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 03/28/2025] Open
Abstract
Cognitive impairments following post-stroke significantly hinder neurological recovery and exacerbate patient morbidity, underscoring urgent need for effective therapeutic strategies. Vortioxetine (VTX), a prominent Selective Serotonin Reuptake Inhibitor (SSRI), boasts notable antidepressant, cognition-enhancing, and anti-inflammatory properties. This investigation delves into VTX's influence on motor skills, spatial learning-memory capabilities, and depressive behaviors in Middle Cerebral Artery Occlusion (MCAO) rats, alongside its underlying mechanisms. Our findings reveal that while VTX fails to entirely reverse ischemic-reperfusion damage, it substantially ameliorates spontaneous locomotor functions, augments post-stroke learning-memory capacities, and exhibits potent antidepressant and anxiety-like efficacy. Preliminary data propose that these beneficial effects may stem from inflammation modulation via the Toll-Like Receptor 2 (TLR-2)/Nuclear Factor-Kappa B (NF-κB) signaling pathway. Collectively, our work underscores VTX's promising role in enhancing motor, cognitive functions, and mitigating depressive symptoms following cerebrovascular accidents, potentially through inflammation regulation. These insights pave the way for novel interventions addressing post-stroke complications, warranting further exploration.
Collapse
Affiliation(s)
- Ziqiang Dong
- Department of Anesthesiology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Zhihui Dong
- Department of Planning and Quality Control, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Lili Xu
- Intravenous Drug Dispensing center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Jinfeng Zhang
- Department of Planning and Quality Control, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Lin Li
- Intravenous Drug Dispensing center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Rongjuan Wang
- Intravenous Drug Dispensing center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Xiaoyan Huang
- Intravenous Drug Dispensing center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Zhengqiang Zou
- Department of Anesthesiology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
- Department of Planning and Quality Control, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
- Intravenous Drug Dispensing center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
- Ganzhou Rongjiang New Area People’s Hospital, Ganzhou, China
| |
Collapse
|
3
|
Jiang Y, Wu Q, Dang Y, Peng L, Meng L, You C. Untargeted metabolomics unveils critical metabolic signatures in novel phenotypes of acute ischemic stroke. Metab Brain Dis 2025; 40:130. [PMID: 39969622 DOI: 10.1007/s11011-024-01451-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/01/2024] [Indexed: 02/20/2025]
Abstract
This study aimed to identify metabolic footprints associated with distinct phenotypes of acute ischemic stroke (AIS) using untargeted metabolomics. We included 20 samples each from AIS phenotype A (n = 251), B (n = 213), and C (n = 43) groups, along with 20 age- and gender-matched healthy controls (HCs). Plasma metabolic profiles were analyzed using liquid chromatography-mass spectrometry (LC-MS). Weighted gene correlation network analysis (WGCNA) evaluated associations between metabolite clusters and clinical traits, including the National Institutes of Health Stroke Scale (NIHSS) and the modified Rankin Scale (mRS). We identified three, five, and six key differential metabolites for diagnosing phenotypes A, B, and C, respectively, demonstrating high diagnostic performance. These metabolites were focused on fatty acids, sex hormones, amino acids, and their derivatives. WGCNA identified 12 core metabolites involved in phenotype progression. Notably, phenylalanylphenylalanine and phenylalanylleucine were inversely correlated with disease severity and disability. Metabolites related to energy supply and inflammation were common across phenotypes, with additional changes in ionic homeostasis in phenotype A and decreased neurotransmitter release in phenotype C. Biosynthesis of unsaturated fatty acids and the pentose phosphate pathway (PPP) were relevant across all phenotypes, while the folate biosynthesis pathway was linked to phenotype C and clinical scales. Key metabolites, including phenylalanylphenylalanine and phenylalanylleucine, and pathways such as folate biosynthesis, significantly contribute to AIS severity and differentiation of phenotypes. These findings offer new insights into the pathogenesis and mechanisms underlying AIS phenotypes.
Collapse
Affiliation(s)
- Yao Jiang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Qian Wu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yingqiang Dang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Lingling Peng
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ling Meng
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
4
|
Li W, Feng X, Zhang M, Wang K, Huang K, Zhao Z, Xia M. CLEC7A Knockdown Alleviates Ischemic Stroke by Inhibiting Pyroptosis and Microglia Activation. J Integr Neurosci 2024; 23:219. [PMID: 39735961 DOI: 10.31083/j.jin2312219] [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: 04/15/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is the leading cause of mortality worldwide. Herein, we aimed to identify novel biomarkers and explore the role of C-type lectin domain family 7 member A (CLEC7A) in IS. METHODS Differentially expressed genes (DEGs) were screened using the GSE106680, GSE97537, and GSE61616 datasets, and hub genes were identified through construction of protein-protein interaction networks. An IS model was established by middle cerebral artery occlusion and reperfusion (MCAO/R). Neural function was assessed using triphenyl tetrazolium chloride, hematoxylin-eosin, and terminal deoxynucleotidyl transferase-mediated nick-end labeling. A cell counting kit was used to detect cell viability following oxygen-glucose deprivation/reperfusion (OGD/R). Inflammatory factors were detected using enzyme-linked immunosorbent assay. The mRNA and protein expression levels were detected using reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. RESULTS Fc fragment of Immunoglobulin G (IgG) receptor IIIa (FCGR3A), Fc fragment of Immunoglobulin E (IgE) receptor Ig (FCER1G), Complement component 5a receptor 1 (C5AR1), CLEC7A, Plasminogen activator, urokinase (PLAU), and C-C motif chemokine ligand 6 (CCL6) were identified as important hub genes, from which CLEC7A was selected as the primary subject of this study. The activation of microglia and pyroptosis were observed in MCAO/R model with increased levels of interleukin (IL)-1β, IL-18, tumor necrosis factor-α, and lactate dehydrogenase. CLEC7A knockdown was found to promote cell viability in BV2 cells and inhibiting pyroptosis in HT22 cells. CLEC7A knockdown in microglia also decreased infarct volume and neurological deficit scores, and alleviated injury and neuronal apoptosis in IS rats. CLEC7A knockdown inhibited pyroptosis and microglial activation in the MCAO/R model. A pyroptosis activator reversed the effect of CLEC7A knockdown on the viability of OGD/R-treated HT22 cells. CONCLUSION CLEC7A is a promising biomarker of IS. CLEC7A knockdown alleviates IS by inhibiting pyroptosis and microglial activation.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Xiaoli Feng
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Manyu Zhang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Kangmeng Wang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Kailai Huang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Zhenqiang Zhao
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Min Xia
- Department of Neurology, Hainan West Central Hospital, 571799 Danzhou, Hainan, China
| |
Collapse
|
5
|
Yin H, Shen J, Qian X, Zhai L, Guan Q, Shen H, Wang G. Dimethyl phthalate exposure induces cognitive impairment via COX2-mediated neuroinflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:117039. [PMID: 39270417 DOI: 10.1016/j.ecoenv.2024.117039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
AIM The present work explored the mechanism of dimethyl phthalate (DMP, the environmental contaminant) exposure in inducing cognitive impairment. METHODS Targets and regulatory networks related to DMP-brain injury-cognitive impairment were analyzed through network pharmacology. DMP exposure was carried out to simulate DMP environmental uptake, whereas Morris water maze was performed for examining cognitive impairment. Additionally, inflammatory cytokine levels within tissues were measured. hematoxylin-eosin staining(H&E) and Nissl staining was conducted to examine brain tissue injury, while Western blot was carried out for identifying protein levels. After applying.Small interfering RNA(siRNA-COX2) and celecoxib-COX2 inhibitors separately, we analyzed impacts of DMP. Besides, in vitro experiments were performed to analyze impacts of DMP on microglial activation. RESULTS As suggested by network pharmacology,Cyclooxygenase-2-PTGS2 (COX2) showed significant relation to DMP, and it exerted its effect via COX2. Following DMP exposure, mice experienced obvious cognitive impairment and brain damage, besides, microglial cells were activated, and inflammatory cytokines were up-regulated. Applying siRNA-COX2 and celecoxib-COX2 suppressed DMP's impact and mitigated mouse cognitive impairment. Based on in vitro analysis, DMP led to microglial activation and neuroinflammation. CONCLUSION DMP exposure causes neuroinflammation via the COX2-regulated microglial activation, thus leading to cognitive impairment. COX2 may serve as the key action target of DMP.
Collapse
Affiliation(s)
- Haoyuan Yin
- The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jian Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, China.
| | | | - Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Genghuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
6
|
Du R, Pei H, He Z, Wang J, Zhou X, Li W, Zhu D, Zhang C. Astragalin improves cognitive disorder in Alzheimer's disease: Based on network pharmacology and molecular docking simulation. CNS Neurosci Ther 2024; 30:e14799. [PMID: 39107952 PMCID: PMC11303257 DOI: 10.1111/cns.14799] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/03/2024] [Accepted: 05/26/2024] [Indexed: 08/10/2024] Open
Abstract
We investigate the mechanism of action of astragalin (AST) in the treatment of Alzheimer's disease (AD). Network pharmacology was conducted to analyze the relationships among AST, AD, and neuroinflammation, The APP/PS1 transgenic mice with AD were used in the experiments; to be specific, the influence of AST on the behavior of mice was analyzed by Morris water maze and eight-arm radial maze tests, the tissue inflammatory factor levels were detected by ELISA, and pathological changes were analyzed by H&E and immunohistochemical staining. Analysis results of network pharmacology suggested that AST exerted the multi-target effect on neuroinflammation in AD. Through molecular docking and dynamics analyses, COX2 might be the target of AST. Moreover, animal experimental results demonstrated that AST improved the behavior of AD mice, and enhanced the motor and memory abilities, meanwhile, it suppressed the expression of inflammatory factors in tissues and the activation of microglial cells. this study discovers that AST can suppress microglial cell activation via COX2 to improve neuroinflammation in AD.
Collapse
Affiliation(s)
- Rui Du
- College of Chinese Medicinal MaterialsJilin Agricultural UniversityChangchunChina
| | - Hongyan Pei
- College of Chinese Medicinal MaterialsJilin Agricultural UniversityChangchunChina
| | - Zhongmei He
- College of Chinese Medicinal MaterialsJilin Agricultural UniversityChangchunChina
| | - Jin Wang
- The Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xiaohong Zhou
- The Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Wenyan Li
- The Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Diwei Zhu
- The Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Caiqun Zhang
- The Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| |
Collapse
|
7
|
Tao X, Hu Y, Mao N, Shen M, Fang M, Zhang M, Lou J, Fang Y, Guo X, Lin Z. Echinatin alleviates inflammation and pyroptosis in hypoxic-ischemic brain damage by inhibiting TLR4/ NF-κB pathway. Int Immunopharmacol 2024; 136:112372. [PMID: 38850784 DOI: 10.1016/j.intimp.2024.112372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Hypoxic ischemic encephalopathy (HIE) is a primary cause of neonatal death and disabilities. The pathogenetic process of HIE is closely associated with neuroinflammation. Therefore, targeting and suppressing inflammatory pathways presents a promising therapeutic strategy for the treatment of HIE. Echinatin is an active component of glycyrrhiza, with anti-inflammatory and anti-oxidative properties. It is commonly combined with other traditional Chinese herbs to exert heat-clearing and detoxifying effects. This study aimed to investigate the anti-inflammatory and neuroprotective effects of Echinatin in neonatal rats with hypoxic-ischemic brain damage, as well as in PC12 cells exposed to oxygen-glucose deprivation (OGD). In vivo, Echinatin effectively reduced cerebral edema and infarct volume, protected brain tissue morphology, improved long-term behavioral functions, and inhibited microglia activation. These effects were accompanied by the downregulation of inflammatory factors and pyroptosis markers. The RNA sequencing analysis revealed an enrichment of inflammatory genes in rats with hypoxic-ischemic brain damage, and Protein-protein interaction (PPI) network analysis identified TLR4, MyD88, and NF-κB as the key regulators. In vitro, Echinatin reduced the levels of TLR4 relevant proteins, inhibited nuclear translocation of NF-κB, reduced the expression of downstreams inflammatory cytokines and pyroptosis proteins, and prevented cell membrane destructions. These findings demonstrated that Echinatin could inhibit the TLR4/NF-κB pathway, thereby alleviating neuroinflammation and pyroptosis. This suggests that Echinatin could be a potential candidate for the treatment of HIE.
Collapse
Affiliation(s)
- Xiaoyue Tao
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Yingying Hu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Niping Mao
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Ming Shen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Mingchu Fang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Min Zhang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Jia Lou
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Yu Fang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China
| | - Xiaoling Guo
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China; Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang,325027, China.
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|