1
|
Rosendo-Chalma P, Díaz-Landy EN, Antonio-Véjar V, Ortiz Tejedor JG, Reytor-González C, Simancas-Racines D, Bigoni-Ordóñez GD. Endometriosis: Challenges in Clinical Molecular Diagnostics and Treatment. Int J Mol Sci 2025; 26:3979. [PMID: 40362218 PMCID: PMC12072088 DOI: 10.3390/ijms26093979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Endometriosis is a chronic disease affecting approximately 10% (190 million) of women and girls of reproductive age worldwide. It is associated with a variety of often debilitating symptoms, including severe pelvic pain, pain during intercourse, bowel movements and/or urination, bloating, nausea, fatigue, risk of infertility, as well as depression and anxiety in some cases. This review summarized the pathogenesis of endometriosis and the criteria for clinical diagnosis, proposed a panel of potential biomarkers for predictive molecular diagnosis, as well as choice of treatments for pain and infertility management.
Collapse
Affiliation(s)
- Pedro Rosendo-Chalma
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México (IIB-UNAM), Mexico City 14080, Mexico;
- Unidad Académica de Salud y Bienestar, Carrera de Bioquímica y Farmacia, Universidad Católica de Cuenca, Cuenca 010101, Ecuador;
- Unidad Académica de Posgrado, Maestría en Diagnóstico de Laboratorio Clínico y Molecular, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Erick Nicolás Díaz-Landy
- Unidad de Ginecología y Obstetricia, Hospital Santa Inés, Cuenca 010107, Ecuador;
- Ginecología y Obstetricia, Universidad del Azuay, Cuenca 010204, Ecuador
| | - Verónica Antonio-Véjar
- Laboratorio de Virología y Patología Traslacional, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico;
| | - Jonnathan Gerardo Ortiz Tejedor
- Unidad Académica de Salud y Bienestar, Carrera de Bioquímica y Farmacia, Universidad Católica de Cuenca, Cuenca 010101, Ecuador;
- Unidad Académica de Posgrado, Maestría en Diagnóstico de Laboratorio Clínico y Molecular, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Claudia Reytor-González
- Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador;
| | - Daniel Simancas-Racines
- Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador;
| | | |
Collapse
|
2
|
Dou S, Wei Y, Lin Z, Wu H, Yang F, Cen X, Lu W, Qin H, Wang R, Wang J. A new perspective on endometriosis: Integrating eQTL mendelian randomization with transcriptomics and single-cell data analyses. Funct Integr Genomics 2025; 25:75. [PMID: 40140093 PMCID: PMC11947010 DOI: 10.1007/s10142-025-01543-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 03/28/2025]
Abstract
Endometriosis is caused by the migration of endometrial cells to locations outside the uterine lining. Despite the increasing prevalence of endometriosis, there has been limited research on genetic effects, and its molecular mechanisms remain unclear. This study aimed to investigate the mechanisms underlying the development of endometriosis and to identify new genetic targets for endometriosis by integrating data from gene chips, single-cell mapping, and genome-wide association study databases. Using the Gene Expression Omnibus database, we downloaded data on normal endometrium, eutopic endometrium, and ectopic lesion tissues to explore the differentially expressed genes (DEGs) between normal and eutopic endometrium, and between eutopic and ectopic endometrium. Assessment of the relationships between DEGs and endometriosis involved differential expression, expression quantitative trait loci (eQTL), and Mendelian randomization (MR) analyses. Two single-cell atlas datasets were then analyzed to explore the mechanisms underlying disease development and progression. Intersection of MR results with DEGs between normal and eutopic endometrium highlighted 28 candidate biomarker genes (17 upregulated and 11 downregulated). Similarly, we identified two additional candidate biomarker genes by intersecting the DEGs between eutopic and ectopic endometrium with MR results. Among these 30 candidates, further filtering using single-cell datasets revealed that the histamine N-methyltransferase (HNMT), coiled-coil domain containing 28 A (CCDC28A), fatty acid desaturase 1 (FADS1) and mahogunin ring finger 1 (MGRN1) genes were differentially expressed between the normal and eutopic groups, consistent with transcriptomic and MR results. Our findings suggested that eutopic endometrium exhibits epithelial-mesenchymal transition (EMT). Cell communication analysis focused on ciliated epithelial cells expressing CDH1 and KRT23 revealed that, in the eutopic endometrium, ciliated epithelial cells are strongly correlated and interact with natural killer cells, T cells, and B cells. We identified four novel biomarker genes and found evidence for EMT in the eutopic endometrium. The mechanism of endometriosis progression may be closely related to EMT and changes in the immune microenvironment triggered by damage to ciliated epithelial cells.
Collapse
Affiliation(s)
- Sheng Dou
- The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Yi Wei
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Zongyun Lin
- The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Hui Wu
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Fenglian Yang
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Xuechang Cen
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Wenjing Lu
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Haimei Qin
- The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
| | - Rong Wang
- The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China
- Blood transfusion department, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Junli Wang
- The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
- Youjiang Medical University for Nationalities, Baise, China.
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
3
|
Liu X, Zhang D, Qiu H. NMF typing and machine learning algorithm-based exploration of preeclampsia-related mechanisms on ferroptosis signature genes. Cell Biol Toxicol 2024; 41:14. [PMID: 39707003 PMCID: PMC11662041 DOI: 10.1007/s10565-024-09963-5] [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: 08/18/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Globally, pre-eclampsia (PE) poses a major threat to the health and survival of pregnant women and fetuses, contributing significantly to morbidity and mortality. Recent studies suggest a pathological link between PE and ferroptosis. We aim to utilize non-negative matrix factorization (NMF) clustering and machine learning algorithms to pinpoint disease-specific genes related to the process of ferroptosis in PE and investigate likely underlying biochemistry mechanisms. METHODS The acquisition of four microarray datasets from the Gene Expression Omnibus (GEO) repository, the integration of these datasets, and the elimination of batch effects formed the core procedure. Genes related to ferroptosis in PE (DE-FRG) were identified. NMF clustering was performed on DE-FRG for unsupervised analysis, generating a heatmap for clustering validation via principal component analysis. Immunocyte infiltration differences between different subtypes were compared to elucidate the impact of ferroptosis on immune infiltration in the placental tissue of PE patients. The application of weighted gene co-expression network analysis (WGCNA) revealed important module genes linked to sample subtypes and disease status. The screening of PE feature genes involved employing SVM, RF, GLM, and XGB machine learning algorithms, and their predictive performance was validated using various analyses and an external dataset. The iRegulon tool was utilized to predict upstream transcription factors associated with ferroptosis feature genes, from which differentially expressed transcription factors were screened to construct a "Transcription Factor-FRG-ferroptosis" regulatory network. Finally, in vitro (cultured cells) and in vivo (rat) models were utilized to evaluate the regulatory mechanisms of ferroptosis in normal and PE placental tissues. RESULTS Differential analysis of the four merged GEO datasets identified 41 DE-FRGs. NMF clustering based on DE-FRGs revealed two PE subtypes. Immunocyte infiltration analysis indicated significant differences in immune levels between these subtypes. Further WGCNA analysis identified module genes associated with PE and these two subtypes. Subsequently, we developed an integrated machine learning model incorporating five FRGs and validated its predictive efficacy using various analyses and an external validation dataset. Finally, based on the transcription factor ARID3A and ferroptosis feature genes EPHB3 and PAPPA2, we constructed a "Transcription Factor-FRG-ferroptosis" regulatory network, with in vitro and in vivo experiments confirming that ARID3A promotes the progression of PE and ferroptosis by activating the expression of EPHB3 and PAPPA2. CONCLUSION This analytical journey illuminated a critical regulatory nexus in PE, underscoring the central influence of ARID3A on PE through ferroptosis-mediated pathways.
Collapse
Affiliation(s)
- Xuemin Liu
- Department of Obsterics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Di Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Hui Qiu
- Department of Obsterics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
4
|
Wang Y, Liu Y, Wang N, Liu Z, Qian G, Li X, Huang H, Zhuo W, Xu L, Zhang J, Lv H, Gao Y. Identification of novel mitophagy-related biomarkers for Kawasaki disease by integrated bioinformatics and machine-learning algorithms. Transl Pediatr 2024; 13:1439-1456. [PMID: 39263286 PMCID: PMC11384439 DOI: 10.21037/tp-24-230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024] Open
Abstract
Background Kawasaki disease (KD) is a systemic vasculitis primarily affecting the coronary arteries in children. Despite growing attention to its symptoms and pathogenesis, the exact mechanisms of KD remain unclear. Mitophagy plays a critical role in inflammation regulation, however, its significance in KD has only been minimally explored. This study sought to identify crucial mitophagy-related biomarkers and their mechanisms in KD, focusing on their association with immune cells in peripheral blood. Methods This research used four datasets from the Gene Expression Omnibus (GEO) database that were categorized as the merged and validation datasets. Screening for differentially expressed mitophagy-related genes (DE-MRGs) was conducted, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. A weighted gene co-expression network analysis (WGCNA) identified the hub module, while machine-learning algorithms [random forest-recursive feature elimination (RF-RFE) and support vector machine-recursive feature elimination (SVM-RFE)] pinpointed the hub genes. Receiver operating characteristic (ROC) curves were generated for these genes. Additionally, the CIBERSORT algorithm was used to assess the infiltration of 22 immune cell types to explore their correlations with hub genes. Interactions between transcription factors (TFs), genes, and Gene-microRNAs (miRNAs) of hub genes were mapped using the NetworkAnalyst platform. The expression difference of the hub genes was validated using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Results Initially, 306 DE-MRGs were identified between the KD patients and healthy controls. The enrichment analysis linked these MRGs to autophagy, mitochondrial function, and inflammation. The WGCNA revealed a hub module of 47 KD-associated DE-MRGs. The machine-learning algorithms identified cytoskeleton-associated protein 4 (CKAP4) and serine-arginine protein kinase 1 (SRPK1) as critical hub genes. In the merged dataset, the area under the curve (AUC) values for CKAP4 and SRPK1 were 0.933 [95% confidence interval (CI): 0.901 to 0.964] and 0.936 (95% CI: 0.906 to 0.966), respectively, indicating high diagnostic potential. The validation dataset results corroborated these findings with AUC values of 0.872 (95% CI: 0.741 to 1.000) for CKAP4 and 0.878 (95% CI: 0.750 to 1.000) for SRPK1. The CIBERSORT analysis connected CKAP4 and SRPK1 with specific immune cells, including activated cluster of differentiation 4 (CD4) memory T cells. TFs such as MAZ, SAP30, PHF8, KDM5B, miRNAs like hsa-mir-7-5p play essential roles in regulating these hub genes. The qRT-PCR results confirmed the differential expression of these genes between the KD patients and healthy controls. Conclusions CKAP4 and SRPK1 emerged as promising diagnostic biomarkers for KD. These genes potentially influence the progression of KD through mitophagy regulation.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Nana Wang
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zhiheng Liu
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xuan Li
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Hongbiao Huang
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Wenyu Zhuo
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lei Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jiaying Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Yang Gao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang (Lianyungang Clinical College of Nanjing Medical University), Lianyungang, China
| |
Collapse
|
5
|
Pei FL, Jia JJ, Lin SH, Chen XX, Wu LZ, Lin ZX, Sun BW, Zeng C. Construction and evaluation of endometriosis diagnostic prediction model and immune infiltration based on efferocytosis-related genes. Front Mol Biosci 2024; 10:1298457. [PMID: 38370978 PMCID: PMC10870152 DOI: 10.3389/fmolb.2023.1298457] [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: 09/21/2023] [Accepted: 12/07/2023] [Indexed: 02/20/2024] Open
Abstract
Background: Endometriosis (EM) is a long-lasting inflammatory disease that is difficult to treat and prevent. Existing research indicates the significance of immune infiltration in the progression of EM. Efferocytosis has an important immunomodulatory function. However, research on the identification and clinical significance of efferocytosis-related genes (EFRGs) in EM is sparse. Methods: The EFRDEGs (differentially expressed efferocytosis-related genes) linked to datasets associated with endometriosis were thoroughly examined utilizing the Gene Expression Omnibus (GEO) and GeneCards databases. The construction of the protein-protein interaction (PPI) and transcription factor (TF) regulatory network of EFRDEGs ensued. Subsequently, machine learning techniques including Univariate logistic regression, LASSO, and SVM classification were applied to filter and pinpoint diagnostic biomarkers. To establish and assess the diagnostic model, ROC analysis, multivariate regression analysis, nomogram, and calibration curve were employed. The CIBERSORT algorithm and single-cell RNA sequencing (scRNA-seq) were employed to explore immune cell infiltration, while the Comparative Toxicogenomics Database (CTD) was utilized for the identification of potential therapeutic drugs for endometriosis. Finally, immunohistochemistry (IHC) and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were utilized to quantify the expression levels of biomarkers in clinical samples of endometriosis. Results: Our findings revealed 13 EFRDEGs associated with EM, and the LASSO and SVM regression model identified six hub genes (ARG2, GAS6, C3, PROS1, CLU, and FGL2). Among these, ARG2, GAS6, and C3 were confirmed as diagnostic biomarkers through multivariate logistic regression analysis. The ROC curve analysis of GSE37837 (AUC = 0.627) and GSE6374 (AUC = 0.635), along with calibration and DCA curve assessments, demonstrated that the nomogram built on these three biomarkers exhibited a commendable predictive capacity for the disease. Notably, the ratio of nine immune cell types exhibited significant differences between eutopic and ectopic endometrial samples, with scRNA-seq highlighting M0 Macrophages, Fibroblasts, and CD8 Tex cells as the cell populations undergoing the most substantial changes in the three biomarkers. Additionally, our study predicted seven potential medications for EM. Finally, the expression levels of the three biomarkers in clinical samples were validated through RT-qPCR and IHC, consistently aligning with the results obtained from the public database. Conclusion: we identified three biomarkers and constructed a diagnostic model for EM in this study, these findings provide valuable insights for subsequent mechanistic research and clinical applications in the field of endometriosis.
Collapse
Affiliation(s)
- Fang-Li Pei
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jin-Jin Jia
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu-Hong Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Xin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li-Zheng Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zeng-Xian Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo-Wen Sun
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Zeng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Xie Y, Shi H, Han B. Bioinformatic analysis of underlying mechanisms of Kawasaki disease via Weighted Gene Correlation Network Analysis (WGCNA) and the Least Absolute Shrinkage and Selection Operator method (LASSO) regression model. BMC Pediatr 2023; 23:90. [PMID: 36829193 PMCID: PMC9951419 DOI: 10.1186/s12887-023-03896-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/07/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Kawasaki disease (KD) is a febrile systemic vasculitis involvingchildren younger than five years old. However, the specific biomarkers and precise mechanisms of this disease are not fully understood, which can delay the best treatment time, hence, this study aimed to detect the potential biomarkers and pathophysiological process of KD through bioinformatic analysis. METHODS The Gene Expression Omnibus database (GEO) was the source of the RNA sequencing data from KD patients. Differential expressed genes (DEGs) were screened between KD patients and healthy controls (HCs) with the "limma" R package. Weighted gene correlation network analysis (WGCNA) was performed to discover the most corresponding module and hub genes of KD. The node genes were obtained by the combination of the least absolute shrinkage and selection operator (LASSO) regression model with the top 5 genes from five algorithms in CytoHubba, which were further validated with the receiver operating characteristic curve (ROC curve). CIBERSORTx was employed to discover the constitution of immune cells in KDs and HCs. Functional enrichment analysis was performed to understand the biological implications of the modular genes. Finally, competing endogenous RNAs (ceRNA) networks of node genes were predicted using online databases. RESULTS A total of 267 DEGs were analyzed between 153 KD patients and 92 HCs in the training set, spanning two modules according to WGCNA. The turquoise module was identified as the hub module, which was mainly enriched in cell activation involved in immune response, myeloid leukocyte activation, myeloid leukocyte mediated immunity, secretion and leukocyte mediated immunity biological processes; included type II diabetes mellitus, nicotinate and nicotinamide metabolism, O-glycan biosynthesis, glycerolipid and glutathione metabolism pathways. The node genes included ADM, ALPL, HK3, MMP9 and S100A12, and there was good performance in the validation studies. Immune cell infiltration analysis revealed that gamma delta T cells, monocytes, M0 macrophage, activated dendritic cells, activated mast cells and neutrophils were elevated in KD patients. Regarding the ceRNA networks, three intact networks were constructed: NEAT1/NORAD/XIST-hsa-miR-524-5p-ADM, NEAT1/NORAD/XIST-hsa-miR-204-5p-ALPL, NEAT1/NORAD/XIST-hsa-miR-524-5p/hsa-miR-204-5p-MMP9. CONCLUSION To conclude, the five-gene signature and three ceRNA networks constructed in our study are of great value in the early diagnosis of KD and might help to elucidate our understanding of KD at the RNA regulatory level.
Collapse
Affiliation(s)
- Yaxue Xie
- Department of Pediatrics, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Hongshuo Shi
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250021, Shandong, China
| | - Bo Han
- Department of Pediatrics, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China. .,Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|