1
|
Boen HM, Pype LL, Papadimitriou K, Altintas S, Teuwen LA, Anguille S, Saevels K, Verlinden A, Delrue L, Heggermont WA, Bosman M, Guns PJ, Heidbuchel H, Van De Heyning CM, Van Craenenbroeck EM, Franssen C. Dynamics of SERPINA3 in response to anthracycline treatment and cardiovascular dysfunction. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:27. [PMID: 40087712 PMCID: PMC11907982 DOI: 10.1186/s40959-025-00324-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND SERPINA3 recently emerged as potential prognostic biomarker in heart failure. In a population of cancer survivors with cancer therapy-related cardiac dysfunction (CTRCD) circulating SERPINA3 was elevated compared to age-matched controls. We aimed to assess the longitudinal dynamics of circulating SERPINA3 levels in patients with cancer treated with anthracycline chemotherapy (AnC) and its relation to CTRCD. METHODS In this single centre cohort study, 55 patients with cancer scheduled for AnC were prospectively enrolled. Cardiac evaluation (echocardiography, high-sensitive cardiac troponin I and NT-proBNP) was performed and SERPINA3 levels in plasma were assessed at 4 timepoints: before chemotherapy, directly after the end of chemotherapy, three months and twelve months after the end of chemotherapy. RESULTS Forty-two out of 55 patients (76.4%) developed CTRCD within 1 year after end of treatment. CTRCD was mild in 32 and moderate in 10 patients, defined as a change in cardiac biomarkers or GLS and LVEF decline < 50% respectively. Overall, median SERPINA3 levels decreased from baseline to three months after AnC (215.7 [62.0-984.0] to 176.9 [94.7-678.0] µg/ml, p = 0.031). This decrease was most prominent in patients without CTRCD (30.8% decrease, p = 0.007), followed by mild CTRCD (9.0% decrease, p = 0.022), while patients with moderate CTRCD did not show a reduction in SERPINA3 (5.1% increase, p = 0.987). SERPINA3 values at three months after AnC were positively correlated with NT-proBNP (r = 0.47, p = 0.002). Several malignancy, treatment and patient characteristics were associated with higher SERPINA3 values. CONCLUSION Circulating SERPINA3 levels show dynamic changes in a population of patients with cancer, with an overall decrease following AnC. However, in patients that developed moderate CTRCD, SERPINA3 levels remained elevated. The potential of SERPINA3 dynamics as a biomarker for CTRCD, deserves validation in larger cohorts.
Collapse
Affiliation(s)
- Hanne M Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.
| | - Lobke L Pype
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | | | - Sevilay Altintas
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Sébastien Anguille
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Kirsten Saevels
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Anke Verlinden
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Leen Delrue
- Department of Cardiology, Olv Hospital Aalst, Aalst, Belgium
| | | | - Matthias Bosman
- Research Group Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Pieter-Jan Guns
- Research Group Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Caroline M Van De Heyning
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
2
|
Jardanowska-Kotuniak M, Dramiński M, Własnowolski M, Łapiński M, Sengupta K, Agarwal A, Filip A, Ghosh N, Pancaldi V, Grynberg M, Saha I, Plewczynski D, Dąbrowski MJ. Unveiling epigenetic regulatory elements associated with breast cancer development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623187. [PMID: 39605637 PMCID: PMC11601335 DOI: 10.1101/2024.11.12.623187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Breast cancer is the most common cancer in women and the 2nd most common cancer worldwide, yearly impacting over 2 million females and causing 650 thousand deaths. It has been widely studied, but its epigenetic variation is not entirely unveiled. We aimed to identify epigenetic mechanisms impacting the expression of breast cancer related genes to detect new potential biomarkers and therapeutic targets. We considered The Cancer Genome Atlas database with over 800 samples and several omics datasets such as mRNA, miRNA, DNA methylation, which we used to select 2701 features that were statistically significant to differ between cancer and control samples using the Monte Carlo Feature Selection and Interdependency Discovery algorithm, from an initial total of 417,486. Their biological impact on cancerogenesis was confirmed using: statistical analysis, natural language processing, linear and machine learning models as well as: transcription factors identification, drugs and 3D chromatin structure analyses. Classification of cancer vs control samples on the selected features returned high classification weighted Accuracy from 0.91 to 0.98 depending on feature-type: mRNA, miRNA, DNA methylation, and classification algorithm. In general, cancer samples showed lower expression of differentially expressed genes and increased β-values of differentially methylated sites. We identified mRNAs whose expression is well explained by miRNA expression and differentially methylated sites β-values. We recognized differentially methylated sites possibly affecting NRF1 and MXI1 transcription factors binding, causing a disturbance in NKAPL and PITX1 expression, respectively. Our 3D models showed more loosely packed chromatin in cancer. This study successfully points out numerous possible regulatory dependencies.
Collapse
Affiliation(s)
- Marta Jardanowska-Kotuniak
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dramiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Własnowolski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Marcin Łapiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Kaustav Sengupta
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Abhishek Agarwal
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Adam Filip
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Nimisha Ghosh
- Department of Computer Science and Information Technology, Institute of Technical Education and Research, Siksha O Anusandhan University, Bhubaneswar, Odisha, 751030, India
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Indrajit Saha
- Department of Computer Science and Engineering, National Institute of Technical Teachers’ Training and Research, Kolkata 700106, India
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Michał J. Dąbrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
3
|
Yu L, Cai S, Guo X. m6A RNA methylation modification is involved in the disease course of heart failure. Biotechnol Genet Eng Rev 2024; 40:961-975. [PMID: 36943073 DOI: 10.1080/02648725.2023.2191086] [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: 02/10/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023]
Abstract
We explored N6-methyladenosine (m6A) RNA methylation as one of the gene regulatory mechanisms in heart failure (HF) biology. Understanding the different physiological mechanisms will facilitate the prevention and individualized treatment of HF. The Gene Expression Omnibus (GEO) database served as the source of the data. In GSE116250, differential analysis between ischemic cardiomyopathy (ICM), dilated cardiomyopathy (DCM) and controls yielded differentially expressed m6A regulators. Differential analysis between HF and controls in GSE131296 identifies m6A-modified genes and then performs enrichment analysis. Protein-protein interaction (PPI) network analysis was performed for the differentially expressed ICM- or DCM-associated genes in GSE116250 and GSE55296, respectively. Finally, the diagnostic genes for ICM and DCM were predicted using receiver operating characteristic (ROC) curve. YTHDC1, HNRNPC and HNRNPA2B1 were significantly downregulated in GSE116250 in DCM and ICM compared with controls. A total of 195 genes were identified in GSE131296 as subject to m6A alteration. These genes may play a role in HF through the MAPK signaling pathway and p53 signaling pathway. PPI network analysis identified CCL5, CXCR4 and CCL2 as key genes for ICM and IL-6 as a key gene for DCM. Through ROC curves, we identified m6A-modified APLP1, KLF2 as potential diagnostic genes for ICM, and m6A-modified FGF7, FREM1 and C14orf132 as potential diagnostic genes for DCM. Our findings support m6A modifying mechanisms in HF etiology that contribute to the treatment of HF. Thus, our data suggest that m6A methylation may be an interesting target for therapeutic intervention.
Collapse
Affiliation(s)
- Liyan Yu
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| | - Shuxia Cai
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| | - Xiuli Guo
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
4
|
Andrzejczyk K, Abou Kamar S, van Ommen AM, Canto ED, Petersen TB, Valstar G, Akkerhuis KM, Cramer MJ, Umans V, Rutten FH, Teske A, Boersma E, Menken R, van Dalen BM, Hofstra L, Verhaar M, Brugts J, Asselbergs F, den Ruijter H, Kardys I. Identifying plasma proteomic signatures from health to heart failure, across the ejection fraction spectrum. Sci Rep 2024; 14:14871. [PMID: 38937570 PMCID: PMC11211454 DOI: 10.1038/s41598-024-65667-0] [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: 03/21/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Circulating proteins may provide insights into the varying biological mechanisms involved in heart failure (HF) with preserved ejection fraction (HFpEF) and reduced ejection fraction (HFrEF). We aimed to identify specific proteomic patterns for HF, by comparing proteomic profiles across the ejection fraction spectrum. We investigated 4210 circulating proteins in 739 patients with normal (Stage A/Healthy) or elevated (Stage B) filling pressures, HFpEF, or ischemic HFrEF (iHFrEF). We found 2122 differentially expressed proteins between iHFrEF-Stage A/Healthy, 1462 between iHFrEF-HFpEF and 52 between HFpEF-Stage A/Healthy. Of these 52 proteins, 50 were also found in iHFrEF vs. Stage A/Healthy, leaving SLITRK6 and NELL2 expressed in lower levels only in HFpEF. Moreover, 108 proteins, linked to regulation of cell fate commitment, differed only between iHFrEF-HFpEF. Proteomics across the HF spectrum reveals overlap in differentially expressed proteins compared to stage A/Healthy. Multiple proteins are unique for distinguishing iHFrEF from HFpEF, supporting the capacity of proteomics to discern between these conditions.
Collapse
Affiliation(s)
- Karolina Andrzejczyk
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sabrina Abou Kamar
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Anne-Mar van Ommen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elisa Dal Canto
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Teun B Petersen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gideon Valstar
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maarten Jan Cramer
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands
| | - Frans H Rutten
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arco Teske
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Roxana Menken
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Bas M van Dalen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Leonard Hofstra
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Marianne Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jasper Brugts
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Folkert Asselbergs
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
5
|
Chen X, Zhong X, Luo D, Lei Y, Huang R. Plasma SMOC2 Predicts Prognosis in Patients with Heart Failure: A Prospective Cohort. Int J Gen Med 2024; 17:1651-1664. [PMID: 38706743 PMCID: PMC11069073 DOI: 10.2147/ijgm.s445457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
Background Heart failure (HF) is a chronic disease with a poor prognosis, making it extremely important to assess the prognosis of patients with HF for accurate treatment. Secreted modular calcium-binding protein 2 (SMOC2) is a cysteine-rich acidic secreted protein that plays a pathophysiological role in many diseases, including regulation of vascular growth factor activity. It has previously been found that SMOC2 plays an essential role in cardiac fibrosis in our previous preclinical study, but whether it can be used as a clinical marker in heart failure patients remains unclear. The purpose of this research was to evaluate the correlation between plasma levels of SMOC2 and the prognosis for individuals with HF. Methods HF patients diagnosed with ischemic cardiomyopathy were enrolled from January to December 2021. Baseline plasma levels of SMOC2 were measured after demographic and clinical features were collected. Linear and nonlinear multivariate Cox regression models were used to determine the association between plasma SMOC2 and patient outcomes during follow-up. All analysis was performed using SPSS, EmpowerStats, and R software. Results The study included 188 patients, and the average follow-up time was 489.5±88.3 days. The plasma SMOC2 concentrations were positively correlated with N-terminal pro-B-type Natriuretic Peptide (NT-proBNP), left ventricular end-diastolic diameter (LVEDd), and length of hospital stay and were negatively correlated with left ventricular ejection fraction (LVEF) at baseline. A total of 53 patients (28.2%) were rehospitalized due to cardiac deterioration, 14 (7.4%) died, and 37 (19.7%) developed malignant arrhythmias. A fully adjusted multivariate COX regression model showed that SMOC2 is associated with readmission (HR = 1.02, 95% CI:1.012-1.655). A significant increase in rehospitalization risk was observed in group Q2 (HR =1.064, 95% CI: 1.037, 3.662, p=0.005) and group Q3 (HR =1.085, 95% CI:1.086, 3.792, p=0.009) in comparison with group Q1. The p for trend also shows a linear correlation across the three models (P < 0.001). SMOC2 was associated with the severity of HF in patients, but not with all-cause deaths and arrhythmias during follow-up. Conclusion Plasma SMOC2 is associated with the severity of HF and readmission rate, and is a good predictor of the risk of readmission in patients.
Collapse
Affiliation(s)
- Xin Chen
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei Province, People’s Republic of China
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshii, Hubei Province, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bio applications, Enshii, Hubei Province, People’s Republic of China
| | - Xing Zhong
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei Province, People’s Republic of China
- Department of Medicine, Hubei Minzu University, Enshi, Hubei Province, People’s Republic of China
| | - Dan Luo
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei Province, People’s Republic of China
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshii, Hubei Province, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bio applications, Enshii, Hubei Province, People’s Republic of China
| | - Yuhua Lei
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei Province, People’s Republic of China
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshii, Hubei Province, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bio applications, Enshii, Hubei Province, People’s Republic of China
| | - Rui Huang
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei Province, People’s Republic of China
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshii, Hubei Province, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bio applications, Enshii, Hubei Province, People’s Republic of China
| |
Collapse
|
6
|
You H, Dong M. Prediction of diagnostic gene biomarkers for hypertrophic cardiomyopathy by integrated machine learning. J Int Med Res 2023; 51:3000605231213781. [PMID: 38006610 PMCID: PMC10683566 DOI: 10.1177/03000605231213781] [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: 05/27/2023] [Accepted: 10/26/2023] [Indexed: 11/27/2023] Open
Abstract
OBJECTIVES Hypertrophic cardiomyopathy (HCM), a leading cause of heart failure and sudden death, requires early diagnosis and treatment. This study investigated the underlying pathogenesis and explored potential diagnostic gene biomarkers for HCM. METHODS Transcriptional profiles of myocardial tissues from patients with HCM (dataset GSE36961) were downloaded from the Gene Expression Omnibus database and subjected to bioinformatics analyses, including differentially expressed gene (DEG) identification, enrichment analyses, and protein-protein interaction (PPI) network analysis. Least absolute shrinkage and selection operator (LASSO) regression and support vector machine recursive feature elimination were performed to identify candidate diagnostic gene biomarkers. mRNA expression levels of candidate biomarkers were tested in an external dataset (GSE141910); area under the receiver operating characteristic curve (AUC) values were obtained to validate diagnostic efficacy. RESULTS Overall, 156 DEGs (109 downregulated, 47 upregulated) were identified. Enrichment and PPI network analyses indicated that the DEGs were involved in biological functions and molecular pathways including inflammatory response, platelet activity, complement and coagulation cascades, extracellular matrix organization, phagosome, apoptosis, and VEGFA-VEGFR2 signaling. RASD1, CDC42EP4, MYH6, and FCN3 were identified as diagnostic biomarkers for HCM. CONCLUSIONS RASD1, CDC42EP4, MYH6, and FCN3 might be diagnostic gene biomarkers for HCM and can provide insights concerning HCM pathogenesis.
Collapse
Affiliation(s)
- Hongjun You
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Mengya Dong
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Ren Y, Wu Y, He W, Tian Y, Zhao X. SMOC2 plays a role in heart failure via regulating TGF-β1/Smad3 pathway-mediated autophagy. Open Med (Wars) 2023; 18:20230752. [PMID: 37465345 PMCID: PMC10350896 DOI: 10.1515/med-2023-0752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
Heart failure (HF) is a major global cause of morbidity and mortality. This study aimed to elucidate the role of secreted protein acidic and rich in cysteine-related modular calcium-binding protein 2 (SMOC2) in HF development and its underlying mechanism. Using a rat HF model, SMOC2 expression was examined and then knocked down via transfection to assess its impact on cardiac function and damage. The study also evaluated the effects of SMOC2 knockdown on autophagy-related molecules and the transforming growth factor beta 1 (TGF-β1)/SMAD family member 3 (Smad3) signaling pathway. Intraperitoneal injection of the TGF-β agonist (SRI-011381) into the HF rat model was performed to explore the SMOC2-TGF-β1/Smad3 pathway relationship. SMOC2 expression was elevated in HF rats, while its downregulation improved cardiac function and damage. SMOC2 knockdown reversed alterations in the LC3-II/I ratio, Beclin-1, and p62 levels in HF rats. Through transmission electron microscope, we observed that SMOC2 knockdown restored autophagosome levels. Furthermore, SMOC2 downregulation inhibited the TGF-β1/Smad3 signaling pathway, which was counteracted by SRI-011381. In conclusion, SMOC2 knockdown inhibits HF development by modulating TGF-β1/Smad3 signaling-mediated autophagy, suggesting its potential as a therapeutic target for HF.
Collapse
Affiliation(s)
- Yu Ren
- Scientific Research Department, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
| | - Yun Wu
- Cardiology Department, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
- Clinical Medical Research Center in Cardiovascular Diseases, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
| | - Wenshuai He
- Cardiology Department, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
- Clinical Medical Research Center in Cardiovascular Diseases, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
| | - Yingjie Tian
- Cardiology Department, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
- Clinical Medical Research Center in Cardiovascular Diseases, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
| | - Xingsheng Zhao
- Cardiology Department, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
- Clinical Medical Research Center in Cardiovascular Diseases, Inner Mongolia People’s Hospital, Inner Mongolia Autonomous Region, Hohhot, 010017, China
| |
Collapse
|
8
|
Zhang M, Wang X, Chen W, Liu W, Xin J, Yang D, Zhang Z, Zheng X. Integrated bioinformatics analysis for identifying key genes and pathways in female and male patients with dilated cardiomyopathy. Sci Rep 2023; 13:8977. [PMID: 37268658 DOI: 10.1038/s41598-023-36117-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of heart failure, and males are more likely to suffer from DCM than females. This research aimed at exploring possible DCM-associated genes and their latent regulatory effects in female and male patients. WGCNA analysis found that in the yellow module, 341 and 367 key DEGs were identified in females and males, respectively. A total of 22 hub genes in females and 17 hub genes in males were identified from the PPI networks of the key DEGs based on Metascape database. And twelve and eight potential TFs of the key DEGs were also identified in females and males, respectively. Eight miRNAs of 15 key DEGs were screened in both females and males, which may be differentially expressed in females and males. Dual-luciferase reporter assay demonstrated that miR-21-5P could directly target the key gene MATN2. Furthermore, Sex differences in KEGG pathways were identified. Both KOBAS and GSEA analysis identified 19 significantly enriched pathways related to immune response in both females and males, and the TGF-β signaling pathway was exclusively identified in males. Network pharmacology analysis revealed that seven key DEGs were potential targets for the treatment of DCM, of which the OLR1 gene was only identified in males, the expression levels of the seven genes were verified by RT-PCR. The above results could offer a novel understanding of sex differences in key genes and pathways in DCM progression.
Collapse
Affiliation(s)
- Min Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xinzhou Wang
- The Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Wenbo Chen
- School of Medicine, Henan Polytechnic University, Jiaozuo, 454000, Henan, China
| | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, 450002, China
| | - Jile Xin
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Debao Yang
- The Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Zhongyuan Zhang
- The Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
9
|
Zhang L, Lin Y, Wang K, Han L, Zhang X, Gao X, Li Z, Zhang H, Zhou J, Yu H, Fu X. Multiple-model machine learning identifies potential functional genes in dilated cardiomyopathy. Front Cardiovasc Med 2023; 9:1044443. [PMID: 36712235 PMCID: PMC9874116 DOI: 10.3389/fcvm.2022.1044443] [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: 09/14/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Machine learning (ML) has gained intensive popularity in various fields, such as disease diagnosis in healthcare. However, it has limitation for single algorithm to explore the diagnosing value of dilated cardiomyopathy (DCM). We aim to develop a novel overall normalized sum weight of multiple-model MLs to assess the diagnosing value in DCM. Methods Gene expression data were selected from previously published databases (six sets of eligible microarrays, 386 samples) with eligible criteria. Two sets of microarrays were used as training; the others were studied in the testing sets (ratio 5:1). Totally, we identified 20 differently expressed genes (DEGs) between DCM and control individuals (7 upregulated and 13 down-regulated). Results We developed six classification ML methods to identify potential candidate genes based on their overall weights. Three genes, serine proteinase inhibitor A3 (SERPINA3), frizzled-related proteins (FRPs) 3 (FRZB), and ficolin 3 (FCN3) were finally identified as the receiver operating characteristic (ROC). Interestingly, we found all three genes correlated considerably with plasma cells. Importantly, not only in training sets but also testing sets, the areas under the curve (AUCs) for SERPINA3, FRZB, and FCN3 were greater than 0.88. The ROC of SERPINA3 was significantly high (0.940 in training and 0.918 in testing sets), indicating it is a potentially functional gene in DCM. Especially, the plasma levels in DCM patients of SERPINA3, FCN, and FRZB were significant compared with healthy control. Discussion SERPINA3, FRZB, and FCN3 might be potential diagnosis targets for DCM, Further verification work could be implemented.
Collapse
Affiliation(s)
- Lin Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yexiang Lin
- Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Kaiyue Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lifeng Han
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zheng Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | - Jiashun Zhou
- Tianjin Jinghai District Hospital, Tianjin, China
| | - Heshui Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,*Correspondence: Heshui Yu,
| | - Xuebin Fu
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States,Xuebin Fu,
| |
Collapse
|
10
|
Wang J, Xie S, Cheng Y, Li X, Chen J, Zhu M. Identification of potential biomarkers of inflammation-related genes for ischemic cardiomyopathy. Front Cardiovasc Med 2022; 9:972274. [PMID: 36082132 PMCID: PMC9445158 DOI: 10.3389/fcvm.2022.972274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveInflammation plays an important role in the pathophysiology of ischemic cardiomyopathy (ICM). We aimed to identify potential biomarkers of inflammation-related genes for ICM and build a model based on the potential biomarkers for the diagnosis of ICM.Materials and methodsThe microarray datasets and RNA-Sequencing datasets of human ICM were downloaded from the Gene Expression Omnibus database. We integrated 8 microarray datasets via the SVA package to screen the differentially expressed genes (DEGs) between ICM and non-failing control samples, then the differentially expressed inflammation-related genes (DEIRGs) were identified. The least absolute shrinkage and selection operator, support vector machine recursive feature elimination, and random forest were utilized to screen the potential diagnostic biomarkers from the DEIRGs. The potential biomarkers were validated in the RNA-Sequencing datasets and the functional experiment of the ICM rat, respectively. A nomogram was established based on the potential biomarkers and evaluated via the area under the receiver operating characteristic curve (AUC), calibration curve, decision curve analysis (DCA), and Clinical impact curve (CIC).Results64 DEGs and 19 DEIRGs were identified, respectively. 5 potential biomarkers (SERPINA3, FCN3, PTN, CD163, and SCUBE2) were ultimately selected. The validation results showed that each of these five potential biomarkers showed good discriminant power for ICM, and their expression trends were consistent with the bioinformatics results. The results of AUC, calibration curve, DCA, and CIC showed that the nomogram demonstrated good performance, calibration, and clinical utility.ConclusionSERPINA3, FCN3, PTN, CD163, and SCUBE2 were identified as potential biomarkers associated with the inflammatory response to ICM. The proposed nomogram could potentially provide clinicians with a helpful tool to the diagnosis and treatment of ICM from an inflammatory perspective.
Collapse
Affiliation(s)
- Jianru Wang
- Department of Cardiovascular, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Central Laboratory, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Shiyang Xie
- Department of Cardiovascular, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Central Laboratory, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanling Cheng
- Department of Cardiovascular, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaohui Li
- Department of Cardiovascular, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jian Chen
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Vascular Anomalies, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Jian Chen,
| | - Mingjun Zhu
- Department of Cardiovascular, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Mingjun Zhu,
| |
Collapse
|
11
|
Song H, Chen S, Zhang T, Huang X, Zhang Q, Li C, Chen C, Chen S, Liu D, Wang J, Tu Y, Wu Y, Liu Y. Integrated Strategies of Diverse Feature Selection Methods Identify Aging-Based Reliable Gene Signatures for Ischemic Cardiomyopathy. Front Mol Biosci 2022; 9:805235. [PMID: 35300115 PMCID: PMC8921505 DOI: 10.3389/fmolb.2022.805235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Objective: Ischemic cardiomyopathy (ICM) is a major cardiovascular state associated with prominently increased morbidity and mortality. Our purpose was to detect reliable gene signatures for ICM through integrated feature selection strategies.Methods: Transcriptome profiles of ICM were curated from the GEO project. Classification models, including least absolute shrinkage and selection operator (LASSO), support vector machine (SVM), and random forest, were adopted for identifying candidate ICM-specific genes for ICM. Immune cell infiltrates were estimated using the CIBERSORT method. Expressions of candidate genes were verified in ICM and healthy myocardial tissues via Western blotting. JC-1 staining, flow cytometry, and TUNEL staining were presented in hypoxia/reoxygenation (H/R)-stimulated H9C2 cells with TRMT5 deficiency.Results: Following the integration of three feature selection methods, we identified seven candidate ICM-specific genes including ASPN, TRMT5, LUM, FCN3, CNN1, PCNT, and HOPX. ROC curves confirmed the excellent diagnostic efficacy of this combination of previous candidate genes in ICM. Most of them presented prominent interactions with immune cell infiltrates. Their deregulations were confirmed in ICM than healthy myocardial tissues. TRMT5 expressions were remarkedly upregulated in H/R-stimulated H9C2 cells. TRMT5 deficiency enhanced mitochondrial membrane potential and reduced apoptosis in H/R-exposed H9C2 cells.Conclusion: Collectively, our findings identified reliable gene signatures through combination strategies of diverse feature selection methods, which facilitated the early detection of ICM and revealed the underlying mechanisms.
Collapse
Affiliation(s)
- Huafeng Song
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China
| | - Tingting Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofei Huang
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiyu Zhang
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Cuizhi Li
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunlin Chen
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoxian Chen
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, School of Medicine, Guangdong Provincial People’s Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
| | - Dehui Liu
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Yingfeng Tu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Yueheng Wu
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, School of Medicine, Guangdong Provincial People’s Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Youbin Liu
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| |
Collapse
|
12
|
Rui H, Zhao F, Yuhua L, Hong J. Suppression of SMOC2 alleviates myocardial fibrosis via the ILK/p38 pathway. Front Cardiovasc Med 2022; 9:951704. [PMID: 36935650 PMCID: PMC10017443 DOI: 10.3389/fcvm.2022.951704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/13/2022] [Indexed: 03/06/2023] Open
Abstract
Background Fibrosis of the myocardium is one of the main pathological changes of adverse cardiac remodeling, which is associated with unsatisfactory outcomes in patients with heart disease. Further investigations into the precise molecular mechanisms of cardiac fibrosis are urgently required to seek alternative therapeutic strategies for individuals suffering from heart failure. SMOC2 has been shown to be essential to exert key pathophysiological roles in various physiological processes in vivo, possibly contributing to the pathogenesis of fibrosis. A study investigating the relationship between SMOC2 and myocardial fibrosis has yet to be conducted. Methods Mice received a continuous ISO injection subcutaneously to induce cardiac fibrosis, and down-regulation of SMOC2 was achieved by adeno-associated virus-9 (AAV9)-mediated shRNA knockdown. Neonatal fibroblasts were separated and cultured in vitro with TGFβ to trigger fibrosis and infected with either sh-SMOC2 or sh-RNA as a control. The role and mechanisms of SMOC2 in myocardial fibrosis were further examined and analyzed. Results SMOC2 knockdown partially reversed cardiac functional impairment and cardiac fibrosis in vivo after 21 consecutive days of ISO injection. We further demonstrated that targeting SMOC2 expression effectively slowed down the trans-differentiation and collagen deposition of cardiac fibroblasts stimulated by TGFβ. Mechanistically, targeting SMOC2 expression inhibited the induction of ILK and p38 in vivo and in vitro, and ILK overexpression increased p38 phosphorylation activity and compromised the protective effects of sh-SMOC2-mediated cardiac fibrosis. Conclusion Therapeutic SMOC2 silencing alleviated cardiac fibrosis through inhibition of the ILK/p38 signaling, providing a preventative and control strategy for cardiac remodeling management in clinical practice.
Collapse
Affiliation(s)
- Huang Rui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Fang Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lei Yuhua
- Department of Cardiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi City, China
| | - Jiang Hong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Jiang Hong,
| |
Collapse
|
13
|
Wang T, Tian J, Jin Y. VCAM1 expression in the myocardium is associated with the risk of heart failure and immune cell infiltration in myocardium. Sci Rep 2021; 11:19488. [PMID: 34593936 PMCID: PMC8484263 DOI: 10.1038/s41598-021-98998-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023] Open
Abstract
Ischemic heart disease (IHD) and dilated cardiomyopathy (DCM) are the two most common etiologies of heart failure (HF). Both forms share common characteristics including ventricle dilation in the final stage. Immune mechanisms in HF are increasingly highlighted and have been implicated in the pathogeneses of IHD and DCM. A better understanding of adhesion molecule expression and correlated immune cell infiltration could enhance disease detection and improve therapeutic targets. This study was performed to explore the common mechanisms underlying IHD and DCM. After searching the Gene Expression Omnibus database, we selected the GSE42955, GSE76701, GSE5406, GSE133054 and GSE57338 datasets for different expressed gene (DEGs) selection and new cohort establishment. We use xcell to calculate immune infiltration degree, ssGSEA and GSEA to calculate the pathway and biological enrichment score, consensus cluster to identify the m6A modification pattern, and LASSO regression to make risk predicting model and use new combined cohort to validate the results. The screening stage revealed that vascular cell adhesion molecule 1 (VCAM1) play pivotal roles in regulating DEGs. Subsequent analyses revealed that VCAM1 was differentially expressed in the myocardium and involved in regulating immune cell infiltration. We also found that dysregulated VCAM1 expression was associated with a higher risk of HF by constructing a clinical risk-predicting model. Besides, we also find a connection among the m6A RNA modification ,expression of VCAM1 and immune regulation. Those connection can be linked by the Wnt pathway enrichment alternation. Collectively, our results suggest that VCAM-1 have the potential to be used as a biomarker or therapy target for HF and the m6A modification pattern is associated with the VCAM1 expression and immune regulation.
Collapse
Affiliation(s)
- Tongyu Wang
- The Fourth Affiliated Hospital of China Medical University, Yuanzhe Jin, No. 4 Chongshan East Road, Huanggu District, Shenyang, Liaoning Province, China
| | - Jiahu Tian
- The Fourth Affiliated Hospital of China Medical University, Yuanzhe Jin, No. 4 Chongshan East Road, Huanggu District, Shenyang, Liaoning Province, China
| | - Yuanzhe Jin
- The Fourth Affiliated Hospital of China Medical University, Yuanzhe Jin, No. 4 Chongshan East Road, Huanggu District, Shenyang, Liaoning Province, China.
| |
Collapse
|