1
|
Yang YL, Li XW, Chen HB, Tang QD, Li YH, Xu JY, Xie JJ. Single-cell transcriptomics reveals writers of RNA modification-mediated immune microenvironment and cardiac resident Macro-MYL2 macrophages in heart failure. BMC Cardiovasc Disord 2024; 24:432. [PMID: 39152369 PMCID: PMC11328403 DOI: 10.1186/s12872-024-04080-x] [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: 01/09/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Heart failure (HF), which is caused by cardiac overload and injury, is linked to significant mortality. Writers of RNA modification (WRMs) play a crucial role in the regulation of epigenetic processes involved in immune response and cardiovascular disease. However, the potential roles of these writers in the immunological milieu of HF remain unknown. METHODS We comprehensively characterized the expressions of 28 WRMs using datasets GSE145154 and GSE141910 to map the cardiac immunological microenvironment in HF patients. Based on the expression of WRMs, the immunological cells in the datasets were scored. RESULTS Single-cell transcriptomics analysis (GSE145154) revealed immunological dysregulation in HF as well as differential expression of WRMs in immunological cells from HF and non-HF (NHF) samples. WRM-scored immunological cells were positively correlated with the immunological response, and the high WRM score group exhibited elevated immunological cell infiltration. WRMs are involved in the differentiation of T cells and myeloid cells. WRM scores of T cell and myeloid cell subtypes were significantly reduced in the HF group compared to the NHF group. We identified a myogenesis-related resident macrophage population in the heart, Macro-MYL2, that was characterized by an increased expression of cardiomyocyte structural genes (MYL2, TNNI3, TNNC1, TCAP, and TNNT2) and was regulated by TRMT10C. Based on the WRM expression pattern, the transcriptomics data (GSE141910) identified two distinct clusters of HF samples, each with distinct functional enrichments and immunological characteristics. CONCLUSION Our study demonstrated a significant relationship between the WRMs and immunological microenvironment in HF, as well as a novel resident macrophage population, Macro-MYL2, characterized by myogenesis. These results provide a novel perspective on the underlying mechanisms and therapeutic targets for HF. Further experiments are required to validate the regulation of WRMs and Macro-MYL2 macrophage subtype in the cardiac immunological milieu.
Collapse
Affiliation(s)
- Yao-Lin Yang
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Xiao-Wei Li
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Hai-Bin Chen
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Qi-Dong Tang
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Yu-Hui Li
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Ji-Ying Xu
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China
| | - Jia-Jia Xie
- First Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospi-tal of Jinan University, NO. 466, Xingang Middle Road, Haizhu District, Guangzhou City, China.
| |
Collapse
|
2
|
Dai H, Liu Y, Zhu M, Tao S, Hu C, Luo P, Jiang A, Zhang G. Machine learning and experimental validation of novel biomarkers for hypertrophic cardiomyopathy and cancers. J Cell Mol Med 2024; 28:e70034. [PMID: 39160643 PMCID: PMC11333198 DOI: 10.1111/jcmm.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 08/21/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary cardiac disorder marked by anomalous thickening of the myocardium, representing a significant contributor to mortality. While the involvement of immune inflammation in the development of cardiac ailments is well-documented, its specific impact on HCM pathogenesis remains uncertain. Five distinct machine learning algorithms, namely LASSO, SVM, RF, Boruta and XGBoost, were utilized to discover new biomarkers associated with HCM. A unique nomogram was developed using two newly identified biomarkers and subsequently validated. Furthermore, samples of HCM and normal heart tissues were gathered from our institution to confirm the variance in expression levels and prognostic significance of GATM and MGST1. Five novel biomarkers (DARS2, GATM, MGST1, SDSL and ARG2) associated with HCM were identified. Subsequent validation revealed that GATM and MGST1 exhibited significant diagnostic utility for HCM in both the training and test cohorts, with all AUC values exceeding 0.8. Furthermore, a novel risk assessment model for HCM patients based on the expression levels of GATM and MGST1 demonstrated favourable performance in both the training (AUC = 0.88) and test cohorts (AUC = 0.9). Furthermore, our study revealed that GATM and MGST1 exhibited elevated expression levels in HCM tissues, demonstrating strong discriminatory ability between HCM and normal cardiac tissues (AUC of GATM = 0.79; MGST1 = 0.86). Our findings suggest that two specific cell types, monocytes and multipotent progenitors (MPP), may play crucial roles in the pathogenesis of HCM. Notably, GATM and MGST1 were found to be highly expressed in various tumours and showed significant prognostic implications. Functionally, GATM and MGST1 are likely involved in xenobiotic metabolism and epithelial mesenchymal transition in a wide range of cancer types. GATM and MGST1 have been identified as novel biomarkers implicated in the progression of both HCM and cancer. Additionally, monocytes and MPP may also play a role in facilitating the progression of HCM.
Collapse
Affiliation(s)
- Hualei Dai
- Cardiovascular CenterThe Affiliated Hospital of Yunnan University, Yunnan UniversityKunmingYunnanChina
- School of MedicineYunnan UniversityKunmingYunnanChina
| | - Ying Liu
- Department of GynecologyYunnan Cancer Hospital and The Third Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Meng Zhu
- Department of GeriatricsThe Affiliated Huaian Hospital of Xuzhou Medical University, Huaian Second People's HospitalHuaianJiangsuChina
| | - Siming Tao
- Cardiovascular CenterThe Affiliated Hospital of Yunnan University, Yunnan UniversityKunmingYunnanChina
| | - Chengcheng Hu
- Cardiovascular CenterThe Affiliated Hospital of Yunnan University, Yunnan UniversityKunmingYunnanChina
| | - Peng Luo
- Department of OncologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Aimin Jiang
- Department of UrologyChangzheng Hospital, Naval Medical UniversityShanghaiChina
| | - Guimin Zhang
- Cardiovascular CenterThe Affiliated Hospital of Yunnan University, Yunnan UniversityKunmingYunnanChina
- School of MedicineYunnan UniversityKunmingYunnanChina
| |
Collapse
|
3
|
Gu J, Zhao Y, Ben Y, Zhang S, Hua L, He S, Liu R, Chen X, Sheng H. A personalized mRNA signature for predicting hypertrophic cardiomyopathy applying machine learning methods. Sci Rep 2024; 14:17023. [PMID: 39043774 PMCID: PMC11266364 DOI: 10.1038/s41598-024-67201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) may lead to cardiac dysfunction and sudden death. This study was designed to develop a HCM signature applying bioinformatics and machine learning methods. Data of HCM and normal tissues were obtained from public databases to screen differentially expressed genes (DEGs) using the R software limma package. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed for enrichment analysis of HCM-associated DEGs. Hub genes for HCM were determined using weighted gene co-expression network analysis (WGCNA) together with two machine learning algorithms (SVM-RFE and LASSO). Finally, we introduced a zebrafish model to simulate changes in the hub genes in the HCM and to observe their effects on cardiac disease development. The mRNA expression data from a total of 106 HCM tissues and 39 normal samples were collected and we screened 157 DEGs. Enrichment analysis showed that immune pathways played an important role in the pathogenesis of HCM. Three hub genes (FCN3, MYH6 and RASD1) were identified using WGCNA, SVM-RFE, and LASSO analysis. In a zebrafish model, knockdown of MYH6 and RASD1 resulted in cardiac malformations with reduced ventricular capacity and heart rate, which validated the clinical significance of these genes in the diagnosis of HCM. Based on machine learning algorithms, our study created a signature with potential impact on cardiac function and cardiac quality index for HCM. The current findings had important implications for the early diagnosis and treatment of HCM.
Collapse
Affiliation(s)
- Jue Gu
- Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, 226000, Jiangsu Province, China
| | - Yamin Zhao
- Nantong Second People's Hospital, Nantong, China
| | - Yue Ben
- Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, 226000, Jiangsu Province, China
| | - Siming Zhang
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Liqi Hua
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Songnian He
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Ruizi Liu
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Xu Chen
- Medical School of Nantong University, Nantong University, Nantong, China.
| | - Hongzhuan Sheng
- Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, 226000, Jiangsu Province, China.
| |
Collapse
|
4
|
Zhou L, Zhang L, Lv Y, Qian J, Huang L, Qu C. YTHDC1 inhibits autophagy-dependent NF-κB signaling by stabilizing Beclin1 mRNA in macrophages. J Inflamm (Lond) 2024; 21:22. [PMID: 38877444 PMCID: PMC11179287 DOI: 10.1186/s12950-024-00393-y] [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: 10/31/2023] [Accepted: 05/15/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND YTHDC1, a key m(6)A nuclear reader, plays a crucial role in regulating mRNA splicing, export, and stability. However, the functional significance and regulatory mechanisms of YTHDC1 in inflammatory bowel disease (IBD) remain to be explored. METHODS We established a dextran sulfate sodium (DSS)-induced murine colitis model in vivo and LPS/IFN-γ-stimulated macrophage inflammation in vitro. The expression of YTHDC1 was determined. Colocalization of YTHDC1 and macrophages was assayed by immunofluorescence staining. LV-YTHDC1 or shYTHDC1 lentiviruses were applied for YTHDC1 overexpression or inhibition. For NF-κB inhibition, JSH-23 was utilized. The interaction of YTHDC1 and Beclin1 mRNA was determined by RIP, and the m6A modification of Beclin1 was confirmed by MeRIP. RESULTS In DSS-induced colitis and LPS/IFN-γ-treated RAW264.7 macrophages, we observed a significant downregulation of YTHDC1. Overexpression of YTHDC1 resulted in decreased levels of iNOS, CD86, and IL-6 mRNA, along with inhibited NF-κB activation in LPS/IFN-γ-treated RAW264.7 cells. Conversely, downregulation of YTHDC1 promoted iNOS expression and inhibited autophagy. Additionally, the effect of YTHDC1 knockdown on CD86 and IL-6 mRNA induced by LPS/IFN-γ was abolished by the NF-κB inhibitor JSH-23. Mechanistically, YTHDC1 interacted with Beclin1 mRNA, thereby stabilizing Beclin1 mRNA and enhancing Beclin1 expression and autophagy. These effects ultimately led to the inhibition of NF-κB signaling in LPS/IFN-γ-challenged macrophages. CONCLUSIONS YTHDC1 inhibited the macrophage-mediated inflammatory response by stabilizing Beclin1 mRNA, which may be a potential therapeutic target for the treatment of IBD.
Collapse
Affiliation(s)
- Li Zhou
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Ling Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Yan Lv
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Jiasheng Qian
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Long Huang
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China.
| | - Chenjiang Qu
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China.
| |
Collapse
|
5
|
Menezes Junior ADS, de França-e-Silva ALG, de Oliveira HL, de Lima KBA, Porto IDOP, Pedroso TMA, Silva DDME, Freitas AF. Genetic Mutations and Mitochondrial Redox Signaling as Modulating Factors in Hypertrophic Cardiomyopathy: A Scoping Review. Int J Mol Sci 2024; 25:5855. [PMID: 38892064 PMCID: PMC11173352 DOI: 10.3390/ijms25115855] [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: 04/22/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heart condition characterized by cellular and metabolic dysfunction, with mitochondrial dysfunction playing a crucial role. Although the direct relationship between genetic mutations and mitochondrial dysfunction remains unclear, targeting mitochondrial dysfunction presents promising opportunities for treatment, as there are currently no effective treatments available for HCM. This review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews guidelines. Searches were conducted in databases such as PubMed, Embase, and Scopus up to September 2023 using "MESH terms". Bibliographic references from pertinent articles were also included. Hypertrophic cardiomyopathy (HCM) is influenced by ionic homeostasis, cardiac tissue remodeling, metabolic balance, genetic mutations, reactive oxygen species regulation, and mitochondrial dysfunction. The latter is a common factor regardless of the cause and is linked to intracellular calcium handling, energetic and oxidative stress, and HCM-induced hypertrophy. Hypertrophic cardiomyopathy treatments focus on symptom management and complication prevention. Targeted therapeutic approaches, such as improving mitochondrial bioenergetics, are being explored. This includes coenzyme Q and elamipretide therapies and metabolic strategies like therapeutic ketosis. Understanding the biomolecular, genetic, and mitochondrial mechanisms underlying HCM is crucial for developing new therapeutic modalities.
Collapse
Affiliation(s)
- Antonio da Silva Menezes Junior
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Ana Luísa Guedes de França-e-Silva
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Henrique Lima de Oliveira
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Khissya Beatryz Alves de Lima
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Iane de Oliveira Pires Porto
- Faculdade de Medicina, Universidade de Rio Verde (UniRV), Campus Aparecida, Aparecida de Goiânia 74345-030, Brazil; (I.d.O.P.P.); (T.M.A.P.)
| | - Thays Millena Alves Pedroso
- Faculdade de Medicina, Universidade de Rio Verde (UniRV), Campus Aparecida, Aparecida de Goiânia 74345-030, Brazil; (I.d.O.P.P.); (T.M.A.P.)
| | - Daniela de Melo e Silva
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Aguinaldo F. Freitas
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| |
Collapse
|
6
|
Xu X, Shen L, Qu Y, Li D, Zhao X, Wei H, Yue S. Experimental validation and comprehensive analysis of m6A methylation regulators in intervertebral disc degeneration subpopulation classification. Sci Rep 2024; 14:8417. [PMID: 38600232 PMCID: PMC11006851 DOI: 10.1038/s41598-024-58888-w] [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: 11/06/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is one of the most prevalent causes of chronic low back pain. The role of m6A methylation modification in disc degeneration (IVDD) remains unclear. We investigated immune-related m6A methylation regulators as IVDD biomarkers through comprehensive analysis and experimental validation of m6A methylation regulators in disc degeneration. The training dataset was downloaded from the GEO database and analysed for differentially expressed m6A methylation regulators and immunological features, the differentially regulators were subsequently validated by a rat IVDD model and RT-qPCR. Further screening of key m6A methylation regulators based on machine learning and LASSO regression analysis. Thereafter, a predictive model based on key m6A methylation regulators was constructed for training sets, which was validated by validation set. IVDD patients were then clustered based on the expression of key m6A regulators, and the expression of key m6A regulators and immune infiltrates between clusters was investigated to determine immune markers in IVDD. Finally, we investigated the potential role of the immune marker in IVDD through enrichment analysis, protein-to-protein network analysis, and molecular prediction. By analysising of the training set, we revealed significant differences in gene expression of five methylation regulators including RBM15, YTHDC1, YTHDF3, HNRNPA2B1 and ALKBH5, while finding characteristic immune infiltration of differentially expressed genes, the result was validated by PCR. We then screen the differential m6A regulators in the training set and identified RBM15 and YTHDC1 as key m6A regulators. We then used RBM15 and YTHDC1 to construct a predictive model for IVDD and successfully validated it in the training set. Next, we clustered IVDD patients based on the expression of RBM15 and YTHDC1 and explored the immune infiltration characteristics between clusters as well as the expression of RBM15 and YTHDC1 in the clusters. YTHDC1 was finally identified as an immune biomarker for IVDD. We finally found that YTHDC1 may influence the immune microenvironment of IVDD through ABL1 and TXK. In summary, our results suggest that YTHDC1 is a potential biomarker for the development of IVDD and may provide new insights for the precise prevention and treatment of IVDD.
Collapse
Affiliation(s)
- Xiaoqian Xu
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Lianwei Shen
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Yujuan Qu
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Danyang Li
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaojing Zhao
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Wei
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China
| | - Shouwei Yue
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
7
|
Zhang Y, Zhao J, Jin Q, Zhuang L. Transcriptomic Analyses and Experimental Validation Identified Immune-Related lncRNA-mRNA Pair MIR210HG- BPIFC Regulating the Progression of Hypertrophic Cardiomyopathy. Int J Mol Sci 2024; 25:2816. [PMID: 38474063 DOI: 10.3390/ijms25052816] [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: 01/24/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a disease in which the myocardium of the heart becomes asymmetrically thickened, malformed, disordered, and loses its normal structure and function. Recent studies have demonstrated the significant involvement of inflammatory responses in HCM. However, the precise role of immune-related long non-coding RNAs (lncRNAs) in the pathogenesis of HCM remains unclear. In this study, we performed a comprehensive analysis of immune-related lncRNAs in HCM. First, transcriptomic RNA-Seq data from both HCM patients and healthy individuals (GSE180313) were reanalyzed thoroughly. Key HCM-related modules were identified using weighted gene co-expression network analysis (WGCNA). A screening for immune-related lncRNAs was conducted within the key modules using immune-related mRNA co-expression analysis. Based on lncRNA-mRNA pairs that exhibit shared regulatory microRNAs (miRNAs), we constructed a competing endogenous RNA (ceRNA) network, comprising 9 lncRNAs and 17 mRNAs that were significantly correlated. Among the 26 lncRNA-mRNA pairs, only the MIR210HG-BPIFC pair was verified by another HCM dataset (GSE130036) and the isoprenaline (ISO)-induced HCM cell model. Furthermore, knockdown of MIR210HG increased the regulatory miRNAs and decreased the mRNA expression of BPIFC correspondingly in AC16 cells. Additionally, the analysis of immune cell infiltration indicated that the MIR210HG-BPIFC pair was potentially involved in the infiltration of naïve CD4+ T cells and CD8+ T cells. Together, our findings indicate that the decreased expression of the lncRNA-mRNA pair MIR210HG-BPIFC was significantly correlated with the pathogenesis of the disease and may be involved in the immune cell infiltration in the mechanism of HCM.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jiuxiao Zhao
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiao Jin
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lenan Zhuang
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
8
|
Zhao L, Wang Y, Mu P, Zhang X, Qi R, Zhang Y, Zhang H, Zhu X, Dong Z, Dong Y. IGFBP3 induces PD-L1 expression to promote glioblastoma immune evasion. Cancer Cell Int 2024; 24:60. [PMID: 38326861 PMCID: PMC10851611 DOI: 10.1186/s12935-024-03234-3] [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: 11/18/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) characterized by immune escape is the most malignant primary brain tumors, which has strong immunosuppressive effect. Programmed death ligand-1 (PD-L1) is a recognized immunosuppressive member on the surface of tumor cells, and plays a crucial role in immune evasion of tumors. Actually, little is known about the regulation of PD-L1 expression in GBM. Insulin-like growth factor binding protein 3 (IGFBP3) is upregulated in GBM and is related to poor patient prognosis. However, it remains unclear whether IGFBP3 plays a role in the regulation of PD-L1 expression in GBM. METHODS The role of IGFBP3 in the glioma immune microenvironment was investigated using the CIBERSORT algorithm. The correlation between IGFBP3 and PD-L1 expression was analyzed using TCGA and CGGA databases. QRT-PCR, immunoblotting and RNA-seq were used to examine the regulatory effect of IGFBP3 on PD-L1 expression. Co-culture assay, cell counting kit (CCK-8), qRT-PCR, ELISA and flow cytometry were performed to explore the function of IGFBP3 in inducing immunosuppression. The biological role of IGFBP3 was verified using immunohistochemical, immunofluorescence and mice orthotopic tumor model. RESULTS In this study, we analyzed immune cells infiltration in gliomas and found that IGFBP3 may be associated with an immunosuppressive microenvironment. Then, by analyzing TCGA and CGGA databases, our results showed that IGFBP3 and PD-L1 expression were positively correlated in GBM patients, but not in LGG patients. In vitro experiments conducted on different GBM cell lines revealed that the overexpression of IGFBP3 led to an increase in PD-L1 expression, which was reversible upon knockdown IGFBP3. Mechanistically, IGFBP3 activated the JAK2/STAT3 signaling pathway, leading to an increase in PD-L1 expression. Additionally, co-culture experiments results showed IGFBP3 overexpression induced upregulation of PD-L1 expression promoted apoptosis in Jurkat cells, and this effect was blocked by IGFBP3 antibody and PDL-1 inhibitors. Importantly, in vivo experiments targeting IGFBP3 suppressed tumor growth and significantly prolonged the survival of mice. CONCLUSIONS This research demonstrated IGFBP3 is a novel regulator for PD-L1 expression in GBM, and identified a new mechanism by which IGFBP3 regulates immune evasion through PD-L1, suggesting that IGFBP3 may be a potential novel target for GBM therapy.
Collapse
Affiliation(s)
- Leilei Zhao
- Department of Immunology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China
| | - Yudi Wang
- Department of Immunology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China
| | - Peizheng Mu
- School of Computer and Normal Engineering, Yantai University, Qingquan Road 30, Yantai, 264005, Shandong, China
| | - Xuehua Zhang
- Department of Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Ruomei Qi
- Department of Immunology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Xiao Zhu
- School of Computer and Normal Engineering, Yantai University, Qingquan Road 30, Yantai, 264005, Shandong, China.
| | - Zhouyan Dong
- Department of Pathogenic Biology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China.
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, Shandong, China.
| |
Collapse
|
9
|
Ramos-Medina MJ, Echeverría-Garcés G, Kyriakidis NC, León Cáceres Á, Ortiz-Prado E, Bautista J, Pérez-Meza ÁA, Abad-Sojos A, Nieto-Jaramillo K, Espinoza-Ferrao S, Ocaña-Paredes B, López-Cortés A. CardiOmics signatures reveal therapeutically actionable targets and drugs for cardiovascular diseases. Heliyon 2024; 10:e23682. [PMID: 38187312 PMCID: PMC10770621 DOI: 10.1016/j.heliyon.2023.e23682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/27/2023] [Accepted: 12/09/2023] [Indexed: 01/09/2024] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, with heart failure being a complex condition that affects millions of individuals. Single-nucleus RNA sequencing has recently emerged as a powerful tool for unraveling the molecular mechanisms behind cardiovascular diseases. This cutting-edge technology enables the identification of molecular signatures, intracellular networks, and spatial relationships among cardiac cells, including cardiomyocytes, mast cells, lymphocytes, macrophages, lymphatic endothelial cells, endocardial cells, endothelial cells, epicardial cells, adipocytes, fibroblasts, neuronal cells, pericytes, and vascular smooth muscle cells. Despite these advancements, the discovery of essential therapeutic targets and drugs for precision cardiology remains a challenge. To bridge this gap, we conducted comprehensive in silico analyses of single-nucleus RNA sequencing data, functional enrichment, protein interactome network, and identification of the shortest pathways to physiological phenotypes. This integrated multi-omics analysis generated CardiOmics signatures, which allowed us to pinpoint three therapeutically actionable targets (ADRA1A1, PPARG, and ROCK2) and 15 effective drugs, including adrenergic receptor agonists, adrenergic receptor antagonists, norepinephrine precursors, PPAR receptor agonists, and Rho-associated kinase inhibitors, involved in late-stage cardiovascular disease clinical trials.
Collapse
Affiliation(s)
- María José Ramos-Medina
- German Cancer Research Center (DKFZ), Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | - Nikolaos C. Kyriakidis
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Ángela León Cáceres
- Heidelberg Institute of Global Health, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
- Instituto de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Esteban Ortiz-Prado
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Jhommara Bautista
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Álvaro A. Pérez-Meza
- Escuela de Medicina, Colegio de Ciencias de La Salud COCSA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | | | - Karol Nieto-Jaramillo
- School of Biological Sciences and Engineering, Yachay Tech University, Urcuqui, Ecuador
| | | | - Belén Ocaña-Paredes
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
10
|
Song B, Xie B, Liu M, Li H, Shi D, Zhao F. Bibliometric and visual analysis of RAN methylation in cardiovascular disease. Front Cardiovasc Med 2023; 10:1110718. [PMID: 37063953 PMCID: PMC10098125 DOI: 10.3389/fcvm.2023.1110718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundRNA methylation is associated with cardiovascular disease (CVD) occurrence and development. The purpose of this study is to visually analyze the results and research trends of global RNA methylation in CVD.MethodsArticles and reviews on RNA methylation in CVD published before 6 November 2022 were searched in the Web of Science Core Collection. Visual and statistical analysis was performed using CiteSpace 1.6.R4 advanced and VOSviewer 1.6.18.ResultsThere were 847 papers from 1,188 institutions and 63 countries/regions. Over approximately 30 years, there was a gradual increase in publications and citations on RNA methylation in CVD. America and China had the highest output (284 and 259 papers, respectively). Nine of the top 20 institutions that published articles were from China, among which Fudan University represented the most. The International Journal of Molecular Sciences was the journal with the most studies. Nature was the most co-cited journal. The most influential writers were Zhang and Wang from China and Mathiyalagan from the United States. After 2015, the primary keywords were cardiac development, heart, promoter methylation, RNA methylation, and N6-methyladenosine. Nuclear RNA, m6A methylation, inhibition, and myocardial infarction were the most common burst keywords from 2020 to the present.ConclusionsA bibliometric analysis reveals research hotspots and trends of RNA methylation in CVD. The regulatory mechanisms of RNA methylation related to CVD and the clinical application of their results, especially m6A methylation, are likely to be the focus of future research.
Collapse
Affiliation(s)
- Boce Song
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Beili Xie
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingwang Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haohao Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Fuhai Zhao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- Correspondence: Fuhai Zhao
| |
Collapse
|
11
|
Zhu Y, Li J, Yang H, Yang X, Zhang Y, Yu X, Li Y, Chen G, Yang Z. The potential role of m6A reader YTHDF1 as diagnostic biomarker and the signaling pathways in tumorigenesis and metastasis in pan-cancer. Cell Death Dis 2023; 9:34. [PMID: 36707507 PMCID: PMC9883452 DOI: 10.1038/s41420-023-01321-4] [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: 11/26/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/29/2023]
Abstract
m6A is an important RNA methylation in progression of various human cancers. As the m6A reader protein, YTHDF1 is reported to accelerate m6A-modified mRNAs translation in cytoplasm. It is highly expressed in various human cancers and contributes to the progression and metastasis of cancers. YTHDF1 was closely associated with poor prognosis and also used as a molecular marker for clinical diagnosis or therapy in human cancers. It has been reported to promote chemoresistance to Adriamycin, Cisplatin and Olaparib by increasing mRNA stability of its target molecule. Moreover, it contributes to CSC-like characteristic of tumor cells and inducing the antitumor immune microenvironment. Here, we reviewed the clinical diagnostic and prognostic values of YTHDF1, as well as the molecular mechanisms of YTHDF1 in progression and metastasis of human cancers.
Collapse
Affiliation(s)
- Yanan Zhu
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Jing Li
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Hang Yang
- grid.415444.40000 0004 1800 0367Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, 650106 Kunming, Yunnan China
| | - Xinyi Yang
- grid.413458.f0000 0000 9330 9891Guizhou Medical University, 550004 Guiyang, Guizhou China
| | - Ya Zhang
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Xinchao Yu
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Ying Li
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Gangxian Chen
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| | - Zuozhang Yang
- grid.452826.fBone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), 650118 Kunming, Yunnan China
| |
Collapse
|