1
|
Chen S, Long S, Liu Y, Wang S, Hu Q, Fu L, Luo D. Evaluation of a three-gene methylation model for correlating lymph node metastasis in postoperative early gastric cancer adjacent samples. Front Oncol 2024; 14:1432869. [PMID: 39484038 PMCID: PMC11524798 DOI: 10.3389/fonc.2024.1432869] [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: 05/14/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background Lymph node metastasis (LNM) has a profound impact on the treatment and prognosis of early gastric cancer (EGC), yet the existing evaluation methods lack accuracy. Recent research has underscored the role of precancerous lesions in tumor progression and metastasis. The objective of this study was to utilize the previously developed EGC LNM prediction model to further validate and extend the analysis in paired adjacent tissue samples. Methods We evaluated the model in a monocentric study using Methylight, a methylation-specific PCR technique, on postoperative fresh-frozen EGC samples (n = 129) and paired adjacent tissue samples (n = 129). Results The three-gene methylation model demonstrated remarkable efficacy in both EGC and adjacent tissues. The model demonstrated excellent performance, with areas under the curve (AUC) of 0.85 and 0.82, specificities of 85.1% and 80.5%, sensitivities of 83.3% and 73.8%, and accuracies of 84.5% and 78.3%, respectively. It is noteworthy that the model demonstrated superior performance compared to computed tomography (CT) imaging in the adjacent tissue group, with an area under the curve (AUC) of 0.86 compared to 0.64 (p < 0.001). Furthermore, the model demonstrated superior diagnostic capability in these adjacent tissues (AUC = 0.82) compared to traditional clinicopathological features, including ulceration (AUC = 0.65), invasional depth (AUC = 0.66), and lymphovascular invasion (AUC = 0.69). Additionally, it surpassed traditional models based on these features (AUC = 0.77). Conclusion The three-gene methylation prediction model for EGC LNM is highly effective in both cancerous and adjacent tissue samples in a postoperative setting, providing reliable diagnostic information. This extends its clinical utility, particularly when tumor samples are scarce, making it a valuable tool for evaluating LNM status and assisting in treatment planning.
Collapse
Affiliation(s)
- Shang Chen
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Shoubin Long
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
| | - Yaru Liu
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
- School of the First Clinical Medical, Ningxia Medical University, Yinchuan, China
| | - Shenglong Wang
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
- School of the First Clinical Medical, Ningxia Medical University, Yinchuan, China
| | - Qian Hu
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Dixian Luo
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen University, Shenzhen, China
- School of the First Clinical Medical, Ningxia Medical University, Yinchuan, China
- Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
2
|
Zhang Z, Wu W, Li X, Lin S, Lei Q, Yu L, Lin J, Sun L, Zhang H, Lin L. Prediction and verification of benignancy and malignancy of pulmonary nodules based on inflammatory related biological markers. Heliyon 2024; 10:e34585. [PMID: 39144966 PMCID: PMC11320450 DOI: 10.1016/j.heliyon.2024.e34585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024] Open
Abstract
Objective Inflammation plays an important role in the transformation of pulmonary nodules (PNs) from benign to malignant. Prediction of benignancy and malignancy of PNs is still lacking efficacy methods. Although Mayo or Brock model have been widely applied in clinical practices, their application conditions are limited. This study aims to construct a diagnostic model of PNs by machine learning using inflammation-related biological markers (IRBMs). Methods Inflammatory related genes (IRGs) were first extracted from GSE135304 chip data. Then, differentially expressed genes (DEGs) and infiltrating immune cells were screened between malignant pulmonary nodules (MN) and benign pulmonary nodule (BN). Correlation analysis was performed on DEGs and infiltrating immune cells. Molecular modules of IRGs were identified through Consistency cluster analysis. Subsequently, IRBMs in IRGs modules were filtered through Weighted gene co-expression network analysis (WGCNA). An optimal diagnostic model was established using machine learning methods. Finally, external dataset GSE108375 was used to verify this result. Results 4 hub IRGs and 3 immune cells showed significantly difference between MN and BN, C1 and C2 module, namely PRTN3, ELANE, NFKB1 and CTLA4, T cells CD4 naïve, NK cells activated and Monocytes. IRBMs were screened from black module and yellowgreen module through WGCNA analysis. The Support vector machines (SVM) was identified as the optimal model with the Area Under Curve (AUC) was 0.753. A nomogram was established based on 5 hub IRBMs, namely HS.137078, KLC3, C13ORF15, STOM and KCTD13. Finally, external dataset GSE108375 verified this result, with the AUC was 0.718. Conclusion SVM model established by 5 hub IRBMs was able to effectively identify MN or BN. Accumulating inflammation and immune dysfunction were important to the transformation from BN to MN.
Collapse
Affiliation(s)
- Zexin Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenfeng Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuewei Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siqi Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiwei Lei
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Yu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jietao Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Sun
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haibo Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Batochir C, Kim IA, Jo EJ, Kim EB, Kim HJ, Hur JY, Kim DW, Park HK, Lee KY. Discrimination of Lung Cancer and Benign Lung Diseases Using BALF Exosome DNA Methylation Profile. Cancers (Basel) 2024; 16:2765. [PMID: 39123492 PMCID: PMC11311347 DOI: 10.3390/cancers16152765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Benign lung diseases are common and often do not require specific treatment, but they pose challenges in the distinguishing of them from lung cancer during low-dose computed tomography (LDCT). This study presents a comprehensive methylation analysis using real-time PCR for minimally invasive diagnoses of lung cancer via employing BALF exosome DNA. A panel of seven epigenetic biomarkers was identified, exhibiting specific methylation patterns in lung cancer BALF exosome DNA. This panel achieved an area under the curve (AUC) of 0.97, with sensitivity and specificity rates of 88.24% and 97.14%, respectively. Each biomarker showed significantly higher mean methylation levels (MMLs) in both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) compared to non-cancer groups, with fold changes from 1.7 to 13.36. The MMLs of the biomarkers were found to be moderately elevated with increasing patient age and smoking history, regardless of sex. A strong correlation was found between the MMLs and NSCLC stage progression, with detection sensitivities of 79% for early stages and 92% for advanced stages. In the validation cohort, the model demonstrated an AUC of 0.95, with 94% sensitivity and specificity. Sensitivity for early-stage NSCLC detection improved from 88.00% to 92.00% when smoking history was included as an additional risk factor.
Collapse
Affiliation(s)
- Chinbayar Batochir
- Seasun Biomaterials, Inc., Daejeon 34015, Republic of Korea; (C.B.); (E.J.J.); (E.-B.K.); (D.W.K.); (H.K.P.)
| | - In Ae Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Republic of Korea; (I.A.K.); (H.J.K.); (J.Y.H.)
| | - Eun Ji Jo
- Seasun Biomaterials, Inc., Daejeon 34015, Republic of Korea; (C.B.); (E.J.J.); (E.-B.K.); (D.W.K.); (H.K.P.)
| | - Eun-Bi Kim
- Seasun Biomaterials, Inc., Daejeon 34015, Republic of Korea; (C.B.); (E.J.J.); (E.-B.K.); (D.W.K.); (H.K.P.)
| | - Hee Joung Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Republic of Korea; (I.A.K.); (H.J.K.); (J.Y.H.)
| | - Jae Young Hur
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Republic of Korea; (I.A.K.); (H.J.K.); (J.Y.H.)
| | - Do Won Kim
- Seasun Biomaterials, Inc., Daejeon 34015, Republic of Korea; (C.B.); (E.J.J.); (E.-B.K.); (D.W.K.); (H.K.P.)
| | - Hee Kyung Park
- Seasun Biomaterials, Inc., Daejeon 34015, Republic of Korea; (C.B.); (E.J.J.); (E.-B.K.); (D.W.K.); (H.K.P.)
| | - Kye Young Lee
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Republic of Korea; (I.A.K.); (H.J.K.); (J.Y.H.)
| |
Collapse
|
4
|
Xue Y, Zhao G, Song L, Qiao L, Huang C, Wang K, Wang T. The signature of cancer methylation markers in maternal plasma: Factors influencing the development and application of cancer liquid biopsy assay. Gene 2024; 906:148261. [PMID: 38342253 DOI: 10.1016/j.gene.2024.148261] [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: 10/16/2023] [Revised: 01/17/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND DNA methylation is highly correlated with cancer and embryo development, and plasma-based methylation markers have been widely used for cancer early detection. However, whether the commonly used cancer methylation markers cause "false positives" in the plasma of pregnant women has not been comprehensively evaluated. METHODS We conducted a case-control study from February 2021 to March 2023, which included 138 pregnant women and 44 control women. Plasma cell-free DNA (cfDNA) was isolated and bisulfite-converted, and then the methylation levels of eight methylated markers related to gastrointestinal cancer (SEPT9, SDC2, C9orf50, KCNQ5, CLIP4, TFPI2, ELMO1 and ZNF582) and three markers related to lung cancer (SHOX2, RASSF1A and PTGER4) were analyzed. RESULTS When comparing the plasma of pregnant women to that of control women, SEPT9, CLIP4, ZNF582, SHOX2, RASSF1A and PTGER showed significantly higher levels of methylation (p < 0.05). These positive signals originate from the placenta/fetus rather than the mother. We found no discernible difference in DNA methylation levels between fetal cfDNA fractions of < 10 % and ≥ 10 % in pregnant women (p > 0.05), while CLIP4 and PTGER4 showed high methylation levels in the assisted fertilization group compared to the natural fertilization group (p < 0.05). CONCLUSION Our study shows that cancer and fetus/placenta exhibit similar DNA methylation patterns, and some gastrointestinal cancer and lung cancer-related methylation markers also show positives in maternal plasma. This is an important consideration in the design and application of plasma-based cancer liquid biopsy assays.
Collapse
Affiliation(s)
- Ying Xue
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Guodong Zhao
- ZJUT Yinhu Research Institute of Innovation and Entrepreneurship Fuyang District, Hangzhou, Zhejiang 311400, China; Zhejiang University Kunshan Biotechnology Laboratory, Zhejiang University Kunshan Innovation Institute, Kunshan, Jiangsu 215300, China; Suzhou VersaBio Technologies Co. Ltd., Kunshan, Jiangsu 215300, China.
| | - Lishuang Song
- Suzhou VersaBio Technologies Co. Ltd., Kunshan, Jiangsu 215300, China
| | - Longwei Qiao
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Chao Huang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Kai Wang
- Suzhou VersaBio Technologies Co. Ltd., Kunshan, Jiangsu 215300, China
| | - Ting Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
5
|
Qu Y, Zhang X, Qiao R, Di F, Song Y, Wang J, Ji L, Zhang J, Gu W, Fang Y, Han B, Yang R, Dai L, Ouyang S. Blood FOLR3 methylation dysregulations and heterogeneity in non-small lung cancer highlight its strong associations with lung squamous carcinoma. Respir Res 2024; 25:59. [PMID: 38273401 PMCID: PMC10809478 DOI: 10.1186/s12931-024-02691-8] [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: 09/30/2023] [Accepted: 01/14/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) accounts for the vast majority of lung cancers. Early detection is crucial to reduce lung cancer-related mortality. Aberrant DNA methylation occurs early during carcinogenesis and can be detected in blood. It is essential to investigate the dysregulated blood methylation markers for early diagnosis of NSCLC. METHODS NSCLC-associated methylation gene folate receptor gamma (FOLR3) was selected from an Illumina 850K array analysis of peripheral blood samples. Mass spectrometry was used for validation in two independent case-control studies (validation I: n = 2548; validation II: n = 3866). Patients with lung squamous carcinoma (LUSC) or lung adenocarcinoma (LUAD), normal controls (NCs) and benign pulmonary nodule (BPN) cases were included. FOLR3 methylations were compared among different populations. Their associations with NSCLC clinical features were investigated. Receiver operating characteristic analyses, Kruskal-Wallis test, Wilcoxon test, logistics regression analysis and nomogram analysis were performed. RESULTS Two CpG sites (CpG_1 and CpG_2) of FOLR3 was significantly lower methylated in NSCLC patients than NCs in the discovery round. In the two validations, both LUSC and LUAD patients presented significant FOLR3 hypomethylations. LUSC patients were highlighted to have significantly lower methylation levels of CpG_1 and CpG_2 than BPN cases and LUAD patients. Both in the two validations, CpG_1 methylation and CpG_2 methylation could discriminate LUSC from NCs well, with areas under the curve (AUCs) of 0.818 and 0.832 in validation I, and 0.789 and 0.780 in validation II. They could also differentiate LUAD from NCs, but with lower efficiency. CpG_1 and CpG_2 methylations could also discriminate LUSC from BPNs well individually in the two validations. With the combined dataset of two validations, the independent associations of age, gender, and FOLR3 methylation with LUSC and LUAD risk were shown and the age-gender-CpG_1 signature could discriminate LUSC and LUAD from NCs and BPNs, with higher efficiency for LUSC. CONCLUSIONS Blood-based FOLR3 hypomethylation was shown in LUSC and LUAD. FOLR3 methylation heterogeneity between LUSC and LUAD highlighted its stronger associations with LUSC. FOLR3 methylation and the age-gender-CpG_1 signature might be novel diagnostic markers for the early detection of NSCLC, especially for LUSC.
Collapse
Affiliation(s)
- Yunhui Qu
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University and the Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Xiuzhi Zhang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Rong Qiao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Feifei Di
- Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, 210000, China
| | - Yakang Song
- Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, 210000, China
| | - Jun Wang
- Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, 210000, China
| | - Longtao Ji
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Zhang
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210000, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210000, China
| | - Yifei Fang
- Department of Respiratory and Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Rongxi Yang
- Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, 210000, China.
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, 210000, China.
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, 450052, China.
| | - Songyun Ouyang
- Department of Respiratory and Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Zhao H, Jiang R, Feng Z, Wang X, Zhang C. Transcription factor LHX9 (LIM Homeobox 9) enhances pyruvate kinase PKM2 activity to induce glycolytic metabolic reprogramming in cancer stem cells, promoting gastric cancer progression. J Transl Med 2023; 21:833. [PMID: 37980488 PMCID: PMC10657563 DOI: 10.1186/s12967-023-04658-7] [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: 05/22/2023] [Accepted: 10/25/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Glycolytic metabolic reprogramming is a phenomenon in which cells undergo altered metabolic patterns during malignant transformation, mainly involving various aspects of glycolysis, electron transport chain, oxidative phosphorylation, and pentose phosphate pathway. This reprogramming phenomenon can be used as one of the markers of tumorigenesis and development. Pyruvate kinase is the third rate-limiting enzyme in the sugar metabolism process by specifically catalyzing the irreversible conversion of PEP to pyruvate. PURPOSE This study aimed to reveal the critical mediator(s) that regulate glycolytic metabolism reprogramming in gastric cancer and their underlying molecular mechanism and then explore the molecular mechanisms by which LHX9 may be involved in regulating gastric cancer (GC) progression. METHODS Firstly, we downloaded the GC and glycolysis-related microarray datasets from TCGA and MSigDB databases and took the intersection to screen out the transcription factor LHX9 that regulates GC glycolytic metabolic reprogramming. Software packages were used for differential analysis, single gene predictive analysis, and Venn diagram. In addition, an enrichment analysis of the glycolytic pathway was performed. Immunohistochemical staining was performed for LHX9 and PKM2 protein expression in 90 GC patients, and the association between their expressions was evaluated by Spearman's correlation coefficient method. Three human GC cell lines (AGS, NCI-N87, HGC-27) were selected for in vitro experimental validation. Flow cytometry was utilized to determine the stem cell marker CD44 expression status in GCSCs. A sphere formation assay was performed to evaluate the sphere-forming capabilities of GCSCs. In addition, RT-qPCR and Western blot experiments were employed to investigate the tumor stem cell markers OCT4 and SOX2 expression levels in GCSCs. Furthermore, a lentiviral expression vector was constructed to assess the impact of downregulating LHX9 or PKM2 on the glycolytic metabolic reprogramming of GCSCs. The proliferation, migration, and invasion of GCSCs were then detected by CCK-8, EdU, and Transwell assays. Subsequently, the mutual binding of LHX9 and PKM2 was verified using chromatin immunoprecipitation and dual luciferase reporter genes. In vivo experiments were verified by establishing a subcutaneous transplantation tumor model in nude mice, observing the size and volume of tumors in vivo in nude mice, and obtaining fresh tissues for subsequent experiments. RESULTS Bioinformatics analysis revealed that LHX9 might be involved in the occurrence and development of GC through regulating glycolytic metabolism. High LHX9 expression could be used as a reference marker for prognosis prediction of GC patients. Clinical tissue assays revealed that LHX9 and PKM2 were highly expressed in GC tissues. Meanwhile, GC tissues also highly expressed glycolysis-associated protein GLUT1 and tumor cell stemness marker CD44. In vitro cellular assays showed that LHX9 could enhance its activity and induce glycolytic metabolic reprogramming in GCSCs through direct binding to PKM2. In addition, the knockdown of LHX9 inhibited PKM2 activity and glycolytic metabolic reprogramming and suppressed the proliferation, migration, and invasive ability of GCSCs. In vivo animal experiments further confirmed that the knockdown of LHX9 could reduce the tumorigenic ability of GCSCs in nude mice by inhibiting PKM2 activity and glycolytic metabolic reprogramming. CONCLUSION The findings suggest that both LHX9 and PKM2 are highly expressed in GCs, and LHX9 may induce the reprogramming of glycolytic metabolism through transcriptional activation of PKM2, enhancing the malignant biological properties of GCSCs and ultimately promoting GC progression.
Collapse
Affiliation(s)
- Hongying Zhao
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China.
| | - Rongke Jiang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| | - Zhijing Feng
- Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Xue Wang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| | - Chunmei Zhang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| |
Collapse
|
7
|
Wu P, Weng H, Luo W, Zhan Y, Xiong L, Zhang H, Yan H. An improved Yolov5s based on transformer backbone network for detection and classification of bronchoalveolar lavage cells. Comput Struct Biotechnol J 2023; 21:2985-3001. [PMID: 37249972 PMCID: PMC10209489 DOI: 10.1016/j.csbj.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Biological tissue information of the lung, such as cells and proteins, can be obtained from bronchoalveolar lavage fluid (BALF), through which it can be used as a complement to lung biopsy pathology. BALF cells can be confused with each other due to the similarity of their characteristics and differences in the way sections are handled or viewed. This poses a great challenge for cell detection. In this paper, An Improved Yolov5s Based on Transformer Backbone Network for Detection and Classification of BALF Cells is proposed, focusing on the detection of four types of cells in BALF: macrophages, lymphocytes, neutrophils and eosinophils. The network is mainly based on the Yolov5s network and uses Swin Transformer V2 technology in the backbone network to improve cell detection accuracy by obtaining global information; the C3Ghost module (a variant of the Convolutional Neural Network architecture) is used in the neck network to reduce the number of parameters during feature channel fusion and to improve feature expression performance. In addition, embedding intersection over union Loss (EIoU_Loss) was used as a bounding box regression loss function to speed up the bounding box regression rate, resulting in higher accuracy of the algorithm. The experiments showed that our model could achieve mAP of 81.29% and Recall of 80.47%. Compared to the original Yolov5s, the mAP has improved by 3.3% and Recall by 3.67%. We also compared it with Yolov7 and the newly launched Yolov8s. mAP improved by 0.02% and 2.36% over Yolov7 and Yolov8s respectively, while the FPS of our model was higher than both of them, achieving a balance of efficiency and accuracy, further demonstrating the superiority of our model.
Collapse
Affiliation(s)
- Puzhen Wu
- The Faculty of Architecture, Civil and Transportation Engineering, Beijing University of Technology, Beijing 100124, China
- Beijing-Dublin International College, Beijing University of Technology, Beijing 100124, China
| | - Han Weng
- Beijing-Dublin International College, Beijing University of Technology, Beijing 100124, China
| | - Wenting Luo
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Yi Zhan
- Beijing-Dublin International College, Beijing University of Technology, Beijing 100124, China
| | - Lixia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Hongyan Zhang
- Department of Burn, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang 330066, China
| | - Hai Yan
- The Faculty of Architecture, Civil and Transportation Engineering, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
8
|
Gupta MM, Gilhotra R, Deopa D, Bhat AA, Thapa R, Singla N, Kulshrestha R, Gupta G. Epigenetics of Pulmonary Tuberculosis. TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:127-144. [DOI: 10.1007/978-981-99-4780-5_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Bronchoalveolar Lavage Fluid-Isolated Biomarkers for the Diagnostic and Prognostic Assessment of Lung Cancer. Diagnostics (Basel) 2022; 12:diagnostics12122949. [PMID: 36552956 PMCID: PMC9776496 DOI: 10.3390/diagnostics12122949] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Lung cancer is considered one of the most fatal malignant neoplasms because of its late detection. Detecting molecular markers in samples from routine bronchoscopy, including many liquid-based cytology procedures, such as bronchoalveolar lavage fluid (BALF), could serve as a favorable technique to enhance the efficiency of a lung cancer diagnosis. BALF analysis is a promising approach to evaluating the tumor progression microenvironment. BALF's cellular and non-cellular components dictate the inflammatory response in a cancer-proliferating microenvironment. Furthermore, it is an essential material for detecting clinically significant predictive and prognostic biomarkers that may aid in guiding treatment choices and evaluating therapy-induced toxicities in lung cancer. In the present article, we have reviewed recent literature about the utility of BALF analysis for detecting markers in different stages of tumor cell metabolism, employing either specific biomarker assays or broader omics approaches.
Collapse
|
10
|
Yu Y, Xue W, Liu Z, Chen S, Wang J, Peng Q, Xu L, Liu X, Cui C, Fan JB. A novel DNA methylation marker to identify lymph node metastasis of colorectal cancer. Front Oncol 2022; 12:1000823. [PMID: 36313642 PMCID: PMC9614158 DOI: 10.3389/fonc.2022.1000823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Lymph node metastasis (LNM) of colorectal cancer (CRC) is an important factor for both prognosis and treatment. Given the deficiencies of conventional tests, we aim to discover novel DNA methylation markers to efficiently identify LNM status of CRC. In this study, genome-wide methylation sequencing was performed in a cohort (n=30) using fresh CRC tissue to discover differentially methylated markers. These markers were subsequently validated with fluorescence quantitative PCR in a cohort (n=221), and the optimal marker was compared to conventional diagnostic methods. Meanwhile, immunohistochemistry was used to verify the effectiveness of the antibody corresponding to this marker in a cohort (n=56). LBX2 achieved an AUC of 0.87, specificity of 87.3%, sensitivity of 75.7%, and accuracy of 81.9%, which outperformed conventional methods including imaging (CT, PET-CT) with an AUC of 0.52, CA199 with an AUC of 0.58, CEA with an AUC of 0.56. LBX2 was also superior to clinicopathological indicators including the depth of tumor invasion and lymphatic invasion with an AUC of 0.61and 0.63 respectively. Moreover, the AUC of LBX2 antibody was 0.84, which was also better than these conventional methods. In conclusion, A novel methylation marker LBX2 could be used as a simple, cost-effective, and reliable diagnostic method for LNM of CRC.
Collapse
Affiliation(s)
- Yingdian Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenyuan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zefeng Liu
- Department of General Surgery, Zhujiang Hosipital, Southern Medical University, Guangzhou, China
| | - Shang Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Wang
- AnchorDx Medical Co., Ltd., International Bio-Island, Guangzhou, China
| | - Quanzhou Peng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Linhao Xu
- AnchorDx Medical Co., Ltd., International Bio-Island, Guangzhou, China
| | - Xin Liu
- AnchorDx Medical Co., Ltd., International Bio-Island, Guangzhou, China
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hosipital, Southern Medical University, Guangzhou, China
- *Correspondence: Jian-Bing Fan, ; Chunhui Cui,
| | - Jian-Bing Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- AnchorDx Medical Co., Ltd., International Bio-Island, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Jian-Bing Fan, ; Chunhui Cui,
| |
Collapse
|
11
|
Lin F, Hu X, Zhang Y, Ye S, Gu Y, Yan B, Wang L, Jiang Y. Upregulated TIGIT+ and Helios+ regulatory T cell levels in bronchoalveolar lavage fluid of NSCLC patients. Mol Immunol 2022; 147:40-49. [DOI: 10.1016/j.molimm.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/17/2022] [Accepted: 04/17/2022] [Indexed: 12/09/2022]
|
12
|
Yousefi PD, Suderman M, Langdon R, Whitehurst O, Davey Smith G, Relton CL. DNA methylation-based predictors of health: applications and statistical considerations. Nat Rev Genet 2022; 23:369-383. [PMID: 35304597 DOI: 10.1038/s41576-022-00465-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
DNA methylation data have become a valuable source of information for biomarker development, because, unlike static genetic risk estimates, DNA methylation varies dynamically in relation to diverse exogenous and endogenous factors, including environmental risk factors and complex disease pathology. Reliable methods for genome-wide measurement at scale have led to the proliferation of epigenome-wide association studies and subsequently to the development of DNA methylation-based predictors across a wide range of health-related applications, from the identification of risk factors or exposures, such as age and smoking, to early detection of disease or progression in cancer, cardiovascular and neurological disease. This Review evaluates the progress of existing DNA methylation-based predictors, including the contribution of machine learning techniques, and assesses the uptake of key statistical best practices needed to ensure their reliable performance, such as data-driven feature selection, elimination of data leakage in performance estimates and use of generalizable, adequately powered training samples.
Collapse
Affiliation(s)
- Paul D Yousefi
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - Matthew Suderman
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - Ryan Langdon
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - Oliver Whitehurst
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK.
| |
Collapse
|
13
|
Wen SWC, Wen J, Hansen TF, Jakobsen A, Hilberg O. Cell Free Methylated Tumor DNA in Bronchial Lavage as an Additional Tool for Diagnosing Lung Cancer-A Systematic Review. Cancers (Basel) 2022; 14:2254. [PMID: 35565384 PMCID: PMC9099950 DOI: 10.3390/cancers14092254] [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: 03/31/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
This systematic review investigated circulating methylated tumor DNA in bronchial lavage fluid for diagnosing lung cancer. PROSPERO registration CRD42022309470. PubMed, Embase, Medline, and Web of Science were searched on 9 March 2022. Studies of adults with lung cancer or undergoing diagnostic workup for suspected lung cancer were included if they used bronchial lavage fluid, analyzed methylated circulating tumor DNA, and reported the diagnostic properties. Sensitivity, specificity, and lung cancer prevalence were summarized in forest plots. Risk of bias was assessed using the QUADAS-2 tool. A total of 25 studies were included. All were case-control studies, most studies used cell pellet for analysis by quantitative PCR. Diagnostic sensitivity ranged from 0% for a single gene to 97% for a four-gene panel. Specificity ranged from 8% for a single gene to 100%. The studies employing a gene panel decreased the specificity, and no gene panel had a perfect specificity of 100%. In conclusion, methylated circulating tumor DNA can be detected in bronchial lavage, and by employing a gene panel the sensitivity can be increased to clinically relevant levels. The available evidence regarding applicability in routine clinical practice is limited. Prospective, randomized clinical trials are needed to determine the further usefulness of this biomarker.
Collapse
Affiliation(s)
- Sara Witting Christensen Wen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (T.F.H.); (A.J.)
- Department of Regional Health Research, J.B. Winsloews Vej 19, 3rd Floor, 5000 Odense C, Denmark;
| | - Jan Wen
- General Practice, Region of Southern Denmark, Damhaven 12, 7100 Vejle, Denmark;
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (T.F.H.); (A.J.)
- Department of Regional Health Research, J.B. Winsloews Vej 19, 3rd Floor, 5000 Odense C, Denmark;
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (T.F.H.); (A.J.)
- Department of Regional Health Research, J.B. Winsloews Vej 19, 3rd Floor, 5000 Odense C, Denmark;
| | - Ole Hilberg
- Department of Regional Health Research, J.B. Winsloews Vej 19, 3rd Floor, 5000 Odense C, Denmark;
| |
Collapse
|
14
|
Chen S, Yu Y, Li T, Ruan W, Wang J, Peng Q, Yu Y, Cao T, Xue W, Liu X, Chen Z, Yu J, Fan JB. A novel DNA methylation signature associated with lymph node metastasis status in early gastric cancer. Clin Epigenetics 2022; 14:18. [PMID: 35115040 PMCID: PMC8811982 DOI: 10.1186/s13148-021-01219-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Background Lymph node metastasis (LNM) is an important factor for both treatment and prognosis of early gastric cancer (EGC). Current methods are insufficient to evaluate LNM in EGC due to suboptimal accuracy. Herein, we aim to identify methylation signatures for LNM of EGC, facilitate precision diagnosis, and guide treatment modalities. Methods For marker discovery, genome-wide methylation sequencing was performed in a cohort (marker discovery) using 47 fresh frozen (FF) tissue samples. The identified signatures were subsequently characterized for model development using formalin-fixed paraffin-embedded (FFPE) samples by qPCR assay in a second cohort (model development cohort, n = 302, training set: n = 151, test set: n = 151). The performance of the established model was further validated using FFPE samples in a third cohorts (validation cohort, n = 130) and compared with image-based diagnostics, conventional clinicopathology-based model (conventional model), and current standard workups. Results Fifty LNM-specific methylation signatures were identified de novo and technically validated. A derived 3-marker methylation model for LNM diagnosis was established that achieved an AUC of 0.87 and 0.88, corresponding to the specificity of 80.9% and 85.7%, sensitivity of 80.6% and 78.1%, and accuracy of 80.8% and 83.8% in the test set of model development cohort and validation cohort, respectively. Notably, this methylation model outperformed computed tomography (CT)-based imaging with a superior AUC (0.88 vs. 0.57, p < 0.0001) and individual clinicopathological features in the validation cohort. The model integrated with clinicopathological features demonstrated further enhanced AUCs of 0.89 in the same cohort. The 3-marker methylation model and integrated model reduced 39.4% and 41.5% overtreatment as compared to standard workups, respectively. Conclusions A novel 3-marker methylation model was established and validated that shows diagnostic potential to identify LNM in EGC patients and thus reduce unnecessary gastrectomy in EGC. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01219-x.
Collapse
Affiliation(s)
- Shang Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanqi Yu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tao Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weimei Ruan
- AnchorDx Medical Co., Ltd, Unit 502, No. 8, 3rd Luoxuan Road, International Bio-Island, Guangzhou, 510300, China
| | - Jun Wang
- AnchorDx Medical Co., Ltd, Unit 502, No. 8, 3rd Luoxuan Road, International Bio-Island, Guangzhou, 510300, China
| | - Quanzhou Peng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, Shenzhen People's Hospital, Shennan Dong Lu, Luohu District, Shenzhen, 518002, China
| | - Yingdian Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tianfeng Cao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenyuan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Liu
- AnchorDx, Inc., 46305 Landing Pkwy, Fremont, CA, 94538, USA
| | - Zhiwei Chen
- AnchorDx Medical Co., Ltd, Unit 502, No. 8, 3rd Luoxuan Road, International Bio-Island, Guangzhou, 510300, China.,AnchorDx, Inc., 46305 Landing Pkwy, Fremont, CA, 94538, USA
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jian-Bing Fan
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,AnchorDx Medical Co., Ltd, Unit 502, No. 8, 3rd Luoxuan Road, International Bio-Island, Guangzhou, 510300, China.
| |
Collapse
|