1
|
Liang G, He J, Chen T, Zhang L, Yu K, Shen W. Identification of ALDH7A1 as a DNA-methylation-driven gene in lung squamous cell carcinoma. Ann Med 2025; 57:2442529. [PMID: 39711312 DOI: 10.1080/07853890.2024.2442529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Deoxyribose nucleic acid (DNA) methylation is an important epigenetic modification that plays an important role in the occurrence and development of tumors. Identifying key methylation-driven genes that affect the prognosis of lung squamous cell carcinoma (LUSC) can provide direction for targeted therapy research. METHODS AND RESULTS Methylation and RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA). The MethylMix package was used to integrate and analyze the methylation and gene expression data from TCGA, and the LUSC dataset (GSE37745) was downloaded from GEO for validation. Forty-five DNA-methylation-driven genes (MDGs) were obtained, and 3 genes (TRIM61, SMIM22, and ALDH7A1) were significantly associated with survival by using univariate and multivariate Cox regression. A risk model was constructed. KM analysis showed that patients with high-risk scores had poor survival. A nomination plot for prognosis prediction of LUSC patients was constructed, which showed a good predictive efficiency for tumor prognosis. The high expression of ALDH7A1 was an independent risk factor for poor prognosis in LUSC. The expression of ALDH7A1 in LUSC was negatively correlated with its methylation status (COR = -0.655). GSEA analysis showed that high expression of ALDH7A1 could activate multiple signaling pathways (JAK-STAT signaling pathway and mTOR signaling pathway). In vitro cell experiments confirmed that in LUSC, silencing ALDH7A1 could inhibit tumor progression, while overexpression of ALDH7A1 could promote tumor progression. CONCLUSION Our results indicated that ALDH7A1, a newly discovered MDG in LUSC, could act as an independent prognostic factor for OS in LUSC, with the potential to become a potential target for LUSC diagnosis and treatment. High expression of ALDH7A1 in LUSC could promote the occurrence and development of tumors. Signaling pathways, such as JAK-STAT and mTOR signaling pathways, might regulate the high expression of ALDH7A1.
Collapse
Affiliation(s)
- Gaofeng Liang
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jinxian He
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tian Chen
- Department of Radiation Oncology, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Liang Zhang
- cDepartment of Respiratory Medicine, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaizhong Yu
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Weiyu Shen
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Duong HQ, Hoang MC, Nguyen TH, Nguyen PT, Le VT, Dao TN, Ngo VL, Dang TH. Aldehyde Dehydrogenase-1A1 (ALDH1A1): The Novel Regulator of Chemoresistance in Pancreatic Cancer Cells. Cancer Control 2024; 31:10732748241305835. [PMID: 39611960 PMCID: PMC11607765 DOI: 10.1177/10732748241305835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Aldehyde dehydrogenase-1A1 (ALDH1A1), a member of a superfamily of 19 isozymes, exhibits various biological functions and is involved in several important physiological and pathological processes, including those associated with various diseases including cancers such as pancreatic cancer. Chemotherapy is one of the most important strategies for the treatment of pancreatic cancer; however, the chemoresistance exhibited by pancreatic cancer cells is a leading cause of chemotherapy failure. It has been reported that overexpression of ALDH1A1 significantly correlates with poor prognosis and tumor aggressiveness, and is clinically associated with chemoresistance. Additionally, ALDH1A1 may serve as a novel regulator for the diagnosis and prognosis of cancer resistance. In particular, ALDH1A1 can promote cancer progression by facilitating the manifestation of cancer stem cell properties. However, the molecular mechanism by which ALDH1A1 clinically regulates the development of chemoresistance, and its role in prognosis and cancer stem cells, including pancreatic cancer stem cells, remain unclear. Therefore, the current review aims to summarize the clinical functions of ALDH1A1 as a novel regulator of chemoresistance, prognosis, and cancer stem cell development in pancreatic cancer.
Collapse
Affiliation(s)
- Hong-Quan Duong
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| | - Minh-Cong Hoang
- Laboratory Department, Yenphong Medical Center, Bacninh, Vietnam
| | - Thi-Hue Nguyen
- Laboratory Department, Bacgiang General Hospital, Bacgiang, Vietnam
| | | | - Van-Thu Le
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| | - Thi-Nguyet Dao
- Pathology Department, Ducgiang General Hospital, Hanoi, Vietnam
| | - Van-Lang Ngo
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, Vietnam
| | - The-Hung Dang
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| |
Collapse
|
3
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
4
|
Pouliquen DL, Ortone G, Rumiano L, Boissard A, Henry C, Blandin S, Guette C, Riganti C, Kopecka J. Long-Chain Acyl Coenzyme A Dehydrogenase, a Key Player in Metabolic Rewiring/Invasiveness in Experimental Tumors and Human Mesothelioma Cell Lines. Cancers (Basel) 2023; 15:cancers15113044. [PMID: 37297007 DOI: 10.3390/cancers15113044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cross-species investigations of cancer invasiveness are a new approach that has already identified new biomarkers which are potentially useful for improving tumor diagnosis and prognosis in clinical medicine and veterinary science. In this study, we combined proteomic analysis of four experimental rat malignant mesothelioma (MM) tumors with analysis of ten patient-derived cell lines to identify common features associated with mitochondrial proteome rewiring. A comparison of significant abundance changes between invasive and non-invasive rat tumors gave a list of 433 proteins, including 26 proteins reported to be exclusively located in mitochondria. Next, we analyzed the differential expression of genes encoding the mitochondrial proteins of interest in five primary epithelioid and five primary sarcomatoid human MM cell lines; the most impressive increase was observed in the expression of the long-chain acyl coenzyme A dehydrogenase (ACADL). To evaluate the role of this enzyme in migration/invasiveness, two epithelioid and two sarcomatoid human MM cell lines derived from patients with the highest and lowest overall survival were studied. Interestingly, sarcomatoid vs. epithelioid cell lines were characterized by higher migration and fatty oxidation rates, in agreement with ACADL findings. These results suggest that evaluating mitochondrial proteins in MM specimens might identify tumors with higher invasiveness.
Collapse
Affiliation(s)
- Daniel L Pouliquen
- Université d'Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Giacomo Ortone
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | - Letizia Rumiano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | - Alice Boissard
- Université d'Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Cécile Henry
- Université d'Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Stéphanie Blandin
- CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes Université, F-44000 Nantes, France
| | - Catherine Guette
- Université d'Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| |
Collapse
|
5
|
Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: Potential biomarkers and therapeutic targets. MedComm (Beijing) 2023; 4:e195. [PMID: 36694633 PMCID: PMC9842923 DOI: 10.1002/mco2.195] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
The family of aldehyde dehydrogenases (ALDHs) contains 19 isozymes and is involved in the oxidation of endogenous and exogenous aldehydes to carboxylic acids, which contributes to cellular and tissue homeostasis. ALDHs play essential parts in detoxification, biosynthesis, and antioxidants, which are of important value for cell proliferation, differentiation, and survival in normal body tissues. However, ALDHs are frequently dysregulated and associated with various diseases like Alzheimer's disease, Parkinson's disease, and especially solid tumors. Notably, the involvement of the ALDHs in tumor progression is responsible for the maintenance of the stem-cell-like phenotype, triggering rapid and aggressive clinical progressions. ALDHs have captured increasing attention as biomarkers for disease diagnosis and prognosis. Nevertheless, these require further longitudinal clinical studies in large populations for broad application. This review summarizes our current knowledge regarding ALDHs as potential biomarkers in tumors and several non-tumor diseases, as well as recent advances in our understanding of the functions and underlying molecular mechanisms of ALDHs in disease development. Finally, we discuss the therapeutic potential of ALDHs in diseases, especially in tumor therapy with an emphasis on their clinical implications.
Collapse
Affiliation(s)
- Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjingChina
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
6
|
Lu HJ, Chuang CY, Chen MK, Su CW, Yang WE, Yeh CM, Lai KM, Tang CH, Lin CW, Yang SF. The impact of ALDH7A1 variants in oral cancer development and prognosis. Aging (Albany NY) 2022; 14:4556-4571. [PMID: 35613852 PMCID: PMC9186774 DOI: 10.18632/aging.204099] [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: 11/04/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022]
Abstract
The gene encoding aldehyde dehydrogenase 7 family member A1 (ALDH7A1) has been associated with the development and prognosis in multiple cancers; however, the role of ALDH7A1 polymorphisms in oral cancer remains unknown. For this purpose, the influences of ALDH7A1 rs13182402 and rs12659017 on oral cancer development and prognosis were analyzed. Our resulted showed that ALDH7A1 rs13182402 genotype had less pathologic nodal metastasis among betel quid chewer. ALDH7A1 rs13182402 also corresponded to higher expressions in upper aerodigestive mucosa, whole blood, the musculoskeletal system and oral cancer tissues than did the ALDH7A1 wild type. Furthermore, ALDH7A1 overexpression in oral cancer cells increased in vitro migration, whereas its silencing reduced cell migration. Conversely, ALDH7A1 expression in tumor tissues and in patients with advanced disease was lower than that in normal tissues and in patients with early-stage disease. When the patients were classified into ALDH7A1-high and -low-expression groups, the high-ALDH7A1 group had superior outcomes in progression-free survival than the low-ALDH7A1 group (5-year survival of 58.7% vs. 48.0%, P = 0.048) did. In conclusion, patients with high ALDH7A1 expression might, however, have more favorable prognoses than those with low ALDH7A1 expression have.
Collapse
Affiliation(s)
- Hsueh-Ju Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Yi Chuang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Wen Su
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Ming Yeh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Kuan-Ming Lai
- Division of Hematology and Oncology, Department of Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
7
|
Huang G, Zhang J, Gong L, Huang Y, Liu D. A glycolysis-based three-gene signature predicts survival in patients with lung squamous cell carcinoma. BMC Cancer 2021; 21:626. [PMID: 34044809 PMCID: PMC8161559 DOI: 10.1186/s12885-021-08360-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 05/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung cancer is one of the most lethal and most prevalent malignant tumors worldwide, and lung squamous cell carcinoma (LUSC) is one of the major histological subtypes. Although numerous biomarkers have been found to be associated with prognosis in LUSC, the prediction effect of a single gene biomarker is insufficient, especially for glycolysis-related genes. Therefore, we aimed to develop a novel glycolysis-related gene signature to predict survival in patients with LUSC. METHODS The mRNA expression files and LUSC clinical information were obtained from The Cancer Genome Atlas (TCGA) dataset. RESULTS Based on Gene Set Enrichment Analysis (GSEA), we found 5 glycolysis-related gene sets that were significantly enriched in LUSC tissues. Univariate and multivariate Cox proportional regression models were performed to choose prognostic-related gene signatures. Based on a Cox proportional regression model, a risk score for a three-gene signature (HKDC1, ALDH7A1, and MDH1) was established to divide patients into high-risk and low-risk subgroups. Multivariate Cox regression analysis indicated that the risk score for this three-gene signature can be used as an independent prognostic indicator in LUSC. Additionally, based on the cBioPortal database, the rate of genomic alterations in the HKDC1, ALDH7A1, and MDH1 genes were 1.9, 1.1, and 5% in LUSC patients, respectively. CONCLUSION A glycolysis-based three-gene signature could serve as a novel biomarker in predicting the prognosis of patients with LUSC and it also provides additional gene targets that can be used to cure LUSC patients.
Collapse
Affiliation(s)
- Guichuan Huang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ling Gong
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Yi Huang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China
| | - Daishun Liu
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), No 98 Fenghuang Road, Huichuan District, Zunyi, 563000, China.
| |
Collapse
|
8
|
Xu Z, Zhang S, Nian F, Xu S. Identification of a glycolysis-related gene signature associated with clinical outcome for patients with lung squamous cell carcinoma. Cancer Med 2021; 10:4017-4029. [PMID: 33991070 PMCID: PMC8209576 DOI: 10.1002/cam4.3945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022] Open
Abstract
Background Lung squamous cell carcinoma (LUSC), one of the main types of lung cancer, has caused a huge social burden. There has been no significant progress in its therapy in recent years, Resulting in a poor prognosis. This study aims to develop a glycolysis‐related gene signature to predict patients’ survival with LUSC and explore new therapeutic targets. Methods We obtained the mRNA expression and clinical information of 550 patients with LUSC from the Cancer Genome Atlas (TCGA) database. Glycolysis genes were identified by Gene Set Enrichment Analysis (GSEA). The glycolysis‐related gene signature was established using the Cox regression analysis. Results We developed five glycolysis‐related genes signature (HKDC1, AGL, ALDH7A1, SLC16A3, and MIOX) to calculate each patient's risk score. According to the risk score, patients were divided into high‐ and low‐risk groups and exhibited significant differences in overall survival (OS) between the two groups. The ROC curves showed that the AUC was 0.707 for the training cohort and 0.651 for the validation cohort. Additionally, the risk score was confirmed as an independent risk factor for LUSC patients by Cox regression analysis. Conclusion We built a gene signature to clarify the connection between glycolysis and LUSC. This model performs well in evaluating patients’ survival with LUSC and provides new biomarkers for targeted therapy.
Collapse
Affiliation(s)
- Ziming Xu
- Department of Thoracic Surgery, Wuxi 9th People's Hospital affiliated to Soochow University, Wuxi, Jiangsu, China.,Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiwei Zhang
- Department of Thoracic Surgery, Wuxi 9th People's Hospital affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Fulai Nian
- Department of Thoracic Surgery, Wuxi 9th People's Hospital affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Shangyu Xu
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
9
|
Zengin T, Önal-Süzek T. Comprehensive Profiling of Genomic and Transcriptomic Differences between Risk Groups of Lung Adenocarcinoma and Lung Squamous Cell Carcinoma. J Pers Med 2021; 11:154. [PMID: 33672117 PMCID: PMC7926392 DOI: 10.3390/jpm11020154] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the second most frequently diagnosed cancer type and responsible for the highest number of cancer deaths worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are subtypes of non-small-cell lung cancer which has the highest frequency of lung cancer cases. We aimed to analyze genomic and transcriptomic variations including simple nucleotide variations (SNVs), copy number variations (CNVs) and differential expressed genes (DEGs) in order to find key genes and pathways for diagnostic and prognostic prediction for lung adenocarcinoma and lung squamous cell carcinoma. We performed a univariate Cox model and then lasso-regularized Cox model with leave-one-out cross-validation using The Cancer Genome Atlas (TCGA) gene expression data in tumor samples. We generated 35- and 33-gene signatures for prognostic risk prediction based on the overall survival time of the patients with LUAD and LUSC, respectively. When we clustered patients into high- and low-risk groups, the survival analysis showed highly significant results with high prediction power for both training and test datasets. Then, we characterized the differences including significant SNVs, CNVs, DEGs, active subnetworks, and the pathways. We described the results for the risk groups and cancer subtypes separately to identify specific genomic alterations between both high-risk groups and cancer subtypes. Both LUAD and LUSC high-risk groups have more downregulated immune pathways and upregulated metabolic pathways. On the other hand, low-risk groups have both up- and downregulated genes on cancer-related pathways. Both LUAD and LUSC have important gene alterations such as CDKN2A and CDKN2B deletions with different frequencies. SOX2 amplification occurs in LUSC and PSMD4 amplification in LUAD. EGFR and KRAS mutations are mutually exclusive in LUAD samples. EGFR, MGA, SMARCA4, ATM, RBM10, and KDM5C genes are mutated only in LUAD but not in LUSC. CDKN2A, PTEN, and HRAS genes are mutated only in LUSC samples. The low-risk groups of both LUAD and LUSC tend to have a higher number of SNVs, CNVs, and DEGs. The signature genes and altered genes have the potential to be used as diagnostic and prognostic biomarkers for personalized oncology.
Collapse
Affiliation(s)
- Talip Zengin
- Department of Molecular Biology and Genetics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey;
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| | - Tuğba Önal-Süzek
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
- Department of Computer Engineering, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| |
Collapse
|
10
|
Zhang M, Sun L, Ru Y, Zhang S, Miao J, Guo P, Lv J, Guo F, Liu B. A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma. Int J Mol Med 2020; 46:252-264. [PMID: 32377703 PMCID: PMC7255475 DOI: 10.3892/ijmm.2020.4590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 01/30/2020] [Indexed: 12/20/2022] Open
Abstract
Lung squamous cell carcinoma (LSCC) is one of the primary types of non-small cell lung carcinoma, and patients with recurrent LSCC usually have a poor prognosis. The present study was conducted to build a risk score (RS) system for LSCC. Methylation data on LSCC (training set) and on head and neck squamous cell carcinoma (validation set 2) were obtained from The Cancer Genome Atlas database, and GSE39279 (validation set 1) was retrieved from the Gene Expression Omnibus database. Differentially methylated protein-coding genes (DMGs)/long non-coding RNAs (DM-lncRNAs) between recurrence-associated samples and nonrecurrence samples were screened out using the limma package, and their correlation analysis was conducted using the cor.test() function. Following identification of the optimal combinations of DMGs or DM-lncRNAs using the penalized package in R, RS systems were built, and the system with optimal performance was selected. Using the rms package, a nomogram survival model was then constructed. For the differentially expressed genes (DEGs) between the high- and low-risk groups, pathway enrichment analysis was performed by Gene Set Enrichment Analysis. There were 335 DMGs and DM-lncRNAs in total. Following screening out of the top 10 genes (aldehyde dehydrogenase 7 family member A1, chromosome 8 open reading frame 48, cytokine-like 1, heat shock protein 90 alpha family class A member 1, isovaleryl-CoA dehydrogenase, phosphodiesterase 3A, PNMA family member 2, SAM domain, SH3 domain and nuclear localization signals 1, thyroid hormone receptor interactor 13 and zinc finger protein 878) and 6 top lncRNAs, RS systems were constructed. According to Kaplan-Meier analysis, the DNA methylation level-based RS system exhibited the best performance. In combination with independent clinical prognostic factors, a nomogram survival model was built and successfully predicted patient survival. Furthermore, 820 DEGs between the high- and low-risk groups were identified, and 3 pathways were identified to be enriched in this gene set. The 10-DMG methylation level-based RS system and the nomogram survival model may be applied for predicting the outcomes of patients with LSCC.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Libing Sun
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Yi Ru
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Shasha Zhang
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Junjun Miao
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Pengda Guo
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jinghuan Lv
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Feng Guo
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Biao Liu
- Department of Oncology, The Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
11
|
Stoll G, Kremer M, Bloy N, Joseph A, Castedo M, Meurice G, Klein C, Galluzzi L, Michels J, Kroemer G. Metabolic enzymes expressed by cancer cells impact the immune infiltrate. Oncoimmunology 2019; 8:e1571389. [PMID: 31069148 DOI: 10.1080/2162402x.2019.1571389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
The expression of two metabolic enzymes, i.e., aldehyde dehydrogenase 7 family, member A1 (ALDH7A1) and lipase C, hepatic type (LIPC) by malignant cells, has been measured by immunohistochemical methods in non-small cell lung carcinoma (NSCLC) biopsies, and has been attributed negative and positive prognostic value, respectively. Here, we demonstrate that the protein levels of ALDH7A1 and LIPC correlate with the levels of the corresponding mRNAs. Bioinformatic analyses of gene expression data from 4921 cancer patients revealed that the expression of LIPC positively correlates with abundant tumor infiltration by myeloid and lymphoid cells in NSCLC, breast carcinoma, colorectal cancer and melanoma samples. In contrast, high levels of ALDH7A1 were associated with a paucity of immune effectors within the tumor bed. These data reinforce the notion that the metabolism of cancer cells has a major impact on immune and inflammatory processes in the tumor microenvironment, pointing to hitherto unsuspected intersections between oncometabolism and immunometabolism.
Collapse
Affiliation(s)
- Gautier Stoll
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Margerie Kremer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Normal Bloy
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Adrien Joseph
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Castedo
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guillaume Meurice
- Bioinformatics Core Facility, Gustave Roussy Cancer Campus, Villejuif, France
| | - Christophe Klein
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Centre d'Histologie, Imagerie cellulaire et Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Judith Michels
- Department of Medical Oncology, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Xie X, Zhao J, Xie L, Wang H, Xiao Y, She Y, Ma L. Identification of differentially expressed proteins in the injured lung from zinc chloride smoke inhalation based on proteomics analysis. Respir Res 2019; 20:36. [PMID: 30770755 PMCID: PMC6377712 DOI: 10.1186/s12931-019-0995-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Lung injury due to zinc chloride smoke inhalation is very common in military personnel and leads to a high incidence of pulmonary complications and mortality. The aim of this study was to uncover the underlying mechanisms of lung injury due to zinc chloride smoke inhalation using a rat model. Methods: Histopathology analysis of rat lungs after zinc chloride smoke inhalation was performed by using haematoxylin and eosin (H&E) and Mallory staining. A lung injury rat model of zinc chloride smoke inhalation (smoke inhalation for 1, 2, 7 and 14 days) was developed. First, isobaric tags for relative and absolute quantization (iTRAQ) and weighted gene co-expression network analysis (WGCNA) were used to identify important differentially expressed proteins. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were used to study the biological functions of differentially expressed proteins. Then, analysis of lung injury repair-related differentially expressed proteins in the early (day 1 and day 2) and middle-late stages (day 7 and day 14) of lung injury after smoke inhalation was performed, followed by the protein-protein interaction (PPI) analysis of these differentially expressed proteins. Finally, the injury repair-related proteins PARK7 and FABP5 were validated by immunohistochemistry and western blot analysis. Results Morphological changes were observed in the lung tissues after zinc chloride smoke inhalation. A total of 27 common differentially expressed proteins were obtained on days 1, 2, 7 and 14 after smoke inhalation. WGCNA showed that the turquoise module (which involved 909 proteins) was most associated with smoke inhalation time. Myl3, Ckm, Adrm1 and Igfbp7 were identified in the early stages of lung injury repair. Gapdh, Acly, Tnni2, Acta1, Actn3, Pygm, Eno3 and Tpi1 (hub proteins in the PPI network) were identified in the middle-late stages of lung injury repair. Eno3 and Tpi1 were both involved in the glycolysis/gluconeogenesis signalling pathway. The expression of PARK7 and FABP5 was validated and was consistent with the proteomics analysis. Conclusion The identified hub proteins and their related signalling pathways may play crucial roles in lung injury repair due to zinc chloride smoke inhalation.
Collapse
Affiliation(s)
- Xiaowei Xie
- Medical School of Chinese PLA, Medical School of Chinese PLA, Fuxing Road, Beijing, 100853, China
| | - Jingan Zhao
- Medical School of Chinese PLA, Medical School of Chinese PLA, Fuxing Road, Beijing, 100853, China
| | - Lixin Xie
- Medical School of Chinese PLA, Medical School of Chinese PLA, Fuxing Road, Beijing, 100853, China.
| | - Haiyan Wang
- Department of Respiratory, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Xiao
- Department of Respiratory, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yingjia She
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - Lingyun Ma
- Department of Respiratory, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
Andrejeva D, Kugler JM, Nguyen HT, Malmendal A, Holm ML, Toft BG, Loya AC, Cohen SM. Metabolic control of PPAR activity by aldehyde dehydrogenase regulates invasive cell behavior and predicts survival in hepatocellular and renal clear cell carcinoma. BMC Cancer 2018; 18:1180. [PMID: 30486822 PMCID: PMC6264057 DOI: 10.1186/s12885-018-5061-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
Background Changes in cellular metabolism are now recognized as potential drivers of cancer development, rather than as secondary consequences of disease. Here, we explore the mechanism by which metabolic changes dependent on aldehyde dehydrogenase impact cancer development. Methods ALDH7A1 was identified as a potential cancer gene using a Drosophila in vivo metastasis model. The role of the human ortholog was examined using RNA interference in cell-based assays of cell migration and invasion. 1H-NMR metabolite profiling was used to identify metabolic changes in ALDH7A1-depleted cells. Publically available cancer gene expression data was interrogated to identify a gene-expression signature associated with depletion of ALDH7A1. Computational pathway and gene set enrichment analysis was used to identify signaling pathways and cellular processes that were correlated with reduced ALDH7A1 expression in cancer. A variety of statistical tests used to evaluate these analyses are described in detail in the methods section. Immunohistochemistry was used to assess ALDH7A1 expression in tissue samples from cancer patients. Results Depletion of ALDH7A1 increased cellular migration and invasiveness in vitro. Depletion of ALDH7A1 led to reduced levels of metabolites identified as ligands for Peroxisome proliferator-activated receptor (PPARα). Analysis of publically available cancer gene expression data revealed that ALDH7A1 mRNA levels were reduced in many human cancers, and that this correlated with poor survival in kidney and liver cancer patients. Using pathway and gene set enrichment analysis, we establish a correlation between low ALDH7A1 levels, reduced PPAR signaling and reduced patient survival. Metabolic profiling showed that endogenous PPARα ligands were reduced in ALDH7A1-depleted cells. ALDH7A1-depletion led to reduced PPAR transcriptional activity. Treatment with a PPARα agonist restored normal cellular behavior. Low ALDH7A1 protein levels correlated with poor clinical outcome in hepatocellular and renal clear cell carcinoma patients. Conclusions We provide evidence that low ALDH7A1 expression is a useful prognostic marker of poor clinical outcome for hepatocellular and renal clear cell carcinomas and hypothesize that patients with low ALDH7A1 might benefit from therapeutic approaches addressing PPARα activity. Electronic supplementary material The online version of this article (10.1186/s12885-018-5061-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana Andrejeva
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Jan-Michael Kugler
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark.
| | - Hung Thanh Nguyen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Anders Malmendal
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Mette Lind Holm
- Department of Urology, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark
| | | | - Anand C Loya
- Department of Pathology, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark
| | - Stephen M Cohen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
14
|
Milone MR, Lombardi R, Roca MS, Bruzzese F, Addi L, Pucci B, Budillon A. Novel pathways involved in cisplatin resistance identified by a proteomics approach in non‐small‐cell lung cancer cells. J Cell Physiol 2018; 234:9077-9092. [DOI: 10.1002/jcp.27585] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/18/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Maria Rita Milone
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Rita Lombardi
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Laura Addi
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Biagio Pucci
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS‐Fondazione G. Pascale Napoli Italy
| |
Collapse
|
15
|
Duong HQ, You KS, Oh S, Kwak SJ, Seong YS. Silencing of NRF2 Reduces the Expression of ALDH1A1 and ALDH3A1 and Sensitizes to 5-FU in Pancreatic Cancer Cells. Antioxidants (Basel) 2017; 6:antiox6030052. [PMID: 28671577 PMCID: PMC5618080 DOI: 10.3390/antiox6030052] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/15/2017] [Accepted: 06/28/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer remains an intractable cancer with a poor five-year survival rate, which requires new therapeutic modalities based on the biology of pancreatic oncogenesis. Nuclear factor E2 related factor-2 (NRF2), a key cytoprotective nuclear transcription factor, regulates antioxidant production, reduction, detoxification and drug efflux proteins. It also plays an essential role in cell homeostasis, cell proliferation and resistance to chemotherapy. We aimed to evaluate the possibility that modulation of NRF2 expression could be effective in the treatment of pancreatic cancer cells. We investigated whether the depletion of NRF2 by using small interfering RNAs (siRNAs) is effective in the expression of biomarkers of pancreatic cancer stemness such as aldehyde dehydrogenase 1 family, member A1 (ALDH1A1) and aldehyde dehydrogenase 3 family, member A1 (ALDH3A1). NRF2 knockdown markedly reduced the expression of NRF2 and glutamate-cysteine ligase catalytic subunit (GCLC) in cell lines established from pancreatic cancers. NRF2 silencing also decreased the ALDH1A1 and ALDH3A1 expression. Furthermore, this NRF2 depletion enhanced the antiproliferative effects of the chemotherapeutic agent, 5-fluorouracil (5-FU) in pancreatic cancer cells.
Collapse
Affiliation(s)
- Hong-Quan Duong
- Department of Cancer Research, Vinmec Research Institute of Stem Cell and Gene Technology, 458 Minh Khai, Hanoi 10000, Vietnam.
- Institute of Research and Development, Duy Tan University, K7/25 Quang Trung, Danang 59000, Vietnam.
- Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.
| | - Kyu Sic You
- Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Korea.
| | - Seunghoon Oh
- Department of Physiology, College of Medicine, Dankook University, Cheonan 31116, Korea.
| | - Sahng-June Kwak
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Korea.
| | - Yeon-Sun Seong
- Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Korea.
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Korea.
| |
Collapse
|
16
|
Rodriguez-Torres M, Allan AL. Aldehyde dehydrogenase as a marker and functional mediator of metastasis in solid tumors. Clin Exp Metastasis 2015; 33:97-113. [PMID: 26445849 PMCID: PMC4740561 DOI: 10.1007/s10585-015-9755-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence indicating that aldehyde dehydrogenase (ALDH) activity selects for cancer cells with increased aggressiveness, capacity for sustained proliferation, and plasticity in primary tumors. However, emerging data also suggests an important mechanistic role for the ALDH family of isoenzymes in the metastatic activity of tumor cells. Recent studies indicate that ALDH correlates with either increased or decreased metastatic capacity in a cellular context-dependent manner. Importantly, it appears that different ALDH isoforms support increased metastatic capacity in different tumor types. This review assesses the potential of ALDH as biological marker and mechanistic mediator of metastasis in solid tumors. In many malignancies, most notably in breast cancer, ALDH activity and expression appears to be a promising marker and potential therapeutic target for treating metastasis in the clinical setting.
Collapse
Affiliation(s)
- Mauricio Rodriguez-Torres
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada. .,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada. .,London Regional Cancer Program, Room A4-132, 790 Commissioners Road East, London, ON, N6A 4L6, Canada.
| |
Collapse
|
17
|
Abstract
![]()
Aldehyde dehydrogenase 7A1 (ALDH7A1)
is part of lysine catabolism
and catalyzes the NAD+-dependent oxidation of α-aminoadipate
semialdehyde to α-aminoadipate. Herein, we describe a structural
study of human ALDH7A1 focused on substrate recognition. Five crystal
structures and small-angle X-ray scattering data are reported, including
the first crystal structure of any ALDH7 family member complexed with
α-aminoadipate. The product binds with the ε-carboxylate
in the oxyanion hole, the aliphatic chain packed into an aromatic
box, and the distal end of the product anchored by electrostatic interactions
with five conserved residues. This binding mode resembles that of
glutamate bound to the proline catabolic enzyme ALDH4A1. Analysis
of ALDH7A1 and ALDH4A1 structures suggests key interactions that underlie
substrate discrimination. Structures of apo ALDH7A1 reveal dramatic
conformational differences from the product complex. Product binding
is associated with a 16 Å movement of the C-terminus into the
active site, which stabilizes the active conformation of the aldehyde
substrate anchor loop. The fact that the C-terminus is part of the
active site was hitherto unknown. Interestingly, the C-terminus and
aldehyde anchor loop are disordered in a new tetragonal crystal form
of the apoenzyme, implying that these parts of the enzyme are highly
flexible. Our results suggest that the active site of ALDH7A1 is disassembled
when the aldehyde site is vacant, and the C-terminus is a mobile element
that forms quaternary structural interactions that aid aldehyde binding.
These results are relevant to the c.1512delG genetic deletion associated
with pyridoxine-dependent epilepsy, which alters the C-terminus of
ALDH7A1.
Collapse
Affiliation(s)
- Min Luo
- Department of Chemistry, University of Missouri-Columbia , Columbia, Missouri 65211, United States
| | - John J Tanner
- Department of Chemistry, University of Missouri-Columbia , Columbia, Missouri 65211, United States.,Department of Biochemistry, University of Missouri-Columbia , Columbia, Missouri 65211, United States
| |
Collapse
|
18
|
Luo M, Gates KS, Henzl MT, Tanner JJ. Diethylaminobenzaldehyde is a covalent, irreversible inactivator of ALDH7A1. ACS Chem Biol 2015; 10:693-7. [PMID: 25554827 DOI: 10.1021/cb500977q] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
There is growing interest in aldehyde dehydrogenases (ALDHs) because of their overexpression in cancer stem cells and the ability to mediate resistance to cancer drugs. Here, we report the first crystal structure of an aldehyde dehydrogenase complexed with the inhibitor 4-diethylaminobenzaldehyde (DEAB). Contrary to the widely held belief that DEAB is a reversible inhibitor of ALDHs, we show that DEAB irreversibly inactivates ALDH7A1 via formation of a stable, covalent acyl-enzyme species.
Collapse
Affiliation(s)
- Min Luo
- Department
of Chemistry, University of Missouri−Columbia, Columbia, Missouri 65211, United States
| | - Kent S. Gates
- Department
of Chemistry, University of Missouri−Columbia, Columbia, Missouri 65211, United States
| | - Michael T. Henzl
- Department
of Biochemistry, University of Missouri−Columbia, Columbia, Missouri 65211, United States
| | - John J. Tanner
- Department
of Chemistry, University of Missouri−Columbia, Columbia, Missouri 65211, United States
- Department
of Biochemistry, University of Missouri−Columbia, Columbia, Missouri 65211, United States
| |
Collapse
|
19
|
Rosi A, Ricci-Vitiani L, Biffoni M, Grande S, Luciani AM, Palma A, Runci D, Cappellari M, De Maria R, Guidoni L, Pallini R, Viti V. (1) H NMR spectroscopy of glioblastoma stem-like cells identifies alpha-aminoadipate as a marker of tumor aggressiveness. NMR IN BIOMEDICINE 2015; 28:317-26. [PMID: 25581615 DOI: 10.1002/nbm.3254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 11/12/2014] [Accepted: 12/03/2014] [Indexed: 05/24/2023]
Abstract
Patients suffering from glioblastoma multiforme (GBM) face a poor prognosis with median survival of about 14 months. High recurrence rate and failure of conventional treatments are attributed to the presence of GBM cells with stem-like properties (GSCs). Metabolite profiles of 42 GSC lines established from the tumor tissue of adult GBM patients were screened with (1) H NMR spectroscopy and compared with human neural progenitor cells from human adult olfactory bulb (OB-NPCs) and from the developing human brain (HNPCs). A first subset (n=12) of GSCs exhibited a dramatic accumulation of the metabolite α-aminoadipate (αAAD), product of the oxidation of α-aminoadipic semialdehyde catalyzed by the ALDH7A1 aldehyde dehydrogenase (ALDH) family in lysine catabolism. αAAD was low/not detectable in a second GSC subset (n=13) with the same neural metabolic profile as well as in a third GSC subset (n=17) characterized by intense lipid signals. Likewise, αAAD was not detected in the spectra of OB-NPCs or HNPCs. Inhibition of mitochondrial ATP synthase by oligomycin treatment revealed that the lysine degradative pathway leading to αAAD formation proceeds through saccharopine, as usually observed in developing brain. Survival curves indicated that high αAAD levels in GSCs significantly correlated with poor patient survival, similarly to prostate and non-small-cell-lung cancers, where activity of ALDH7A1 correlates with tumor aggressiveness.
Collapse
Affiliation(s)
- Antonella Rosi
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Pors K, Moreb JS. Aldehyde dehydrogenases in cancer: an opportunity for biomarker and drug development? Drug Discov Today 2014; 19:1953-63. [PMID: 25256776 DOI: 10.1016/j.drudis.2014.09.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/31/2014] [Accepted: 09/15/2014] [Indexed: 02/07/2023]
Abstract
Aldehyde dehydrogenases (ALDHs) belong to a superfamily of 19 isozymes that are known to participate in many physiologically important biosynthetic processes including detoxification of specific endogenous and exogenous aldehyde substrates. The high expression levels of an emerging number of ALDHs in various cancer tissues suggest that these enzymes have pivotal roles in cancer cell survival and progression. Mapping out the heterogeneity of tumours and their cancer stem cell (CSC) component will be key to successful design of strategies involving therapeutics that are targeted against specific ALDH isozymes. This review summarises recent progress in ALDH-focused cancer research and discovery of small-molecule-based inhibitors.
Collapse
Affiliation(s)
- Klaus Pors
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| | - Jan S Moreb
- Hematological Malignancies, PO Box 100278, Gainesville, FL 32610-0277, USA.
| |
Collapse
|
21
|
Wang H, Tong L, Wei J, Pan W, Li L, Ge Y, Zhou L, Yuan Q, Zhou C, Yang M. The ALDH7A1 genetic polymorphisms contribute to development of esophageal squamous cell carcinoma. Tumour Biol 2014; 35:12665-70. [PMID: 25213698 DOI: 10.1007/s13277-014-2590-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/03/2014] [Indexed: 01/10/2023] Open
Abstract
Although the entire etiology of esophageal squamous cell carcinoma (ESCC) is still unclear, alcohol drinking has been identified as a major environmental risk factor. The aldehyde dehydrogenase (ALDH) superfamily members are major enzymes involved in the alcohol-metabolizing pathways. Accumulating evidences demonstrated that ALDH7A1, one of ALDH superfamily members, degrades and detoxifies acetaldehyde generated by alcohol metabolism and have been associated with development and prognosis of multiple cancers. However, it is still unknown if ALDH7A1 single nucleotide polymorphisms (SNPs) contribute to ESCC susceptibility. In this study, we examined the association between sixteen ALDH7A1 SNPs and risk of developing ESCC. Genotypes were determined in 2,098 ESCC patients and 2,150 controls (three independent hospital-based case-control sets from different regions of China). Odds ratios (ORs) and 95 % confidence intervals (CIs) were estimated by logistic regression. Our data demonstrated that only the ALDH7A1 rs13182402 SNP confer susceptibility to ESCC (For AG genotype, OR = 0.75, 95 % CI = 0.66-0.91, P = 4.8 × 10(-6); for GG genotype, OR = 0.59, 95 % CI = 0.41-0.88, P = 0.003). These results are consistent to the biological functions of ALDH7A1 during alcohol metabolism and carcinogenesis.
Collapse
Affiliation(s)
- Haiyong Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, P. O. Box 53, Beijing, 100029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Large-scale analysis of posttranslational modifications in the hippocampus of patients with Alzheimer’s disease using pI shift and label-free quantification without enrichment. Anal Bioanal Chem 2014; 406:5433-46. [DOI: 10.1007/s00216-014-7933-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 01/10/2023]
|
23
|
Arasada RR, Amann JM, Rahman MA, Huppert SS, Carbone DP. EGFR blockade enriches for lung cancer stem-like cells through Notch3-dependent signaling. Cancer Res 2014; 74:5572-84. [PMID: 25125655 DOI: 10.1158/0008-5472.can-13-3724] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the epidermal growth factor receptor (EGFR) are the most common actionable genetic abnormalities yet discovered in lung cancer. However, targeting these mutations with kinase inhibitors is not curative in advanced disease and has yet to demonstrate an impact on potentially curable, early-stage disease, with some data suggesting adverse outcomes. Here, we report that treatment of EGFR-mutated lung cancer cell lines with erlotinib, while showing robust cell death, enriches the ALDH(+) stem-like cells through EGFR-dependent activation of Notch3. In addition, we demonstrate that erlotinib treatment increases the clonogenicity of lung cancer cells in a sphere-forming assay, suggesting increased stem-like cell potential. We demonstrate that inhibition of EGFR kinase activity leads to activation of Notch transcriptional targets in a γ secretase inhibitor-sensitive manner and causes Notch activation, leading to an increase in ALDH high(+) cells. We also find a kinase-dependent physical association between the Notch3 and EGFR receptors and tyrosine phosphorylation of Notch3. This could explain the worsened survival observed in some studies of erlotinib treatment at early-stage disease, and suggests that specific dual targeting might overcome this adverse effect.
Collapse
Affiliation(s)
- Rajeswara Rao Arasada
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Joseph M Amann
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Mohammad A Rahman
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Center for Critical Care Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Stacey S Huppert
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio.
| | - David P Carbone
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio.
| |
Collapse
|
24
|
The effect of several intertrial intervals on the 1 Hz interference effect. Can J Neurol Sci 1981; 12:cancers12040961. [PMID: 32295073 PMCID: PMC7225959 DOI: 10.3390/cancers12040961] [Citation(s) in RCA: 40] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/24/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022]
Abstract
Gynecologic cancers cause over 600,000 deaths annually in women worldwide. The development of chemoresistance after initial rounds of chemotherapy contributes to tumor relapse and death due to gynecologic malignancies. In this regard, cancer stem cells (CSCs), a subpopulation of stem cells with the ability to undergo self-renewal and clonal evolution, play a key role in tumor progression and drug resistance. Aldehyde dehydrogenases (ALDH) are a group of enzymes shown to be robust CSC markers in gynecologic and other malignancies. These enzymes also play functional roles in CSCs, including detoxification of aldehydes, scavenging of reactive oxygen species (ROS), and retinoic acid (RA) signaling, making ALDH an attractive therapeutic target in various clinical scenarios. In this review, we discuss the critical roles of the ALDH in driving stemness in different gynecologic malignancies. We review inhibitors of ALDH, both general and isoform-specific, which have been used to target CSCs in gynecologic cancers. Many of these inhibitors have been shown to be effective in preclinical models of gynecologic malignancies, supporting further development in the clinic. Furthermore, ALDH inhibitors, including 673A and CM037, synergize with chemotherapy to reduce tumor growth. Thus, ALDH-targeted therapies hold promise for improving patient outcomes in gynecologic malignancies.
Collapse
|