1
|
Wang G, Liu S, Kong X, Jiao H, Tong F, Guo Z, Zhang M, Guan X, Ren N, Li W, Qi L, Wei Y. Lipocalin-2 induced LDHA expression promotes vascular remodelling in pulmonary hypertension. Cell Prolif 2024; 57:e13717. [PMID: 39021353 PMCID: PMC11628741 DOI: 10.1111/cpr.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
Aerobic glycolysis is involved in the pathogenesis of pulmonary hypertension (PH). The mechanisms by which glycolysis is increased and how it contributes to pulmonary vascular remodelling are not yet fully understood. In this study, we demonstrated that elevated lipocalin-2 (LCN2) in PH significantly enhances aerobic glycolysis in human pulmonary artery smooth muscle cells (PASMCs) by up-regulating LDHA expression. Knockout of Lcn2 or having heterozygous LDHA deficiency in mice significantly inhibits the progression of hypoxic PH. Our study reveals that LCN2 stimulates LDHA expression by activating Akt-HIF-1α signalling pathway. Inhibition of Akt or HIF-1α reduces LDHA expression and proliferation of PASMCs. Both Akt and HIF-1α play critical roles in the development of PH and are suppressed in the pulmonary vessels of hypoxic PH mice lacking LCN2. These findings shed light on the LCN2-Akt-HIF1α-LDHA axis in aerobic glycolysis in PH.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaohui Kong
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Hong Jiao
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Feng Tong
- Department of Cardiac Surgery, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Zhangke Guo
- Department of Cardiac Surgery, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Meng Zhang
- Department of Pathology, Beijing Children's Hospital, Capital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Xiaoxing Guan
- Department of Pathology, Beijing Children's Hospital, Capital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Na Ren
- Department of Clinical Laboratory Center, Beijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Wanzhen Li
- Department of Lipidomics Experimental Platform, State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental BiologyChinese Academy of SciencesBeijingChina
| | - Lihua Qi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa-SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [DOI: https:/doi.org/10.3390/biomedicines12020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3−3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
3
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa- SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [PMID: 38397997 PMCID: PMC11154313 DOI: 10.3390/biomedicines12020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 02/25/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3-3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico;
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico;
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
4
|
Zhang HH, Zhang AQ, Peng P, Huang L, Liu CY, Nie XR, Hou DF, Zhang X, Li SZ. USP5 facilitates bladder cancer progression by stabilizing the c-Jun protein. Cancer Cell Int 2024; 24:32. [PMID: 38229092 DOI: 10.1186/s12935-024-03222-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Bladder cancer is the second most common genitourinary malignancy worldwide. The death rate of bladder cancer has increased every year. However, the molecular mechanism of bladder cancer is not sufficiently studied. Deubiquitinating enzymes (DUBs) play an important role in carcinogenesis. Several studies have demonstrated that USP5 associated with malignancy and pathological progression in hepatocellular carcinoma, colorectal and non-small cell lung cancer. However, the role of USP5 in bladder cancer need to be explored. METHODS The USP5 expression was analysed using the web server GEPIA. To explore USP5 function in bladder cancer, we constructed USP5-knockout cell lines in T24 cells. A FLAG-USP5 (WT USP5) plasmid and a plasmid FLAG-USP5 C335A (catalytic-inactive mutant) used to overexpress USP5 in EJ cells. CCK8, colony formation, transwell and scratch assays were used to assess cell viability, proliferation and migration. RNA sequencing (RNA-seq) and dual-luciferase reporter assays were performed to screen the pathway. Coimmunoprecipitation and immunofluorescence were used to explore the interaction between USP5 and c-Jun. Cycloheximide (CHX) chase assays were performed to establish the effect of USP5 on c-Jun stability. Xenograft mouse model was used to study the role of USP5 in bladder cancer. RESULTS USP5 expression is increased in bladder cancer patients. Genetic ablation of USP5 markedly inhibited bladder cancer cell proliferation, viability, and migration both in vitro and in vivo. RNA-seq and luciferase pathway screening showed that USP5 activated JNK signalling, and we identified the interaction between USP5 and c-Jun. USP5 was found to activate c-Jun by inhibiting its ubiquitination. CONCLUSIONS Our results show that high USP5 expression promotes bladder cancer progression by stabilizing c-Jun and that USP5 is a potential therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Hui-Hui Zhang
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - An-Qi Zhang
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - Peng Peng
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - Liang Huang
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - Cai-Ying Liu
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - Xin-Rui Nie
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - De-Fu Hou
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China
| | - Xia Zhang
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China.
| | - Shang-Ze Li
- Department of Laboratory Medicine, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Yuelu District, Changsha, Hunan, China.
- School of Medicine, Chongqing University, 131 Yubei Road, Shapingba District, Chongqing, China.
| |
Collapse
|
5
|
Wang L, Cao Y, Guo W, Xu J. High expression of cuproptosis-related gene FDX1 in relation to good prognosis and immune cells infiltration in colon adenocarcinoma (COAD). J Cancer Res Clin Oncol 2023; 149:15-24. [PMID: 36173462 PMCID: PMC9889456 DOI: 10.1007/s00432-022-04382-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/24/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cuproptosis induced by FDX1 is a newly discovered mechanism regulating cell death. However, the role of FDX1 in the pathogenesis of colon adenocarcinoma (COAD) remains to be studied. METHODS FDX1 expression was analyzed with The Cancer Genome Atlas (TCGA) database and Human Protein Atlas (HPA) database. Association between FDX1 expression and COAD prognosis was investigated via the Kaplan-Meier (KM) survival curve. The differentially expressed genes (DEGs) of FDX1 were screened with R packages and the PPI were constructed via STRING database. Cytoscape software was used to detect the most profound modules in the PPIs network. CancerSEA database was used to analyze the effect of FDX1 expression levels on different functional status of COAD cells. The relationship between FDX1 expression and immune infiltration of COAD was analyzed by TIMER2.0 database. The COAD patients with high expression of FDX1 by Western blot, and the levels of immune infiltration were measured by flow cytometry. RESULTS FDX1 was low expressed in most cancers, such as BRCA, KICH, and COAD. The overall survival (OS) and disease-specific survival (DSS) of COAD with high FDX1 expression were better than that of the low expression group. GO-KEGG enrichment analysis revealed that FDX1 and its co-expressed genes played an important role in the pathogenesis of COAD. Moreover, FDX1 expression in COAD were positively associated with "quiescence" and "inflammation" but negatively correlated with "invasion". FDX1 expression was positively correlated with infiltration levels of CD8+ T cells, NK cells, and neutrophils. Oppositely, FDX1 expression was negatively correlated with that of CD4+ T cells and cancer-associated fibroblasts (CAFs). Finally, 6 COAD patients with high expression of FDX1 were screened, and the proportion of CD8+ T cells in cancer tissues of these patients was significantly higher than that in paracancerous, while the CD4+ T cells presented the opposite pattern. CONCLUSION FDX1 plays a role in inducing cuproptosis and modulating tumor immunity, which could be considered as potential therapeutic targets in COAD.
Collapse
Affiliation(s)
- Lizong Wang
- grid.452929.10000 0004 8513 0241General Practice Department, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province China
| | - Yi Cao
- grid.443626.10000 0004 1798 4069School of Basic Medicine, Wannan Medical College, NO. 22 Wenchang west road, Wuhu, Anhui Province China
| | - Wei Guo
- grid.443626.10000 0004 1798 4069School of Basic Medicine, Wannan Medical College, NO. 22 Wenchang west road, Wuhu, Anhui Province China
| | - Jingyun Xu
- School of Basic Medicine, Wannan Medical College, NO. 22 Wenchang west road, Wuhu, Anhui Province, China.
| |
Collapse
|
6
|
Non-kinase targeting of oncogenic c-Jun N-terminal kinase (JNK) signaling: the future of clinically viable cancer treatments. Biochem Soc Trans 2022; 50:1823-1836. [PMID: 36454622 PMCID: PMC9788565 DOI: 10.1042/bst20220808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 01/09/2023]
Abstract
c-Jun N-terminal Kinases (JNKs) have been identified as key disease drivers in a number of pathophysiological settings and central oncogenic signaling nodes in various cancers. Their roles in driving primary tumor growth, positively regulating cancer stem cell populations, promoting invasion and facilitating metastatic outgrowth have led JNKs to be considered attractive targets for anti-cancer therapies. However, the homeostatic, apoptotic and tumor-suppressive activities of JNK proteins limit the use of direct JNK inhibitors in a clinical setting. In this review, we will provide an overview of the different JNK targeting strategies developed to date, which include various ATP-competitive, non-kinase and substrate-competitive inhibitors. We aim to summarize their distinct mechanisms of action, review some of the insights they have provided regarding JNK-targeting in cancer, and outline the limitations as well as challenges of all strategies that target JNKs directly. Furthermore, we will highlight alternate drug targets within JNK signaling complexes, including recently identified scaffold proteins, and discuss how these findings may open up novel therapeutic options for targeting discrete oncogenic JNK signaling complexes in specific cancer settings.
Collapse
|