1
|
Li X, Zhang Y, Gong J, Liu W, Zhao H, Xue W, Ren Z, Bao J, Lin Z. Development of a breast cancer invasion score to predict tumor aggressiveness and prognosis via PI3K/AKT/mTOR pathway analysis. Cell Death Discov 2025; 11:157. [PMID: 40204712 PMCID: PMC11982538 DOI: 10.1038/s41420-025-02422-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
Invasiveness is a key indicator of tumor malignancy and is often linked to poor prognosis in breast cancer (BC). To explore the diverse characteristics of invasive cells, single-cell RNA sequencing (scRNA-seq) data from three ductal carcinoma stages were analyzed, classifying samples into invasion and non-invasion groups. Nine genes (MCTS1, PGK1, PCMT1, C8orf76, TMEM242, QPRT, SLC16A2, AFG1L, and SPINK8) were identified as key discriminators between these groups. A breast cancer invasion score (BCIS) model was developed using LASSO Cox regression, revealed that high BCIS correlated with poorer overall survival in TCGA-BRCA patients and was validated across GSE20685 and METABRIC datasets (five-year and ten-year survival). Functional experiments demonstrated that knockdown of PGK1 or PCMT1 inhibited tumor cell proliferation and reduced the phosphorylation levels of mTORC, P70S6K, S6, and AKT, indicating suppression of the PI3K/AKT/mTOR pathways. High-BCIS tumors exhibited enrichment in protein secretion and PI3K/AKT/mTOR pathways, associated with aggressiveness and therapy resistance. This study introduced the BCIS score, distinguishing invasion from non-invasion cells, linked to PI3K/AKT/mTOR pathways, offering insights into BRCA prognosis and tumor aggressiveness.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of General Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Ya Zhang
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianping Gong
- Department of General Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Wenjia Liu
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, 311199, China
| | - Hanchen Zhao
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, 311199, China
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Wei Xue
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, 311199, China
| | - Zhaojun Ren
- Department of Pathology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210000, China.
| | - Jun Bao
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210000, China.
| | - Ziao Lin
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, 311199, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311100, China.
| |
Collapse
|
2
|
Mirzaiebadizi A, Shafabakhsh R, Ahmadian MR. Modulating PAK1: Accessory Proteins as Promising Therapeutic Targets. Biomolecules 2025; 15:242. [PMID: 40001545 PMCID: PMC11852631 DOI: 10.3390/biom15020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The p21-activated kinase (PAK1), a serine/threonine protein kinase, is critical in regulating various cellular processes, including muscle contraction, neutrophil chemotaxis, neuronal polarization, and endothelial barrier function. Aberrant PAK1 activity has been implicated in the progression of several human diseases, including cancer, heart disease, and neurological disorders. Increased PAK1 expression is often associated with poor clinical prognosis, invasive tumor characteristics, and therapeutic resistance. Despite its importance, the cellular mechanisms that modulate PAK1 function remain poorly understood. Accessory proteins, essential for the precise assembly and temporal regulation of signaling pathways, offer unique advantages as therapeutic targets. Unlike core signaling components, these modulators can attenuate aberrant signaling without completely abolishing it, potentially restoring signaling to physiological levels. This review highlights PAK1 accessory proteins as promising and novel therapeutic targets, opening new horizons for disease treatment.
Collapse
Affiliation(s)
- Amin Mirzaiebadizi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Rana Shafabakhsh
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
3
|
Wang B, Zhou R, Wu J, Kim H, Kim K. Inhibition of δ-catenin palmitoylation slows the progression of prostate cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119741. [PMID: 38697304 DOI: 10.1016/j.bbamcr.2024.119741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of death in males. It has been reported that δ-catenin expression is upregulated during the late stage of prostate cancer. Palmitoylation promotes protein transport to the cytomembrane and regulates protein localization and function. However, the effect of δ-catenin palmitoylation on the regulation of cancer remains unknown. In this study, we utilized prostate cancer cells overexpressing mutant δ-catenin (J6A cells) to induce a depalmitoylation phenotype and investigate its effect on prostate cancer. Our results indicated that depalmitoylation of δ-catenin not only reduced its membrane expression but also promoted its degradation in the cytoplasm, resulting in a decrease in the effect of EGFR and E-cadherin signaling. Consequently, depalmitoylation of δ-catenin reduced the proliferation and metastasis of prostate cancer cells. Our findings provide novel insights into potential therapeutic strategies for controlling the progression of prostate cancer through palmitoylation-based targeting of δ-catenin.
Collapse
Affiliation(s)
- Beini Wang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea
| | - Jin Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Kwonseop Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
4
|
Chen M, Pan L, Chen D, Wu Y, Ye J, Li K, Zhang N, Xu J. PAK1 Promotes Inflammation Induced by Sepsis through the Snail/CXCL2 Signaling Pathway. ACS Infect Dis 2024; 10:1370-1378. [PMID: 38503263 DOI: 10.1021/acsinfecdis.4c00052] [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] [Indexed: 03/21/2024]
Abstract
Sepsis is a severe syndrome characterized by organ dysfunction, resulting from a systemic imbalance in response to infection. PAK1 plays a critical role in various diseases. The present study aimed to explore and delineate the mechanism of PAK1 in inflammation induced by sepsis. Bioinformatics analysis was performed to assess PAK1, snail, and CXCL2 expression in the whole blood of septic patients and the pathways enriched with PAK1. To simulate the sepsis model, THP-1 cells were stimulated with lipopolysaccharide. Gene expression was evaluated using qRT-PCR, while cell viability was assessed using CCK-8 assay. Cell apoptosis was tested with flow cytometry. Expression of inflammatory factors in cells following different treatments was analyzed using the enzyme linked immunosorbent assay (ELISA). Dual-luciferase and chromatin immunoprecipitation assays were conducted to verify the binding relationship between PAK1 and the snail. Mouse models of cecal ligation and puncture were established, and hematoxylin and eosin staining and ELISA were employed to detect the infiltration levels of inflammatory cells and the expression of related protective factors in lung, liver, and kidney tissues. The results demonstrated upregulation of PAK1, snail, and CXCL2 in the whole blood of septic patients, with PAK1 being enriched in the chemokine-related pathway. Knockdown of PAK1 significantly promoted the apoptosis of LPS-stimulated THP-1 cells and inhibited the expression of inflammatory factors. PAK1 upregulated the expression of the snail, which in turn promoted the expression of CXCL2. Thus, PAK1 mediated the sepsis-induced inflammatory response through the snail/CXCL2 pathway. In conclusion, PAK1 played a role in promoting inflammation induced by sepsis through the snail/CXCL2 axis, thereby providing a potential therapeutic target for the management of sepsis.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Liuhua Pan
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Deyuan Chen
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yueming Wu
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jianping Ye
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ke Li
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ning Zhang
- Department of Emergency Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Junlong Xu
- Department of Critical Care Medicine, The Six Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| |
Collapse
|
5
|
Tang Q, Wen H, Hu H, Chen X, Xu S, Fan L, Liu L, Li J. Circ_0070203 Promotes Epithelial-mesenchymal Transition in Ovarian Serous Cystadenocarcinoma through miR-370-3p/TGFβR2 Axis. Recent Pat Anticancer Drug Discov 2024; 19:233-246. [PMID: 38214360 DOI: 10.2174/1574892818666230328124804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Circular RNAs (circRNAs) are important biological molecules associated with the pathogenesis of multiple cancers. OBJECTIVE This work aimed to investigate the function and molecular mechanism of circ_0070203 in high-grade serous ovarian cystadenocarcinoma (HGSOC). METHODS circRNA microarray was conducted to detect the circ_0070203 expression in HGSOC tissues. Bioinformatics analysis was used to find the binding sites between circ_0070203, miR- 370-3p and TGFβR2. Real-time quantitative reverse transcription PCR (RT-qPCR) was executed to detect the expressions of circ_0070203, miR-370-3p and TGFβR2 in HGSOC tissues and SKOV3 cells. Dual-luciferase reporter gene assay was used to validate the relationships between miR-370-3p and circ_0070203 or TGFβR2. Besides, transwell assays were conducted to assess the migrative, invasive abilities of ovarian cancer (OC) cells. Western blotting was adopted to detect the expression of epithelial-mesenchymal transition (EMT)-related proteins. The related patents were also studied during the research. RESULTS Circ_0070203 and TGFβR2 were upregulated, while miR-370-3p was downregulated in FIGO stage III-IV HGSOC tissues and SKOV-3 cell lines. circ_0070203 overexpression changed the expression of other EMT-related proteins and enhanced the migrative, invasive abilities of OC cells, while silencing circ_0070203 worked oppositely. Mechanistically, circ_0070203 could upregulate TGFβR2 expression in OC cells via sponging miR-370-3p. CONCLUSION Circ_0070203 could promote the epithelial-mesenchymal transition, invasion, and metastasis of HGSOC via regulating the miR-370-3p/TGFβR2 axis. Our findings provided a potential biomarker for HGSOC therapy.
Collapse
Affiliation(s)
- Qiong Tang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Huiting Wen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Xiaoli Chen
- Department of Obstetrics, Southern Medical University Affiliated Maternal and Children's Hospital of Foshan, Foshan, 528000, Guangdong, People's Republic of China
| | - Shuxiu Xu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Li Fan
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Longyang Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Jing Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, People's Republic of China
| |
Collapse
|
6
|
Surabhi RP, Rajendran S, Srikanth Swamy Swaroop B, Murugan S, Shanmugasundaram G, Joseph LD, Pitani R, Babu PS, Suresh K R, Venkatraman G. Activation of oncogenic signaling kinase PAK1 by ionising radiation confers an aggressive phenotype in head and neck squamous cell carcinoma. Cell Signal 2023; 112:110910. [PMID: 37777103 DOI: 10.1016/j.cellsig.2023.110910] [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: 08/15/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Head and neck squamous cancers are very aggressive tumors often diagnosed in late stages with poor prognosis. HNSCCs are usually treated by a course of radiation (IR) therapy and followed by surgery. These treatment regimens fail to bring a complete response. Molecular signatures in tumors are attributed to this response and an improved understanding of the signaling events could offer new avenues for therapy. Here, we show that P21 activated kinase-1 (PAK1) - an oncogenic signaling serine/threonine kinase, is activated upon exposure to IR and this leads to an accelerated tumorigenic character in HNSCC cells. Our results show that PAK1 is highly expressed in HNSCC cell lines, as compared to normal buccal mucosa cells and when HNSCC cells were exposed to IR, they show activated PAK1 and an aggressive phenotype as determined by in vitro functional assays. PAK1 levels were elevated in HNSCC as compared to adjacent normal oral tissues and our results also show convincing evidence of activated PAK1 in patient tumor samples of post- IR treatment as compared to pre-IR treatment and is associated with poor survival. Pak1 Knockout (KO) clones in HNSCC cells showed that they were more sensitive to IR as compared to wild type (wt) cells. This altered sensitivity to IR was attributed to enhanced DNA damage response modulated by PAK1 in cells. Overall, our results suggest that PAK1 expression in HNSCC could be a critical determinant in IR therapy response and silencing PAK1 is likely to be a treatment modality to improve clinical outcomes.
Collapse
Affiliation(s)
- Rohan Prasad Surabhi
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Swetha Rajendran
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - B Srikanth Swamy Swaroop
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Sowmiya Murugan
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai 600036, India
| | - Gouthaman Shanmugasundaram
- Department of Surgical Oncology, Sri Ramachandra Medical College, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Leena Dennis Joseph
- Department of Pathology, Sri Ramachandra Medical College, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Ravishankar Pitani
- Department of Community Medicine, Sri Ramachandra Medical College, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Pakala Suresh Babu
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana 500046, India
| | - Rayala Suresh K
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai 600036, India.
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India.
| |
Collapse
|
7
|
Wang K, Qiu X, Zhang Z, Xu H, Tan Y, Su R, Gao F, Zhuo J, Li W, Lian Z, He H, Xu X. ATRA sensitized the response of hepatocellular carcinoma to Sorafenib by downregulation of p21-activated kinase 1. Cell Commun Signal 2023; 21:193. [PMID: 37537668 PMCID: PMC10399044 DOI: 10.1186/s12964-023-01194-1] [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: 02/19/2023] [Accepted: 06/10/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Sorafenib resistance greatly reduces the efficacy of treatments in advanced hepatocellular carcinoma (HCC) patients, but the underlying mechanisms are not thoroughly understood. All-trans retinoic acid (ATRA), an anti-leukaemia agent, has attracted considerable attention due to its role in sensitizing cells to other anticancer treatments. We aimed to investigate the combined effect of ATRA and Sorafenib on HCC and the underlying mechanisms. METHODS CCK-8, cell sphere formation, trans-well migration, and wound-healing assays were used to analyse the biological behaviours of HCC cells in vitro. Western blotting and qRT-PCR analysis were conducted to measure the expression of p21 activated kinase 1 (PAK1) and phospho-p21 activated kinase 1 (pPAK1). Xenograft models were established to confirm the synergistic effects of ATRA and Sorafenib in vivo. TUNEL assays and immunohistochemistry were utilized to determine apoptosis, proliferation, PAK1 and pPAK1 levels in tumour tissues. RESULTS We observed that PAK1 was overexpressed in HCC, and its expression was negatively correlated with the survival of patients. PAK1 promoted the proliferation, self-renewal and epithelial-mesenchymal transition of HCC cells. Correlation analysis indicated that the IC50 of Sorafenib was positively correlated with the level of pPAK1 in HCC cell lines. ATRA inhibited the progression of HCC and sensitized HCC response to Sorafenib by downregulation of PAK1, as shown by the calculated coefficient of drug interaction and the data obtained from xenograft models. CONCLUSIONS Our findings indicated that instead of treatment with Sorafenib alone, the combination of ATRA and Sorafenib provides a more effective treatment for HCC patients. Video Abstract.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, People's Republic of China
| | - Xun Qiu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Zhensheng Zhang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Hanzhi Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Yawen Tan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Renyi Su
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Fengqiang Gao
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China
| | - Jianyong Zhuo
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Wangyao Li
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Zhengxing Lian
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, VIC, 3084, Australia.
| | - Xiao Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, People's Republic of China.
- Zhejiang University School of Medicine, Hangzhou, 310058, People's Republic of China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, People's Republic of China.
| |
Collapse
|
8
|
Hammad S, Ogris C, Othman A, Erdoesi P, Schmidt-Heck W, Biermayer I, Helm B, Gao Y, Piorońska W, Holland CH, D'Alessandro LA, de la Torre C, Sticht C, Al Aoua S, Theis FJ, Bantel H, Ebert MP, Klingmüller U, Hengstler JG, Dooley S, Mueller NS. Tolerance of repeated toxic injuries of murine livers is associated with steatosis and inflammation. Cell Death Dis 2023; 14:414. [PMID: 37438332 DOI: 10.1038/s41419-023-05855-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 04/13/2023] [Accepted: 05/05/2023] [Indexed: 07/14/2023]
Abstract
The human liver has a remarkable capacity to regenerate and thus compensate over decades for fibrosis caused by toxic chemicals, drugs, alcohol, or malnutrition. To date, no protective mechanisms have been identified that help the liver tolerate these repeated injuries. In this study, we revealed dysregulation of lipid metabolism and mild inflammation as protective mechanisms by studying longitudinal multi-omic measurements of liver fibrosis induced by repeated CCl4 injections in mice (n = 45). Based on comprehensive proteomics, transcriptomics, blood- and tissue-level profiling, we uncovered three phases of early disease development-initiation, progression, and tolerance. Using novel multi-omic network analysis, we identified multi-level mechanisms that are significantly dysregulated in the injury-tolerant response. Public data analysis shows that these profiles are altered in human liver diseases, including fibrosis and early cirrhosis stages. Our findings mark the beginning of the tolerance phase as the critical switching point in liver response to repetitive toxic doses. After fostering extracellular matrix accumulation as an acute response, we observe a deposition of tiny lipid droplets in hepatocytes only in the Tolerant phase. Our comprehensive study shows that lipid metabolism and mild inflammation may serve as biomarkers and are putative functional requirements to resist further disease progression.
Collapse
Affiliation(s)
- Seddik Hammad
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt.
| | - Christoph Ogris
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Amnah Othman
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Pia Erdoesi
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Schmidt-Heck
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Ina Biermayer
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Barbara Helm
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Yan Gao
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weronika Piorońska
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian H Holland
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Lorenza A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Carolina de la Torre
- Core Facility Next Generation Sequencing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Core Facility Next Generation Sequencing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sherin Al Aoua
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Steven Dooley
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nikola S Mueller
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
9
|
Alqurashi YE, Al-Hetty HRAK, Ramaiah P, Fazaa AH, Jalil AT, Alsaikhan F, Gupta J, Ramírez-Coronel AA, Tayyib NA, Peng H. Harnessing function of EMT in hepatocellular carcinoma: From biological view to nanotechnological standpoint. ENVIRONMENTAL RESEARCH 2023; 227:115683. [PMID: 36933639 DOI: 10.1016/j.envres.2023.115683] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 05/08/2023]
Abstract
Management of cancer metastasis has been associated with remarkable reduction in progression of cancer cells and improving survival rate of patients. Since 90% of mortality are due to cancer metastasis, its suppression can improve ability in cancer fighting. The EMT has been an underlying cause in increasing cancer migration and it is followed by mesenchymal transformation of epithelial cells. HCC is the predominant kind of liver tumor threatening life of many people around the world with poor prognosis. Increasing patient prognosis can be obtained via inhibiting tumor metastasis. HCC metastasis modulation by EMT and HCC therapy by nanoparticles are discussed here. First of all, EMT happens during progression and advanced stages of HCC and therefore, its inhibition can reduce tumor malignancy. Moreover, anti-cancer compounds including all-trans retinoic acid and plumbaging, among others, have been considered as inhibitors of EMT. The EMT association with chemoresistance has been evaluated. Moreover, ZEB1/2, TGF-β, Snail and Twist are EMT modulators in HCC and enhancing cancer invasion. Therefore, EMT mechanism and related molecular mechanisms in HCC are evaluated. The treatment of HCC has not been only emphasized on targeting molecular pathways with pharmacological compounds and since drugs have low bioavailability, their targeted delivery by nanoparticles promotes HCC elimination. Moreover, nanoparticle-mediated phototherapy impairs tumorigenesis in HCC by triggering cell death. Metastasis of HCC and even EMT mechanism can be suppressed by cargo-loaded nanoparticles.
Collapse
Affiliation(s)
- Yaser E Alqurashi
- Department of Biology, College of Science Al-zulfi, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | | | | | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U. P., India
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | - Nahla A Tayyib
- Faculty of Nursing, Umm Al- Qura University, Makkah, Saudi Arabia
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
10
|
Clinical Significance of Combined Epithelial-Mesenchymal Transition Markers Expression and Role of Rac1 in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24021765. [PMID: 36675278 PMCID: PMC9865966 DOI: 10.3390/ijms24021765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in cancer progression, invasion, and metastasis. We aimed to evaluate the correlations between clinicopathological characteristics and EMT markers in patients with hepatocellular carcinoma (HCC) who underwent surgical resection and to identify the key regulator in EMT process. Fresh-frozen HCC tissues and adjacent nontumor liver tissues from 30 patients who underwent surgical resection were provided by the Gachon University Gil Medical Center Bio Bank. Human HCC cell lines, Hep3B, SNU449, and Huh7 cells were transfected with Rac1 siRNA and exposed to hypoxic conditions. The combined EMT markers expression (down-expression of E-cadherin and overexpression of p21-activated kinases 1 (PAK1)/Snail) by Western blot in HCC tissues when compared to adjacent nontumor liver tissues was significantly associated with macrovascular invasion (p = 0.021), microvascular invasion (p = 0.001), large tumor size (p = 0.021), and advanced tumor stage (p = 0.015). Patients with combined EMT markers expression showed early recurrence and poor overall survival. In vitro studies showed that Rac1 knockdown decreased the expression of EMT markers including PAK1 and Snail in hypoxia-induced Hep3B cells and suppressed the migration and invasion of hypoxia-induced HCC cells. Rac1 may be a potential therapeutic target for inhibition of EMT process through the inhibition of PAK1 and Snail in HCC.
Collapse
|
11
|
Qiu X, Xu H, Wang K, Gao F, Xu X, He H. P-21 Activated Kinases in Liver Disorders. Cancers (Basel) 2023; 15:cancers15020551. [PMID: 36672500 PMCID: PMC9857091 DOI: 10.3390/cancers15020551] [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: 12/14/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
The p21 Activated Kinases (PAKs) are serine threonine kinases and play important roles in many biological processes, including cell growth, survival, cytoskeletal organization, migration, and morphology. Recently, PAKs have emerged in the process of liver disorders, including liver cancer, hepatic ischemia-reperfusion injury, hepatitis, and liver fibrosis, owing to their effects in multiple signaling pathways in various cell types. Activation of PAKs promotes liver cancer growth and metastasis and contributes to the resistance of liver cancer to radiotherapy and chemotherapy, leading to poor survival of patients. PAKs also play important roles in the development and progression of hepatitis and other pathological processes of the liver such as fibrosis and ischemia-reperfusion injury. In this review, we have summarized the currently available studies about the role of PAKs in liver disorders and the mechanisms involved, and further explored the potential therapeutic application of PAK inhibitors in liver disorders, with the aim to provide a comprehensive overview on current progress and perspectives of PAKs in liver disorders.
Collapse
Affiliation(s)
- Xun Qiu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hanzhi Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Correspondence: (K.W.); (H.H.)
| | - Fengqiang Gao
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, 145 Studley Rd., Heidelberg, VIC 3084, Australia
- Correspondence: (K.W.); (H.H.)
| |
Collapse
|
12
|
Long X, Wang D, Wu Z, Liao Z, Xu J. Circular RNA hsa_circ_0004689 (circSWT1) promotes NSCLC progression via the miR‐370‐3p/SNAIL axis by inducing cell epithelial‐mesenchymal transition (EMT). Cancer Med 2022; 12:8289-8305. [PMID: 36530171 PMCID: PMC10134258 DOI: 10.1002/cam4.5527] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/10/2022] [Accepted: 11/04/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Previous studies have reported the role of circular RNAs (circRNAs) in the progression of non-small-cell lung cancer (NSCLC). SWT1-derived circRNAs were confirmed to affect the apoptosis of cardiomyocytes; however, the biological functions of SWT1-derived circRNAs in cancers are still unknown. Here, we investigated the potential role of SWT1-derived circRNAs in NSCLC. METHODS We used quantitative real-time polymerase chain reaction (qRT-PCR) to measure the expression of circSWT1 in NSCLC tissues and paired normal tissues. The potential functions of circSWT1 in tumor progression were assessed by CCK-8, colony formation, wound healing, and matrigel transwell assays in vitro and by xenograft tumor models in vivo. Next, epithelial-mesenchymal transition (EMT) was evaluated by western blotting, immunofluorescence, and immunohistochemistry (IHC). Moreover, circRIP, RNA pulldown assays, luciferase reporter gene assays, and FISH were conducted to illuminate the molecular mechanisms of circSWT1 via the miR-370-3p/SNAIL signal pathway. Then, we knocked out SNAIL in A549 and H1299 cells to identify the roles of circSWT1 in the progression and EMT of NSCLC through SNAIL. Finally, circSWT1 functions were confirmed in vivo using xenograft tumor models. RESULTS CircSWT1 expression was significantly upregulated in NSCLC tissues, and high expression of circSWT1 predicted poor prognosis in NSCLC via survival analysis. In addition, overexpression of circSWT1 promoted the invasion and migration of NSCLC cells. Subsequently, we found that overexpression of circSWT1 induced EMT and that knockdown of circSWT1 inhibited EMT in NSCLC cells. Mechanistically, circSWT1 relieved the inhibition of downstream SNAIL by sponging miR-370-3p. Moreover, we found that these effects could be reversed by knocking out SNAIL. Finally, we verified that circSWT1 promoted NSCLC progression and EMT in xenograft tumor models. CONCLUSION CircSWT1 promoted the invasion, migration, and EMT of NSCLC. CircSWT1 could serve as a potential biomarker and a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xiang Long
- Department of Cardiothoracic Surgery The Second Affiliated Hospital of Nanchang University Nanchang People's Republic of China
| | - Ding‐Guo Wang
- Department of Cardiothoracic Surgery The Second Affiliated Hospital of Nanchang University Nanchang People's Republic of China
| | - Zhi‐Bo Wu
- Department of Cardiothoracic Surgery The Second Affiliated Hospital of Nanchang University Nanchang People's Republic of China
| | - Zhong‐Min Liao
- Department of Cardiothoracic Surgery The Second Affiliated Hospital of Nanchang University Nanchang People's Republic of China
| | - Jian‐Jun Xu
- Department of Cardiothoracic Surgery The Second Affiliated Hospital of Nanchang University Nanchang People's Republic of China
| |
Collapse
|
13
|
Li T, Guan L, Tang G, He B, Huang L, Wang J, Li M, Bai Y, Li X, Zhang H. Downregulation of TMEM220 promotes tumor progression in Hepatocellular Carcinoma. Cancer Gene Ther 2022; 29:835-844. [PMID: 34321624 DOI: 10.1038/s41417-021-00370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/22/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
During the process of long-term carcinogenesis, cells accumulate many mutations. Deregulated genes expression causes profound changes in cell proliferation, which is one of the hallmarks of HCC. A comprehensive understanding of these changes will contribute to the molecular mechanism of HCC progression. Through clinical sample analysis, we found that TMEM220 is downregulated in tumor and lower levels of TMEM220 is associated with poor prognosis in HCC patients. Through overexpressing TMEM220 in HCC cell lines, we found that the proliferation of cancer cells was significantly slowed down and metastasis was significantly reduced. For further study of its molecular mechanism, we performed a reverse-phase protein array (RPPA). The results suggest that phenotypic changes caused by TMEM220 in HCC cells might be associated with FOXO and PI3K-Akt pathways. Mechanism studies showed that overexpression of TMEM220 could regulate β-catenin and FOXO3 transcriptional activity by altering their subcellular localization, affecting the expression of downstream gene p21 and SNAIL, and ultimately reducing the progression of HCC. Altogether, our study proposes a working model in which upregulation of TMEM220 expression alters the genes expression involved in cell proliferation, thereby inhibiting HCC progression, which suggests that TMEM220 might serve as a clinical biomarker.
Collapse
Affiliation(s)
- Ting Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China
| | - Lei Guan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China
| | - Guangbo Tang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China
| | - Bing He
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Lili Huang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Juan Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, PR China
| | - Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanxia Bai
- Department of Otolaryngology-Head-Neck Surgery, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Huqin Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
14
|
WNT5A promotes the metastasis of esophageal squamous cell carcinoma by activating the HDAC7/SNAIL signaling pathway. Cell Death Dis 2022; 13:480. [PMID: 35595735 PMCID: PMC9122958 DOI: 10.1038/s41419-022-04901-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide, with high incidence and mortality rates and low survival rates. However, the detailed molecular mechanism of ESCC progression remains unclear. Here, we first showed significantly higher WNT5A and SNAIL expression in ESCC samples than in corresponding paracancerous samples. High WNT5A and SNAIL expression levels correlated positively with lymphatic metastasis and poor prognosis for patients with ESCC based on immunohistochemical (IHC) staining of 145 paired ESCC samples. Spearman's correlation analyses confirmed the strong positive correlation between WNT5A and SNAIL expression, and patients with ESCC presenting coexpression of WNT5A and SNAIL had the worst prognosis. Then, we verified that the upregulation of WNT5A promoted ESCC cell metastasis in vivo and in vitro, suggesting that WNT5A might be a promising therapeutic target for the prevention of ESCC. Furthermore, WNT5A overexpression induced the epithelial-mesenchymal transition via histone deacetylase 7 (HDAC7) upregulation, and HDAC7 silencing significantly reversed WNT5A-induced SNAIL upregulation and ESCC cell metastasis. In addition, we used HDAC7 inhibitors (SAHA and TMP269) to further confirm that HDAC7 participates in WNT5A-mediated carcinogenesis. Based on these results, HDAC7 is involved in WNT5A-mediated ESCC progression, and approaches targeting WNT5A and HDAC7 might be potential therapeutic strategies for ESCC.
Collapse
|
15
|
Chen G, Liao J, Xu Y, Chen Y, Li J, Bu G, Li Q. LINC01232 Promotes Metastasis and EMT by Regulating miR-506-5p/PAK1 Axis in Gastric Cancer. Cancer Manag Res 2022; 14:1729-1740. [PMID: 35592108 PMCID: PMC9113486 DOI: 10.2147/cmar.s352081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/28/2022] [Indexed: 01/19/2023] Open
Abstract
Background Long non-coding RNA LINC01232 plays an important role in the progression of metastasis in several cancers. However, the function of LINC01232 in gastric cancer is limited. Authors aimed to investigate the role and mechanism of LINC01232 in the metastasis of gastric cancer. Methods The expression levels and correlation of LINC01232, miR-506-5p, and PAK1 were analyzed by GEPIA or ENCORI, and the abundance of LINC01232 and miR-506-5p was measured in tissues and cells via qRT-PCR, the location of LINC01232 in gastric cells was analyzed by nuclear and cytoplasmic fractionation, while the protein levels of PAK1, E-cadherin and vimentin were additionally quantified by Western blotting. Interactions between LINC01232, miR-506-5p, and PAK1 were detected through luciferase reporter assays, qRT-PCR and Western blotting. Cellular viability was evaluated through CCK8 assays, migration ability was measured by transwell assays, invasion ability was tested by wound healing experiment. Results LINC01232 was overexpressed in gastric cancer tissues and cells, and mainly located in nucleus. The inhibition of LINC01232 could suppress migration, invasion and EMT of gastric cancer cells. MiR-506-5p was downregulated in gastric cancer tissues and cells. LINC01232 sponged miR-506-5p to accelerate migration and EMT. PAK1 was certified to be a target of miR-506-5p, inhibition of PAK1 could interrupt LINC01232 overexpression-induced migration of gastric cancer cells. Conclusion The LINC01232/miR-506-5p/PAK1 axis promotes metastasis of gastric cancer cells.
Collapse
Affiliation(s)
- Gang Chen
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
| | - Jiangtao Liao
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
| | - Yan Xu
- Geriatrics Departments, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yinyun Chen
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
| | - Juan Li
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
| | - Guangkui Bu
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
| | - Qingqing Li
- Digestive System Department, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China
- Correspondence: Qingqing Li, Digestive System Department, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, People’s Republic of China, Email
| |
Collapse
|
16
|
Micó-Carnero M, Casillas-Ramírez A, Sánchez-González A, Rojano-Alfonso C, Peralta C. The Role of Neuregulin-1 in Steatotic and Non-Steatotic Liver Transplantation from Brain-Dead Donors. Biomedicines 2022; 10:biomedicines10050978. [PMID: 35625715 PMCID: PMC9138382 DOI: 10.3390/biomedicines10050978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/29/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background. Brain death (BD) and steatosis are key risk factors to predict adverse post-transplant outcomes. We investigated the role of Neuregulin-1 (NRG1) in rat steatotic and non-steatotic liver transplantation (LT) from brain death donors (DBD). Methods: NRG1 pathways were characterized after surgery. Results: NRG1 and p21-activated kinase 1 (PAK1) levels increased in steatotic and non-steatotic grafts from DBDs. The abolishment of NRG1 effects reduced PAK1. When the effect of either NRG1 nor PAK1 was inhibited, injury and regenerative failure were exacerbated. The benefits of the NRG-1-PAK1 axis in liver grafts from DBDs were associated with increased vascular endothelial growth factor-A (VEGFA) and insulin growth factor-1 (IGF1) levels, respectively. Indeed, VEGFA administration in non-steatotic livers and IGF1 treatment in steatotic grafts prevented damage and regenerative failure resulting from the inhibition of either NRG1 or PAK-1 activity in each type of liver. Exogenous NRG1 induced greater injury than BD induction. Conclusions: This study indicates the benefits of endogenous NRG1 in liver grafts from DBDs and underscores the specificity of the NRG1 signaling pathway depending on the type of liver: NRG1-PAK1-VEGFA in non-steatotic livers and NRG1-PAK1-IGF1 in steatotic livers. Exogenous NRG1 is not an appropriate strategy to apply to liver grafts from DBD.
Collapse
Affiliation(s)
- Marc Micó-Carnero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico; (A.C.-R.); (A.S.-G.)
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros 87300, Mexico
| | - Alfredo Sánchez-González
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico; (A.C.-R.); (A.S.-G.)
| | - Carlos Rojano-Alfonso
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
- Correspondence: ; Tel.: +34-932-275-400
| |
Collapse
|
17
|
CMAHP promotes metastasis by reducing ubiquitination of Snail and inducing angiogenesis via GM-CSF overexpression in gastric cancer. Oncogene 2022; 41:159-172. [PMID: 34716430 DOI: 10.1038/s41388-021-02087-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/18/2022]
Abstract
Pseudogenes are generally considered "junk" DNA or "genomic fossils" generated during the evolution process that lack biological activity. However, accumulating reports indicate that pseudogenes have biological functions critical for cancer development. Experiments from the current study showed marked overexpression of the cytidine monophospho-N-acetylneuraminic acid hydroxylase pseudogene (CMAHP) in gastric cancer, which was associated with poor overall survival. However, the mechanisms underlying the activity of CMAHP in tumor development are largely unknown. Gene Set Enrichment Analysis (GSEA) revealed that CMAHP-correlated genes are significantly involved in epithelial-mesenchymal transition (EMT) and angiogenesis. Functional studies further confirmed that CMAHP mediates metastasis and angiogenesis in vitro and in vivo. Furthermore, CMAHP promoted cancer cell migration, invasion, and metastasis through Snail overexpression, which decreased ubiquitination mediated by NF-κB signaling. Angiogenesis is known to be induced by granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation. CMAHP increased GM-CSF transactivation via promoting direct binding of c-Jun to the -1981/-1975 region of the GM-CSF promoter. Notably, CMAHP interacts with Histone H1.4 promoting histone acetylation to enhance c-Jun and RelA (p65) expression. Our collective findings provide novel evidence that CMAHP contributes to tumor progression and modulates metastasis and angiogenesis in gastric cancer.
Collapse
|
18
|
Li Y, Li T, Zhou D, Wei J, Li Z, Li X, Jia S, Ouyang Q, Qi S, Chen Z, Zhang B, Yu J, Jia J, Xu A, Huang J. Role of tight junction-associated MARVEL protein marvelD3 in migration and epithelial-mesenchymal transition of hepatocellular carcinoma. Cell Adh Migr 2021; 15:249-260. [PMID: 34338154 PMCID: PMC8331009 DOI: 10.1080/19336918.2021.1958441] [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] [Indexed: 10/29/2022] Open
Abstract
MarvelD3, a recently identified tight junction membrane protein, could be associated with hepatocellular carcinoma (HCC). We aimed to investigate the role of marvelD3 in Epithelial-Mesenchymal Transition (EMT) and migration of HCC and explore the underlying molecular mechanisms. First, we assessed marvlD3 expression in HCC and normal liver tissues and found loss of marvelD3 expression was significantly correlated with the occurrence and TNM stage of HCC. Second, we detected that marvelD3 was downregulated in HCC cells with transforming growth factor β1 and snail/slug-induced EMT. Finally, we analyzed expression of marvelD3 protein was significantly associated with EMT and the NF-κB signaling pathway. Our study demonstrated that MarvelD3 inhibited EMT and migration of HCC cells along with inhibiting NF-κB signaling pathway.Abbreviations: HCC, Hepatocellular carcinoma; TJ, Tight junction; MARVEL, MAL and related proteins for vesicle trafficking and membrane link; EMT, Epithelial-mesenchymal transition; NF-κB, Nuclear factor kappa B; TAMPs, Tight junction-associated marvel proteins; TGF-β1, Transforming growth factor-β1; MMP9, matrix metallopeptidase 9; RT-PCR, Real-time PCR; IHC, Immunohistochemistry; IF, Immunofluorescence.
Collapse
Affiliation(s)
- Yanmeng Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Teng Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Donghu Zhou
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia Wei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenkun Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojin Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Siyu Jia
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qin Ouyang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Saiping Qi
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhibin Chen
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bei Zhang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing Yu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Anjian Xu
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jian Huang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Shi J, Song S, Li S, Zhang K, Lan Y, Li Y. TNF-α/NF-κB signaling epigenetically represses PSD4 transcription to promote alcohol-related hepatocellular carcinoma progression. Cancer Med 2021; 10:3346-3357. [PMID: 33932127 PMCID: PMC8124102 DOI: 10.1002/cam4.3832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Chronic alcohol consumption is more frequently associated with advanced, aggressive hepatocellular carcinoma (HCC) tumors. Alcohol adversely impacts ER/Golgi membrane trafficking and Golgi protein N-glycosylation in hepatocytes; these effects have been attributed (in part) to dysregulated adenosine diphosphate-ribosylation factor (ARF) GTPase signaling. Here, we investigated the role of the ARF GTPase guanine exchange factor PSD4 in HCC progression. METHODS R-based bioinformatics analysis was performed on publicly available array data. Modulating gene expression was accomplished via lentiviral vectors. Gene expression was analyzed using quantitative real-time PCR and immunoblotting. PSD4 promoter methylation was assessed using quantitative methylation-specific PCR. Phospho-p65(S276)/DNMT1 binding to the PSD4 promoter was analyzed via chromatin immunoprecipitation. We constructed ethanol/DEN-induced and DEN only-induced transgenic murine models of HCC. RESULTS We identified PSD4 as a hypermethylated, suppressed gene in alcohol-related HCC tumors; however, PSD4 was not dysregulated in all-cause HCC tumors. Certain HCC cell lines also displayed varying degrees of PSD4 downregulation. PSD4 overexpression or knockdown decreased and increased cell migration and invasiveness, respectively. Mechanistically, PSD4 transcription was repressed by TNF-α-induced phospho-p65(S276)'s recruitment of DNA methyltransferase 1 (DNMT1), resulting in PSD4 promoter methylation. PSD4 inhibited pro-EMT CDC42 activity, resulting in downregulation of E-cadherin and upregulation of N-cadherin and vimentin. Hepatocyte-specific PSD4 overexpression reduced ethanol/DEN-induced HCC tumor progression and EMT marker expression in vivo. CONCLUSIONS PSD4 is a hypermethylated, suppressed gene in alcohol-related HCC tumors that negatively modulated pro-EMT CDC42 activity. Furthermore, we present a novel phospho-NF-κB p65(S276)/DNMT1-mediated promoter methylation mechanism by which TNF-α/NF-κB signaling represses PSD4 transcription in HCC cells.
Collapse
Affiliation(s)
- Jia'ning Shi
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shupeng Song
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuangxing Li
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kaili Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yinghua Lan
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongguo Li
- Department of Infectious Disease, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
20
|
Yao D, Huang J, Wang J, He Z, Zhang J. Design, synthesis and biological evaluation of novel tetrahydrothieno [2,3-c]pyridine substitued benzoyl thiourea derivatives as PAK1 inhibitors in triple negative breast cancer. J Enzyme Inhib Med Chem 2021; 35:1524-1538. [PMID: 32752894 PMCID: PMC7470115 DOI: 10.1080/14756366.2020.1797710] [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] [Indexed: 02/08/2023] Open
Abstract
The overexpression of P21-activated kinase 1 (PAK1) is associated with poor prognosis in several cancers, which has emerged as a promising drug targets. Based on high-throughput virtual screening strategy, tetrahydrothieno [2,3-c]pyridine scaffold was identified as an initial lead for targeting PAK1. Herein we reported our structure-based optimisation strategy to discover a potent PAK1 inhibitor (7j) which displayed potent PAK1 inhibition and antiproliferatory activity in MDA-MB-231 cells. 7j induced obviously G2/M cell cycle arrest via PAK1-cdc25c-cdc2 pathway, and also inhibited MAPK-ERK and MAPK-JNK cascade to induce MDA-MB-231 cell death. Together, these results provided a novel chemical scaffold as PAK1 inhibitor for breast cancer treatment.
Collapse
Affiliation(s)
- Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen, China.,School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Jian Huang
- Drug Development Department, Shenzhen Honghui Bio-Pharmaceutical Co. Ltd., Shenzhen, China
| | - Jinhui Wang
- Drug Development Department, Shenzhen Honghui Bio-Pharmaceutical Co. Ltd., Shenzhen, China
| | - Zhendan He
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen, China.,School of Pharmaceutical Sciences, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Jin Zhang
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen, China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Li Z, Wang Q, Peng S, Yao K, Chen J, Tao Y, Gao Z, Wang F, Li H, Cai W, Lai Y, Li K, Chen X, Huang H. The metastatic promoter DEPDC1B induces epithelial-mesenchymal transition and promotes prostate cancer cell proliferation via Rac1-PAK1 signaling. Clin Transl Med 2020; 10:e191. [PMID: 33135357 PMCID: PMC7536616 DOI: 10.1002/ctm2.191] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 01/19/2023] Open
Abstract
Metastasis is the major cause of prostate cancer (PCa)-related mortality. Epithelial-mesenchymal transition (EMT) is a vital characteristic feature that empowers cancer cells to adapt and survive at the beginning of metastasis. Therefore, it is essential to identify the regulatory mechanism of EMT in metastatic prostate cancer (mPCa) and to develop a novel therapy to block PCa metastasis. Here, we discovered a novel PCa metastasis oncogene, DEP domain containing 1B (DEPDC1B), which was positively correlated with the metastasis status, high Gleason score, advanced tumor stage, and poor prognosis. Functional assays revealed that DEPDC1B enhanced the migration, invasion, and proliferation of PCa cells in vitro and promoted tumor metastasis and growth in vivo. Mechanistic investigations clarified that DEPDC1B induced EMT and enhanced proliferation by binding to Rac1 and enhancing the Rac1-PAK1 pathway. This DEPDC1B-mediated oncogenic effect was reversed by a Rac1-GTP inhibitor or Rac1 knockdown. In conclusion, we discover that the DEPDC1B-Rac1-PAK1 signaling pathway may serve as a multipotent target for clinical intervention in mPCa.
Collapse
Affiliation(s)
- Zean Li
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Qiong Wang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Shirong Peng
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Kai Yao
- Department of UrologySun Yat‐sen University Cancer CenterGuangzhouGuangdongP. R. China
| | - Junxiu Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Yiran Tao
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Ze Gao
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Fen Wang
- Center for Cancer and Stem Cell BiologyTexas A&M Health Science CenterInstitute of Biosciences and TechnologyHoustonTexas
| | - Hui Li
- Department of PathologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginia
| | - Wenli Cai
- Department of RadiologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Yiming Lai
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Kaiwen Li
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Xu Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| | - Hai Huang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
22
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|
23
|
Dang Y, Guo Y, Ma X, Chao X, Wang F, Cai L, Yan Z, Xie L, Guo X. Systemic analysis of the expression and prognostic significance of PAKs in breast cancer. Genomics 2020; 112:2433-2444. [PMID: 31987914 DOI: 10.1016/j.ygeno.2020.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/24/2019] [Accepted: 01/23/2020] [Indexed: 12/23/2022]
Abstract
PAKs (p21-activated kinases) are reported to play crucial roles in a variety of cellular processes and participate in the progression of human cancers. However, the expression and prognostic values of PAKs remain poorly explored in breast cancers. In our study, we examined the mRNA and protein expression levels of PAKs and the prognostic value. We also analyzed the interaction network, genetic alteration, and functional enrichment of PAKs. The results showed that the mRNA levels of PAK1, PAK2, PAK4 and PAK6 were significantly up-regulated in breast cancer compared with normal tissues, while the reverse trend for PAK3 and PAK5 was found, furthermore, the proteins expression of PAK1, PAK2 and PAK4 in breast cancer tissues were higher than that in normal breast tissues. Survival analysis revealed breast cancer patients with low mRNA expression of PAK3 and PAK5 showed worse RFS, conversely, elevated PAK4 levels predicted worse RFS. In addition, the breast cancer patients with PAKs genetic alterations correlated with worse OS. These results indicated that PAKs might be promising potential biomarkers for breast cancer.
Collapse
Affiliation(s)
- Yifang Dang
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Ying Guo
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiaoyu Ma
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiaoyu Chao
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Fei Wang
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Linghao Cai
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Zhongyi Yan
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Longxiang Xie
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiangqian Guo
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| |
Collapse
|
24
|
Zhao H, Tao S. MiRNA-221 protects islet β cell function in gestational diabetes mellitus by targeting PAK1. Biochem Biophys Res Commun 2019; 520:218-224. [PMID: 31587871 DOI: 10.1016/j.bbrc.2019.09.139] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/30/2019] [Indexed: 12/24/2022]
Abstract
To elucidate the potential function of miRNA-221 in gestational diabetes mellitus (GDM) and the underlying mechanism. MiRNA-221 level was analyzed in the microarray containing placental tissues of GDM rats. After constructing GDM model in rats, miRNA-221 level in placental tissues of GDM rats or controls was determined as well. The relationship between miRNA-221 level and blood glucose in GDM rats was analyzed by Spearman correlation test. Regulatory effects of miRNA-221 on proliferation, apoptosis and insulin secretion in INS-1 cells were assessed. Through dual-luciferase reporter gene assay, the direct target of miRNA-221, PAK1 was identified. At last, potential influences of miRNA-221/PAK1 axis on INS-1 cell phenotypes were determined. MiRNA-221 was downregulated in placental tissues of GDM rats, and its level was negatively correlated to that of blood glucose level in GDM rats. Overexpression of miRNA-221 stimulated insulin secretion, cell proliferation and suppressed apoptosis in INS-1 cells. Knockdown of miRNA-221 achieved the opposite results. PAK1 was proved as the direct target of miRNA-221. Notably, PAK1 was able to reverse regulatory effects of miRNA-221 on INS-1 cell phenotypes. MiRNA-221 regulates proliferation, apoptosis and insulin secretion in islet β cells through targeting PAK1, thus protecting GDM-induced islet dysfunction.
Collapse
Affiliation(s)
- Hongqiang Zhao
- Department of Gerontology, Jinan People's Hospital of Shandong Province, Jinan, China.
| | - Shujuan Tao
- Department of Obstetrics, Jinan Second Maternal and Child Health Hospital of Shandong Province, Jinan, China
| |
Collapse
|