1
|
Guo Y, Shang S, Liang L, Liu E. ZNF385A was identified as a novel colorectal cancer-related functional gene by analysis of the interaction and immune characteristics of oxidative stress and the inflammatory response. Discov Oncol 2025; 16:290. [PMID: 40064736 PMCID: PMC11893970 DOI: 10.1007/s12672-025-02024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Recently, oxidative stress and inflammatory responses have been shown to directly impact tumor growth and the tumor microenvironment (TME). However, more research is necessary to fully understand the relationship between oxidative stress and inflammatory responses and colorectal cancer (CRC). METHODS The FindCluster algorithm was used to extract CRC Single-cell RNA sequencing (scRNA-seq) data and identify tumor cell groupings. From the MSigDB database, genes associated with oxidative stress and the inflammatory response were taken. We identified molecular subtypes and built a predictive risk model with the LASSO-Cox method using the ConsensusClusterPlus software suite. We incorporated the prognostic risk model and other clinicopathological parameters into a column-line chart. Finally, we used Quantitative Polymerase Chain Reaction (qPCR) and immunohistochemistry to check the expression of the unreported hub model genes. Cell proliferation was assessed using EDU and colony formation assays. Reactive Oxygen Species (ROS) tests were used to quantitatively determine the ROS content in CRC cells. The ability of CRC cells to invade and migrate was examined using transwell experiments. The regulatory functions of hub model genes were discovered in vivo using a xenograft model tumor assay. RESULTS Oxidative stress and inflammatory response factors in monocytic/macrophages of CRC were significantly upregulated, and their oxidative stress and inflammatory response functions were significantly higher than those of other cell subgroups, as indicated by the enrichment score. These factors showed significant synergistic overexpression and enrichment in this cell population. We constructed a prognostic risk model consisting of seven signatures. The good and stable prognostic evaluation efficacy of the model was confirmed, and risk scores were determined to be independent prognostic factors for CRC. We explored the relationship between the risk score model and malignant progression of tumor cells, tumor immune microenvironment, genomic variation, chemotherapy resistance, and immune response. Further qPCR and immunohistochemistry analysis showed that the expression of ZNF385A was high in CRC tissues. The functional experiment results indicated that interfering with the expression of ZNF385A could suppress the proliferation, ROS, migration and invasion of SW620 cells in vitro and the growth of xenograft tumors in vivo. CONCLUSION In this study, we investigated the critical expression patterns of oxidative stress- and inflammatory response-related genes in CRC, which may contribute to the prognosis and immunotherapy of CRC. Additionally, we discovered ZNF385A to be a novel oncogene in CRC. These findings imply that this model may be applied to assess prognostic risk and identify potential therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Yaqi Guo
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Shipeng Shang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Leilei Liang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Enrui Liu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Zhang W, Huang H, Wang L, Lehmann BD, Chen SX. An Integrative Multi-Omics Random Forest Framework for Robust Biomarker Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641533. [PMID: 40093058 PMCID: PMC11908250 DOI: 10.1101/2025.03.05.641533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
High-throughput technologies now produce a wide array of omics data, from genomic and transcriptomic profiles to epigenomic and proteomic measurements. Integrating these diverse data types can yield deeper insights into the biological mechanisms driving complex traits and diseases. Yet, extracting key shared biomarkers from multiple data layers remains a major challenge. We present a multivariate random forest (MRF)-based framework enhanced by a novel inverse minimal depth (IMD) metric for integrative variable selection. By assigning response variables to tree nodes and employing IMD to rank predictors, our approach efficiently identifies essential features across different omics types, even when confronted with high-dimensionality and noise. Through extensive simulations and analyses of multi-omics datasets from The Cancer Genome Atlas, we demonstrate that our method outperforms established integrative techniques in uncovering biologically meaningful biomarkers and pathways. Our findings show that selected biomarkers not only correlate with known regulatory and signaling networks but can also stratify patient subgroups with distinct clinical outcomes. The method's scalable, interpretable, and user-friendly implementation ensures broad applicability to a range of research questions. This MRF-based framework advances robust biomarker discovery and integrative multi-omics analyses, accelerating the translation of complex molecular data into tangible biological and clinical insights.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Hanchen Huang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Brian D. Lehmann
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven X. Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
3
|
Gupta VG, Roby KF, Pathak HB, Godwin AK, Gunewardena S, Khabele D. The Tie2 antagonist rebastinib reduces ovarian cancer growth in a syngeneic murine model. BMC Cancer 2025; 25:233. [PMID: 39930466 PMCID: PMC11812249 DOI: 10.1186/s12885-025-13640-z] [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: 07/01/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND The receptor tyrosine kinase TIE2 and its ligands, angiopoietins (ANGPTs), promote angiogenesis. In addition to expression on vascular endothelial cells, TIE2 is expressed on M2-like pro-tumorigenic macrophages. Thus, the TIE2 inhibitor rebastinib was developed as a potential therapy to address multiple cancers. The objective of this study was to determine the effects of rebastinib alone and combined with chemotherapy in a syngeneic murine model of ovarian cancer. METHODS Female C57Bl6J mice were intraperitoneally injected with syngeneic ID8 ovarian cancer cells. Once tumors were established, mice were untreated (control) or treated with rebastinib, carboplatin plus paclitaxel (chemotherapy), or rebastinib plus chemotherapy. In one set of experiments, survival was followed for 140 days. In other experiments, ascites was harvested 24 h after the last treatment and analyzed by flow cytometry. In in vitro experiments, RNA sequencing was performed on ID8 cells and murine peritoneal macrophage cells (PMJ2R) after treatment with rebastinib, chemotherapy, or rebastinib plus chemotherapy. RESULTS Tumor-bearing mice treated with rebastinib plus chemotherapy had longer median survival than mice treated with chemotherapy (132.5 vs. 127 days, P < 0.01). Ascites from mice treated with rebastinib had more CD45 + macrophages (P < 0.03) and cytotoxic T cells (P < 0.0001) than ascites from mice treated with chemotherapy. Rebastinib had no significant effect on the numbers of regulatory T cells, Tie2 + macrophages, or Tie2 + M2 macrophages. In ID8 cells, in vitro, rebastinib treatment upregulated 1528 genes and downregulated 3115 genes. In macrophages, in vitro, rebastinib treatment upregulated 2302 genes and downregulated 2970 genes. Rebastinib differentially regulated ANGPT-like proteins in both types of cells, including several ANGPT-like genes involved in tumorigenesis, angiogenesis, and proliferation. ANGPTL1, an anti-angiogenic and anti-apoptotic gene, was increased tenfold in ID8 cells treated with rebastinib (P < 0.001) but was not altered in macrophages. CONCLUSIONS Rebastinib plus chemotherapy extends survival in a syngeneic murine model of ovarian cancer. Rebastinib alters proportions of immune cell subsets, increases cytotoxic T cells in ascites, and alters gene expression in tumor cells and macrophages.
Collapse
Affiliation(s)
- Vijayalaxmi G Gupta
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 660 S. Euclid Avenue, Mailstop 8064-37-1005, Saint Louis, MO, 63110, USA.
| | - Katherine F Roby
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Harsh B Pathak
- Department of Pathology, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 660 S. Euclid Avenue, Mailstop 8064-37-1005, Saint Louis, MO, 63110, USA.
| |
Collapse
|
4
|
Fang WQ, Zhang XB, Yu Y, Ge J, Meng R. Propofol reduces breast cancer cell stemness via FOXO3/SOX2 axis. J Cancer 2025; 16:1555-1562. [PMID: 39991565 PMCID: PMC11843230 DOI: 10.7150/jca.104142] [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: 09/24/2024] [Accepted: 12/21/2024] [Indexed: 02/25/2025] Open
Abstract
Objective: Propofol is a common intravenous anesthetic in cancer resection surgery, which is considered to exhibit anti-tumor effect in various cancer types. This study was aimed at investigating the role and mechanism of propofol in breast cancer stemness and proliferation. Methods: The breast cancer cells with propofol treatment were sequenced. The expression of FOXO3 in propofol treated cells was detected by RT-qPCR and Western blot. The CSC properties were analyzed by screen cells with ESA+CD44+CD24-/low through flow cytometry and the proliferation capacity were also detected. The expression correlation of FOXO3 and target genes were detected by western blot. The potential binding site of FOXO3 on SOX2 was predicted by JASPAR and verified by dual-luciferase reporter assay and ChIP assay. Results: FOXO3 was found to be upregulated in propofol 24h-treated cells. Propofol could inhibit the capacity of breast cancer cell stemness and proliferation by upregulation FOXO3, which inhibited SOX2 expression transcriptionally. Conclusion: In this study, we uncovered the role of propofol-FOXO3-SOX2 in breast cancer cell stemness and proliferation, which might serve as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Wen-Qian Fang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Hospital of Gynecology and Obstetrics, Tianjin 300100, China
| | - Xiao-Bei Zhang
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yue Yu
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jie Ge
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ran Meng
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
5
|
Wang Z, Li R, Yang G, Wang Y. Cancer stem cell biomarkers and related signalling pathways. J Drug Target 2024; 32:33-44. [PMID: 38095181 DOI: 10.1080/1061186x.2023.2295222] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Abstract
Cancer stem cells (CSCs) represent a distinct subset of neoplastic cells characterised by their heightened capacity for tumorigenesis. These cells are implicated in the facilitation of cancer metastasis, recurrence, and resistance to conventional therapeutic interventions. Extensive scientific research has been devoted to the identification of biomarkers and the elucidation of molecular mechanisms in order to improve targeted therapeutic approaches. Accurate identification of cancer stem cells based on biomarkers can provide a theoretical basis for drug combinations of malignant tumours. Targeted biomarker-based therapies also offer a silver lining for patients with advanced malignancies. This review aims comprehensively to consolidate the latest findings on CSCs biomarkers, targeted agents as well as biomarkers associated signalling pathways in well-established cancer types, thereby contributing to improved prognostic outcomes.
Collapse
Affiliation(s)
- Zhe Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Rui Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guilin Yang
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yijin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
6
|
Sharma M, Dey U, Das AS, Olymon K, Kumar A, Mukhopadhyay R. Anti-tumor potential of high salt in breast Cancer cell lines. Mol Biol Rep 2024; 51:1002. [PMID: 39305332 DOI: 10.1007/s11033-024-09925-4] [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: 06/29/2024] [Accepted: 09/09/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Recent 23Na-MRI reports show higher salt deposition in malignant breast tissue than in surrounding normal tissue. The effect of high salt on cancer progression remains controversial. Here, we investigated the direct effect of high salt on breast cancer progression in vitro. METHODS Here, the impact of high salt on apoptosis, proliferation, cell cycle, adhesion, and migration of MDA-MB-231 and MCF-7 cells was studied using MTT, scratch, and clonogenic assays, as well as RT-PCR and flow cytometry. Gene expression was analyzed using Real-Time PCR and western blotting. The effect of high salt on global transcriptomics changes in MDA MB-231 cells was studied using RNA-sequencing analysis. RESULTS Flow cytometry with Annexin V and CFSE revealed that high salt-induced dose-dependent apoptosis and inhibited proliferation. High salt-induced cell cycle arrest at the G1/S phase of the cell cycle. p-MDM2 is known to suppress p53, which plays a crucial role in regulating apoptosis and cell cycle arrest under cellular stress conditions. High salt treatment led to decreased p-MDM2 and increased p53 expression, suggesting that high salt induces apoptosis through p53 stabilization. decreased p-MDM2 and increased p53 expression. High salt also reduced migration and adhesion of cells in a dose-dependent manner suggesting its inhibitory effect on metastatic properties as evident from wound healing assay. RNA sequencing analysis revealed overexpression of tumor suppressor genes and genes associated with anti-tumor activity (PCDHGA11, EIF3CL, RAVER1, TNFSF15, RANBP3L) and under-expression of genes involved in cancer-promoting activity (MT1X, CLDN14, CSF-2). CONCLUSION Our results unequivocally demonstrate the anti-tumor efficacy of high salt against breast cancer cells, suggesting its potential as a therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Manoj Sharma
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Upalabdha Dey
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Anindhya Sundar Das
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, USA
| | - Kaushika Olymon
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Aditya Kumar
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India.
| | - Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India.
| |
Collapse
|
7
|
Hushmandi K, Saadat SH, Mirilavasani S, Daneshi S, Aref AR, Nabavi N, Raesi R, Taheriazam A, Hashemi M. The multifaceted role of SOX2 in breast and lung cancer dynamics. Pathol Res Pract 2024; 260:155386. [PMID: 38861919 DOI: 10.1016/j.prp.2024.155386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Breast and lung cancers are leading causes of death among patients, with their global mortality and morbidity rates increasing. Conventional treatments often prove inadequate due to resistance development. The alteration of molecular interactions may accelerate cancer progression and treatment resistance. SOX2, known for its abnormal expression in various human cancers, can either accelerate or impede cancer progression. This review focuses on examining the role of SOX2 in breast and lung cancer development. An imbalance in SOX2 expression can promote the growth and dissemination of these cancers. SOX2 can also block programmed cell death, affecting autophagy and other cell death mechanisms. It plays a significant role in cancer metastasis, mainly by regulating the epithelial-to-mesenchymal transition (EMT). Additionally, an imbalanced SOX2 expression can cause resistance to chemotherapy and radiation therapy in these cancers. Genetic and epigenetic factors may affect SOX2 levels. Pharmacologically targeting SOX2 could improve the effectiveness of breast and lung cancer treatments.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Seyedalireza Mirilavasani
- Campus Venlo, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, The Netherlands
| | - Salman Daneshi
- Department of Public Health,School of Health,Jiroft University of Medical Sciences,Jiroft, the Islamic Republic of Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6 Canada
| | - Rasoul Raesi
- Department of Health Services Management, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran.; Department of Nursing, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| |
Collapse
|
8
|
Peng H, Ye T, Deng L, Yang X, Li Q, Tong J, Guo J. Activin and Hepatocyte Growth Factor Promotes Colorectal Cancer Stemness and Metastasis through FOXM1/SOX2/CXCR4 Signaling. Gut Liver 2024; 18:476-488. [PMID: 37458065 PMCID: PMC11096902 DOI: 10.5009/gnl220531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/22/2023] [Accepted: 04/28/2023] [Indexed: 07/18/2023] Open
Abstract
Background/Aims Cancer stem cells (CSCs) are believed to drive tumor development and metastasis. Activin and hepatocyte growth factor (HGF) are important cytokines with the ability to induce cancer stemness. However, the effect of activin and HGF combination treatment on CSCs is still unclear. Methods In this study, we sequentially treated colorectal cancer cells with activin and HGF and examined CSC marker expression, self-renewal, tumorigenesis, and metastasis. The roles of forkhead box M1 (FOXM1) and sex-determining region Y-box 2 (SOX2), two stemness-related transcription factors, in activin/HGF-induced aggressive phenotype were explored. Results Activin and HGF treatment increased the expression of CSC markers and enhanced sphere formation in colorectal cancer cells. The tumorigenic and metastatic capacities of colorectal cancer cells were enhanced upon activin and HGF treatment. Activin and HGF treatment preferentially promoted stemness and metastasis of CD133+ subpopulations sorted from colorectal cancer cells. FOXM1 was upregulated by activin and HGF treatment, and the knockdown of FOXM1 blocked activin/HGF-induced stemness, tumorigenesis, and metastasis of colorectal cancer cells. Similarly, SOX2 was silencing impaired sphere formation of activin/HGF-treated colorectal cancers. Overexpression of SOX2 rescued the stem cell-like phenotype in FOXM1-depleted colorectal cancer cells with activin and HGF treatment. Additionally, the inhibition of FOXM1 via thiostrepton suppressed activin/HGF-induced stemness, tumorigenesis and metastasis. Conclusions Sequential treatment with activin and HGF promotes colorectal cancer stemness and metastasis through activation of the FOXM1/SOX2 signaling. FOXM1 could be a potential target for the treatment of colorectal cancer metastasis.
Collapse
Affiliation(s)
- Hong Peng
- Department of Gastroenterology and Hepatology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Ye
- Department of Gastroenterology and Hepatology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Lei Deng
- Department of Gastroenterology and Hepatology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Xiaofang Yang
- Department of Gastroenterology and Hepatology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Qingling Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jin Tong
- Department of Gastroenterology and Hepatology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Jinjun Guo
- Department of Gastroenterology and Hepatology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Zhang H, Chen K, Zhou Y, Cao Z, Xu C, Zhou L, Wu G, Peng C, Lai S, Wu X. PLA2G2D fosters angiogenesis in non-small cell lung cancer through aerobic glycolysis. Growth Factors 2024; 42:74-83. [PMID: 38164009 DOI: 10.1080/08977194.2023.2297702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Non-small cell lung cancer (NSCLC) stands prominent among the prevailing and formidable oncological entities. The immune and metabolic-related molecule Phospholipase A2, group IID (PLA2G2D) exerts promotional effects on tumor progression. However, its involvement in cancer angiogenesis remains elusive. Therefore, this investigation delved into the functional significance of PLA2G2D concerning angiogenesis in NSCLC. This study analyzed the expression and enriched pathways of PLA2G2D in NSCLC tissues through bioinformatics analysis, and measured the expression of PLA2G2D in NSCLC cells using qRT-PCR and western blot (WB). Subsequently, the viability and angiogenic potential of NSCLC cells were assessed employing CCK-8 and angiogenesis assays, respectively. The expression profile of angiogenic factors was analyzed through WB. Finally, the expression of glycolysis pathway-related genes, extracellular acidification rate and oxygen consumption rate, and the levels of pyruvate, lactate, citrate, and malate were analyzed in NSCLC cells using qRT-PCR, Seahorse XF 96, and related kits. Bioinformatics analysis revealed the upregulation of PLA2G2D in NSCLC tissues and its association with VEGF and glycolysis signaling pathways. Molecular and cellular experiments demonstrated that upregulated PLA2G2D promoted the viability, angiogenic ability, and glycolysis pathway of NSCLC cells. Rescue assays revealed that the effects of high expression of PLA2G2D on the viability, angiogenic ability, and glycolysis of NSCLC cells were weakened after the addition of the glycolysis inhibitor 2-DG. In summary, PLA2G2D plays a key role in NSCLC angiogenesis through aerobic glycolysis, displaying great potential as a target for anti-angiogenesis therapy.
Collapse
Affiliation(s)
- Huaizhong Zhang
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Keng Chen
- Medical College of Hangzhou Normal University, Hangzhou, China
| | - Yongqing Zhou
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Zhuo Cao
- Department of Respiratory and Critical Care Medicine, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Cunlai Xu
- Department of Respiratory and Critical Care Medicine, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Lin Zhou
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Gongzhi Wu
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Congxiong Peng
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Songqing Lai
- Department of Cardiothoracic Surgery, the first Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuhui Wu
- Department of Cardiothoracic Surgery, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| |
Collapse
|
10
|
Chen ZW, Dong ZB, Xiang HT, Chen SS, Yu WM, Liang C. Helicobacter pylori CagA protein induces gastric cancer stem cell-like properties through the Akt/FOXO3a axis. J Cell Biochem 2024; 125:e30527. [PMID: 38332574 DOI: 10.1002/jcb.30527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/24/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
The presence of Helicobacter pylori (H. pylori) infection poses a substantial risk for the development of gastric adenocarcinoma. The primary mechanism through which H. pylori exerts its bacterial virulence is the cytotoxin CagA. This cytotoxin has the potential to induce inter-epithelial mesenchymal transition, proliferation, metastasis, and the acquisition of stem cell-like properties in gastric cancer (GC) cells infected with CagA-positive H. pylori. Cancer stem cells (CSCs) represent a distinct population of cells capable of self-renewal and generating heterogeneous tumor cells. Despite evidence showing that CagA can induce CSCs-like characteristics in GC cells, the precise mechanism through which CagA triggers the development of GC stem cells (GCSCs) remains uncertain. This study reveals that CagA-positive GC cells infected with H. pylori exhibit CSCs-like properties, such as heightened expression of CD44, a specific surface marker for CSCs, and increased ability to form tumor spheroids. Furthermore, we have observed that H. pylori activates the PI3K/Akt signaling pathway in a CagA-dependent manner, and our findings suggest that this activation is associated with the CSCs-like characteristics induced by H. pylori. The cytotoxin CagA, which is released during H. pylori infection, triggers the activation of the PI3K/Akt signaling pathway in a CagA-dependent manner. Additionally, CagA inhibits the transcription of FOXO3a and relocates it from the nucleus to the cytoplasm by activating the PI3K/Akt pathway. Furthermore, the regulatory function of the Akt/FOXO3a axis in the transformation of GC cells into a stemness state was successfully demonstrated.
Collapse
Affiliation(s)
- Zheng-Wei Chen
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Zhe-Bin Dong
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Han-Ting Xiang
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Sang-Sang Chen
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Wei-Ming Yu
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Chao Liang
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Wang D, Li H, Zeng T, Chen Q, Huang W, Huang Y, Liao Y, Jiang Q. Exosome-transmitted ANGPTL1 suppresses angiogenesis in glioblastoma by inhibiting the VEGFA/VEGFR2/Akt/eNOS pathway. J Neuroimmunol 2024; 387:578266. [PMID: 38150891 DOI: 10.1016/j.jneuroim.2023.578266] [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: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Glioblastoma (GBM) is a highly vascularized malignancy that relies on new vessel generation, and thus targeting angiogenesis has been a promising anti-GBM approach. ANGPTL1 is well-known for its anti-angiogenic property; nevertheless, its role in GBM is yet to be explored. Recently, the crucial role of exosomes (Exos) as intercellular communication mediators has gained prominence in GBM therapy. This work aimed to explore the role of exosomal ANGPTL1 in GBM angiogenesis and its mechanisms. METHODS Bioinformatic analysis was performed to evaluate ANGPTL expression in GBM. Human GBM cell lines (U87 and U251) and a xenograft mouse model were employed. Exos were isolated from oe-NC- and oe-ANGPTL-transfected bone mesenchymal stem cells and identified. Cell proliferation, migration, and apoptosis were detected. Immunofluorescence, qRT-PCR, western blotting, co-immunoprecipitation, and immunohistochemistry were used to determine the molecular mechanisms underlying exosomal ANGPTL1 against GBM angiogenesis. Besides, tube generation and transmission electron microscope assays were conducted to assess GBM angiogenesis. RESULTS Low ANGPTL1 expression was observed in GBM tumor tissues and cells. Functionally, e-ANGPTL-Exos inhibited GBM malignant progression and angiogenesis in vitro and in vivo. Mechanically, e-ANGPTL-Exos reduced VEGFA expression and blocked the VEGFR2/Akt/eNOS pathway in GBM cells and tumor tissues. Co-immunoprecipitation revealed a link between ANGPTL1 and VEGFA in GBM cells. Notably, oe-VEGFA abolished the suppressive functions of e-ANGPTL-Exos in GBM progression and angiogenesis and the VEGFR2/Akt/eNOS axis. The VEGFR2 inhibitor, vandetanib, eliminated the promotive effects of oe-VEGFA on GBM angiogenesis with suppressed VEGFR2/Akt/eNOS pathway. CONCLUSIONS Exosomal ANGPTL1 suppressed GBM angiogenesis by inhibiting the VEGFA/VEGFR2/Akt/eNOS axis.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| | - Huichen Li
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Tianxiang Zeng
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Qiang Chen
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Weilong Huang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yujing Huang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yuqing Liao
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Qiuhua Jiang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
12
|
Thorin E, Labbé P, Lambert M, Mury P, Dagher O, Miquel G, Thorin-Trescases N. Angiopoietin-Like Proteins: Cardiovascular Biology and Therapeutic Targeting for the Prevention of Cardiovascular Diseases. Can J Cardiol 2023; 39:1736-1756. [PMID: 37295611 DOI: 10.1016/j.cjca.2023.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Despite the best pharmacologic tools available, cardiovascular diseases (CVDs) remain a major cause of morbidity and mortality in developed countries. After 2 decades of research, new therapeutic targets, such as angiopoietin-like proteins (ANGPTLs), are emerging. ANGPTLs belong to a family of 8 members, from ANGPTL1 to ANGPTL8; they have structural homology with angiopoietins and are secreted in the circulation. ANGPTLs display a multitude of physiological and pathologic functions; they contribute to inflammation, angiogenesis, cell death, senescence, hematopoiesis, and play a role in repair, maintenance, and tissue homeostasis. ANGPTLs-particularly the triad ANGPTL3, 4, and 8-have an established role in lipid metabolism through the regulation of triacylglycerol trafficking according to the nutritional status. Some ANGPTLs also contribute to glucose metabolism. Therefore, dysregulation in ANGPTL expression associated with abnormal circulating levels are linked to a plethora of CVD and metabolic disorders including atherosclerosis, heart diseases, diabetes, but also obesity and cancers. Because ANGPTLs bind to different receptors according to the cell type, antagonists are therapeutically inadequate. Recently, direct inhibitors of ANGPTLs, mainly ANGPTL3, have been developed, and specific monoclonal antibodies and antisense oligonucleotides are currently being tested in clinical trials. The aim of the current review is to provide an up-to-date preclinical and clinical overview on the function of the 8 members of the ANGPTL family in the cardiovascular system, their contribution to CVD, and the therapeutic potential of manipulating some of them.
Collapse
Affiliation(s)
- Eric Thorin
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada.
| | - Pauline Labbé
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Mélanie Lambert
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada
| | - Pauline Mury
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada
| | - Olina Dagher
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada; Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Géraldine Miquel
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
13
|
Guan J, Zhang ZY, Sun JH, Wang XP, Zhou ZQ, Qin L. LITAF inhibits colorectal cancer stemness and metastatic behavior by regulating FOXO1-mediated SIRT1 expression. Clin Exp Metastasis 2023:10.1007/s10585-023-10213-x. [PMID: 37266842 DOI: 10.1007/s10585-023-10213-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/20/2023] [Indexed: 06/03/2023]
Abstract
Lipopolysaccharide-induced tumor necrosis factor alpha factor (LITAF) is a transcription factor that activates the transcription of TNF-α and regulates the inflammatory response. LITAF has been found to have potential anti-cancer effects of in several tumors. However, the role of LITAF in colorectal cancer (CRC) remains unclear. Through a comprehensive pan-cancer analysis of the Cancer Genome Atlas (TCGA), LITAF was identified as a differentially downregulated gene in CRC. We hypothesized that LITAF may participate in the modulation of CRC progression. The present study was aimed to investigate the expression profile of LITAF in CRC and its effect on metastatic behavior and stemness as well as the underlying molecular mechanism. The expression profile of LITAF in CRC, and its relationship with the prognosis of CRC were explored using public databases. LITAF expression was detected by quantitative real-time PCR (qRT-PCR), western blot, and immunohistochemistry. Furthermore, the effects of overexpression or knockdown of LITAF on cell proliferation, apoptosis, migration, invasion, and stemness of CRC cells were investigated in vitro. The regulatory effect of LITAF on forkhead Box O 1 (FOXO1)-sirtuin 1 (SIRT1) signaling axis was also explored. In addition, a xenograft mouse model was used to investigate the in-vivo role of LITAF. LITAF was downregulated in tumor tissues and its expression was associated with the prognosis, pathological stage and liver metastasis. In-vitro experiments confirmed that LITAF inhibited tumor cell proliferation, migration, invasion and stemness, and induced cell apoptosis. In vivo experiments demonstrated that LITAF inhibited the tumorigenicity and liver metastasis in tumor-bearing mice. Additionally, LITAF promoted FOXO1-mediated SIRT1 inhibition, thus regulating cancer stemness and malignant phenotypes. LITAF was silenced in CRC and it participated in the progression of CRC by inhibiting CRC cell stemness, and malignant phenotypes. Therefore, LITAF may serve as a novel biomarker of CRC prognosis.
Collapse
Affiliation(s)
- Jiao Guan
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zheng-Yun Zhang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian-Hua Sun
- Department of Emergency, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xin-Ping Wang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zun-Qiang Zhou
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Lei Qin
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
14
|
Wang Y, Yi Y, Pan S, Zhang Y, Fu J, Wu X, Qin X. Angiopoietin-like protein 3 promotes colorectal cancer progression and liver metastasis partly via the mitogen-activated protein kinase 14 pathway. Mol Carcinog 2023; 62:546-560. [PMID: 36692110 DOI: 10.1002/mc.23506] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/25/2023]
Abstract
Colorectal cancer (CRC) remains one of the most common malignancies worldwide, and liver metastasis represents a considerable challenge during CRC treatment. Aberrant expression of angiopoietin-like protein 3 (ANGPTL3) has been reported in several human cancer types. However, the function and mechanism of ANGPTL3 in CRC remain unclear. In this study, we first explored ANGPTL3 expression profiles in CRC datasets from ONCOMINE and in local samples from patients with CRC. We then elucidated the function of ANGPTL3 via knockdown and overexpression experiments. Bioinformatic analyses were performed to investigate the biological function and associated molecular mechanisms of ANGPTL3 in CRC oncogenesis and development. Finally, a xenograft model of liver metastasis was used to determine the role of ANGPTL3 in CRC metastasis. Our findings indicated that ANGPTL3 expression was upregulated in human CRC tissues, with high ANGPTL3 expression significantly correlated with poor survival of patients with CRC. ANGPTL3 overexpression promoted the proliferation and migration of CRC cells partially through mitogen-activated protein kinase 14 (MAPK14), while ANGPTL3 silencing had the opposite effect. Moreover, ANGPTL3 downregulation suppressed tumor growth and liver metastasis in xenograft mice. Collectively, the results presented here indicate that ANGPTL3 promotes cell proliferation and liver metastasis partly via MAPK14, suggesting that ANGPTL3 plays a tumor-promoting role in CRC progression and thus may represent a therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Yuexia Wang
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Yi Yi
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Shengli Pan
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Yuhao Zhang
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Jun Fu
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Xiaolin Wu
- Central Laboratory, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| | - Xianju Qin
- Department of General Surgery, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai, China
| |
Collapse
|
15
|
Ding LN, Yu YY, Ma CJ, Lei CJ, Zhang HB. SOX2-associated signaling pathways regulate biological phenotypes of cancers. Biomed Pharmacother 2023; 160:114336. [PMID: 36738502 DOI: 10.1016/j.biopha.2023.114336] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
SOX2 is a transcription factor involved in multiple stages of embryonic development. In related reports, SOX2 was found to be abnormally expressed in tumor tissues and correlated with clinical features such as TNM staging, tumor grade, and prognosis in patients with various cancer types. In most cancer types, SOX2 is a tumor-promoting factor that regulates tumor progression and metastasis primarily by maintaining the stemness of cancer cells. In addition, SOX2 also regulates the proliferation, apoptosis, invasion, migration, ferroptosis and drug resistance of cancer cells. However, SOX2 acts as a tumor suppressor in some cases in certain cancer types, such as gastric and lung cancer. These key regulatory functions of SOX2 involve complex regulatory networks, including protein-protein and protein-nucleic acid interactions through signaling pathways and noncoding RNA interactions, modulating SOX2 expression may be a potential therapeutic strategy for clinical cancer patients. Therefore, we sorted out the phenotypes related to SOX2 in cancer, hoping to provide a basis for further clinical translation.
Collapse
Affiliation(s)
- L N Ding
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Y Y Yu
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - C J Ma
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - C J Lei
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - H B Zhang
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Sequential Treatment with Activin and Hepatocyte Growth Factor Induces FOXM1 to Promote Colorectal Cancer Liver Metastasis. Can J Gastroenterol Hepatol 2022; 2022:8996203. [PMID: 36591565 PMCID: PMC9803576 DOI: 10.1155/2022/8996203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are involved in liver metastasis in colorectal cancer (CRC). Activin and hepatocyte growth factor (HGF) are important regulators of stem cell properties. This study was performed to explore the effect of activin and HGF on CRC invasion and metastasis. The key genes involved in the action of activin and HGF in CRC were identified. METHODS HCT116 CRC cells were sequentially treated with activin and HGF and examined for migration and invasion in vitro and liver metastasis in vivo. RNA sequencing was performed to identify differentially expressed genes in response to activin and HGF. RESULTS Sequential treatment with activin and HGF-enhanced CRC cell migration, invasion, and metastasis. CXCR4 and AFP expressions were increased by activin and HGF treatment. Knockdown of FOXM1 blocked liver metastasis from HCT116 cells pretreated with activin and HGF and suppressed CXCR4 and AFP expression. Activin alone increased the mRNA and protein expression of FOXM1. In contrast, HGF alone enhanced the phosphorylation of FOXM1, without altering the total protein level of FOXM1. SMAD2 was required for activin-mediated FOXM1 induction. FOXM1 transactivated CXCR4 by directly binding to the promoter of CXCR4. Additionally, CXCR4 regulated AFP expression through the NF-κB pathway. CONCLUSIONS Sequential treatment with activin and HGF accelerates CRC invasion and liver metastasis, which involves the upregulation and activation of FOXM1 and induction of CXCR4 and AFP.
Collapse
|
17
|
Non-alcoholic fatty liver disease and liver secretome. Arch Pharm Res 2022; 45:938-963. [PMCID: PMC9703441 DOI: 10.1007/s12272-022-01419-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
|
18
|
Zhang Y, Yang X, Liu S, Zhuang Z, Wei M, Deng X, Wang Z. Comprehensive Analysis of Potential Prognostic Values of ANGPTLs in Colorectal Cancer. Genes (Basel) 2022; 13:genes13122215. [PMID: 36553482 PMCID: PMC9777639 DOI: 10.3390/genes13122215] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors in the world. CRC recurrence and metastasis cause poor prognosis. ANGPTLs (angiopoietin-like proteins) are a family of proteins that are widely involved in metabolic disease and tumorigenesis. The roles of ANGPTLs in CRC are still controversial and deserve further research. In this study, several databases were employed to explore the expression profiles, prognostic values, genetic alterations, potential biological function, and immune infiltration correlation of ANGPTLs in CRC. The expression of ANGPTL4 was significantly positively correlated with the stage of CRC. Therefore, cell and molecular experiments were further performed to explore the roles of ANGPTL4. Our results showed that the transcriptions of ANGPTLs in colon cancer and rectal cancer tissues were lower than those in normal tissues, but the protein expression varied among different ANGPTLs. In addition, the high expression of ANGPTLs led to a relatively poor oncological outcome. Specifically, the expression of ANGPTL4 is significantly positively correlated with the stage of CRC. Further investigation revealed that ANGPTLs are mainly involved in signal transduction and the regulation of transcription, while KEGG pathway analyses demonstrated pathways in cancer. Additionally, we also observed that ANGPTL4 could promote the proliferation and migration of CRC cells, and four specific small molecule compounds had potential ANGPTL4-binding capabilities, suggesting the clinical application of these small molecule compounds on CRC treatment. Our findings imply the prognostic values and potential therapeutic targets of ANGPTLs in CRC.
Collapse
Affiliation(s)
- Yang Zhang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sicheng Liu
- Research Laboratory of Cancer Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zixuan Zhuang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingtian Wei
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangbing Deng
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: ; Tel.: +86-028-85422480; Fax: +86-28-81654035
| |
Collapse
|
19
|
Chen Z, Mei K, Xiao Y, Xiong Y, Long W, Wang Q, Zhong J, Di D, Ge Y, Luo Y, Li Z, Huang Y, Gu R, Wang B. Prognostic Assessment of Oxidative Stress-Related Genes in Colorectal Cancer and New Insights into Tumor Immunity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2518340. [PMID: 36299603 PMCID: PMC9590115 DOI: 10.1155/2022/2518340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/21/2022] [Indexed: 11/19/2022]
Abstract
Oxidative stress is crucial to the biology of tumors. Oxidative stress' potential predictive significance in colorectal cancer (CRC) has not been studied; nevertheless here, we developed a forecasting model based on oxidative stress to forecast the result of CRC survival and enhance clinical judgment. The training set was chosen from the transcriptomes of 177 CRC patients in GSE17536. For validation, 65 samples of colon cancer from GSE29621 were utilized. For the purpose of choosing prognostic genes, the expression of oxidative stress-related genes (OXEGs) was found. Prognostic risk models were built using multivariate Cox regression analysis, univariate Cox regression analysis, and LASSO regression analysis. The outcomes of the western blot and transcriptome sequencing tests were finally confirmed. ATF4, CARS2, CRP, GPX1, IL1B, MAPK8, MRPL44, MTFMT, NOS1, OSGIN2, SOD2, AARS2, and FOXO3 were among the 14 OXEGs used to build prognostic characteristics. Patients with CRC were categorized into low-risk and high-risk groups according on their median risk scores. Cox regression analysis using single and multiple variables revealed that OXEG-related signals were independent risk factors for CRC. Additionally, the validation outcomes from western blotting and transcriptome sequencing demonstrated that OXEGs were differently expressed. Using 14 OXEGs, our work creates a predictive signature that may be applied to the creation of new prognostic models and the identification of possible medication candidates for the treatment of CRC.
Collapse
Affiliation(s)
- Zilu Chen
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Kun Mei
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yao Xiao
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Xiong
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Long
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qin Wang
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiang Zhong
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Dongmei Di
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yunxi Ge
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Yi Luo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, China
- Department of Oncology, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, Jiangsu 210028, China
| | - Ziyun Li
- Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Huang
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Renjun Gu
- Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bin Wang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| |
Collapse
|
20
|
ANGPTL1, Foxo3a-Sox2, and colorectal cancer metastasis. Clin Sci (Lond) 2022; 136:1367-1370. [PMID: 36156125 PMCID: PMC9527825 DOI: 10.1042/cs20220394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022]
Abstract
In the present commentary, we discuss new observations stating that angiopoietin-like protein 1 (ANGPTL1) attenuates cancer metastasis and stemness through Forkhead box O-3a (Foxo3a)–SRY-related HMG-box-2 (Sox2) axis in colorectal cancer (Clin. Sci. (2022) 136, 657–673, https://doi.org/10.1042/CS20220043). ANGPTL1 has been reported to play a critical role in cancer progression and metastasis. However, the underlying mechanisms remain controversial. Here, we integrate the possible mechanisms for ANGPTL1 inhibiting colorectal cancer liver metastasis and discuss the regulation of ANGPTL1 on the Foxo3a–Sox2 pathway. Although ANGPTL1 showed multifunctional potential, there is still a long way to go for ANGPTL1 to be an effective treatment strategy in the clinic.
Collapse
|