1
|
Li X, Li X, Qin J, Lei L, Guo H, Zheng X, Zeng X. Machine learning-derived peripheral blood transcriptomic biomarkers for early lung cancer diagnosis: Unveiling tumor-immune interaction mechanisms. Biofactors 2025; 51:e2129. [PMID: 39415336 DOI: 10.1002/biof.2129] [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: 08/07/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
Lung cancer continues to be the leading cause of cancer-related mortality worldwide. Early detection and a comprehensive understanding of tumor-immune interactions are crucial for improving patient outcomes. This study aimed to develop a novel biomarker panel utilizing peripheral blood transcriptomics and machine learning algorithms for early lung cancer diagnosis, while simultaneously providing insights into tumor-immune crosstalk mechanisms. Leveraging a training cohort (GSE135304), we employed multiple machine learning algorithms to formulate a Lung Cancer Diagnostic Score (LCDS) based on peripheral blood transcriptomic features. The LCDS model's performance was evaluated using the area under the receiver operating characteristic (ROC) curve (AUC) in multiple validation cohorts (GSE42834, GSE157086, and an in-house dataset). Peripheral blood samples were obtained from 20 lung cancer patients and 10 healthy control subjects, representing an in-house cohort recruited at the Sixth People's Hospital of Chengdu. We employed advanced bioinformatics techniques to explore tumor-immune interactions through comprehensive immune infiltration and pathway enrichment analyses. Initial screening identified 844 differentially expressed genes, which were subsequently refined to 87 genes using the Boruta feature selection algorithm. The random forest (RF) algorithm demonstrated the highest accuracy in constructing the LCDS model, yielding a mean AUC of 0.938. Lower LCDS values were significantly associated with elevated immune scores and increased CD4+ and CD8+ T-cell infiltration, indicative of enhanced antitumor-immune responses. Higher LCDS scores correlated with activation of hypoxia, peroxisome proliferator-activated receptor (PPAR), and Toll-like receptor (TLR) signaling pathways, as well as reduced DNA damage repair pathway scores. Our study presents a novel, machine learning-derived peripheral blood transcriptomic biomarker panel with potential applications in early lung cancer diagnosis. The LCDS model not only demonstrates high accuracy in distinguishing lung cancer patients from healthy individuals but also offers valuable insights into tumor-immune interactions and underlying cancer biology. This approach may facilitate early lung cancer detection and contribute to a deeper understanding of the molecular and cellular mechanisms underlying tumor-immune crosstalk. Furthermore, our findings on the relationship between LCDS and immune infiltration patterns may have implications for future research on therapeutic strategies targeting the immune system in lung cancer.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xuebing Li
- Department of Respiratory and Critical Care Medicine, People's Hospital of Yaan, Yaan, Sichuan, China
| | - Jiangyue Qin
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Lei
- Department of Oncology, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hua Guo
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xi Zheng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuefeng Zeng
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Liu Y, Ding L, Chen G, Wang P, Wang X. A thermo-sensitive hydrogel with prominent hemostatic effect prevents tumor recurrence via anti-anoikis-resistance. J Nanobiotechnology 2024; 22:496. [PMID: 39164723 PMCID: PMC11334358 DOI: 10.1186/s12951-024-02739-3] [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: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Tumor cells can survive when detached from the extracellular matrix (ECM) or lose cell-cell connections, a phenomenon known as anoikis-resistance (AR). AR is closely associated with tumor cell metastasis and recurrence, enabling tumor cells to disseminate, migrate, and invade after detachment. To address this issue, a novel intervention method combining intraoperative hemostasis with multifunctional nanozyme driven-enhanced chemodynamic therapy (ECDT) has been proposed, which holds the potential to weaken the AR capability of tumor cells and suppress tumor recurrence. Here, a nanocomposite containing a dendritic mesoporous nanoframework with Cu2+ was developed using an anion-assisted approach after surface PEG grafting and glucose oxidase (GOx) anchoring (DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG was further encapsulated in a thermal-sensitive hydrogel (H@DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG utilizes its high peroxidase (POD) activity to elevate intracellular ROS levels, thereby weakening the AR capability of bladder cancer cells. Additionally, through its excellent catalase (CAT) activity, DMSN-Cu@GOx/PEG converts the high level of hydrogen peroxide (H2O2) catalyzed by intracellular GOx into oxygen (O2), effectively alleviating tumor hypoxia, downregulating hypoxia-inducible factor-1α (HIF-1α) expression, inhibiting epithelial-mesenchymal transition (EMT) processes, and ultimately suppressing the migration and invasion of bladder cancer cells. Interestingly, in vivo results showed that the thermosensitive hydrogel H@DMSN-Cu@GOx/PEG could rapidly gel at body temperature, forming a gel film on wounds to eliminate residual tumor tissue after tumor resection surgery. Importantly, H@DMSN-Cu@GOx/PEG exhibited excellent hemostatic capabilities, effectively enhancing tissue coagulation during post-tumor resection surgery and mitigating the risk of cancer cell dissemination and recurrence due to surgical bleeding. Such hydrogels undoubtedly possess strong surgical application. Our developed novel nanosystem and hydrogel can inhibit the AR capability of tumor cells and prevent recurrence post-surgery. This study represents the first report of using dendritic mesoporous silica-based nanoreactors for inhibiting the AR capability of bladder cancer cells and suppressing tumor recurrence post-surgery, providing a new avenue for developing strategies to impede tumor recurrence after surgery.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Lei Ding
- School of Rare Earths, University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, People's Republic of China
| | - Gaojie Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Peiyuan Wang
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research On the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, People's Republic of China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China.
- Medical Research Institute, Wuhan University, Wuhan, 430071, People's Republic of China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
3
|
Ma Y, Yu Y, Yin Y, Wang L, Yang H, Luo S, Zheng Q, Pan Y, Zhang D. Potential role of epithelial-mesenchymal transition induced by periodontal pathogens in oral cancer. J Cell Mol Med 2024; 28:e18064. [PMID: 38031653 PMCID: PMC10805513 DOI: 10.1111/jcmm.18064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
With the increasing incidence of oral cancer in the world, it has become a hotspot to explore the pathogenesis and prevention of oral cancer. It has been proved there is a strong link between periodontal pathogens and oral cancer. However, the specific molecular and cellular pathogenic mechanisms remain to be further elucidated. Emerging evidence suggests that periodontal pathogens-induced epithelial-mesenchymal transition (EMT) is closely related to the progression of oral cancer. Cells undergoing EMT showed increased motility, aggressiveness and stemness, which provide a pro-tumour environment and promote malignant metastasis of oral cancer. Plenty of studies proposed periodontal pathogens promote carcinogenesis via EMT. In the current review, we discussed the association between the development of oral cancer and periodontal pathogens, and summarized various mechanisms of EMT caused by periodontal pathogens, which are supposed to play an important role in oral cancer, to provide targets for future research in the fight against oral cancer.
Collapse
Affiliation(s)
- Yiwei Ma
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yingyi Yu
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yuqing Yin
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Liu Wang
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Huishun Yang
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Shiyin Luo
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Qifan Zheng
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of StomatologyChina Medical UniversityShenyangChina
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of StomatologyChina Medical UniversityShenyangChina
| |
Collapse
|
4
|
Giriyappagoudar M, Vastrad B, Horakeri R, Vastrad C. Study on Potential Differentially Expressed Genes in Idiopathic Pulmonary Fibrosis by Bioinformatics and Next-Generation Sequencing Data Analysis. Biomedicines 2023; 11:3109. [PMID: 38137330 PMCID: PMC10740779 DOI: 10.3390/biomedicines11123109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/24/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with reduced quality of life and earlier mortality, but its pathogenesis and key genes are still unclear. In this investigation, bioinformatics was used to deeply analyze the pathogenesis of IPF and related key genes, so as to investigate the potential molecular pathogenesis of IPF and provide guidance for clinical treatment. Next-generation sequencing dataset GSE213001 was obtained from Gene Expression Omnibus (GEO), and the differentially expressed genes (DEGs) were identified between IPF and normal control group. The DEGs between IPF and normal control group were screened with the DESeq2 package of R language. The Gene Ontology (GO) and REACTOME pathway enrichment analyses of the DEGs were performed. Using the g:Profiler, the function and pathway enrichment analyses of DEGs were performed. Then, a protein-protein interaction (PPI) network was constructed via the Integrated Interactions Database (IID) database. Cytoscape with Network Analyzer was used to identify the hub genes. miRNet and NetworkAnalyst databaseswereused to construct the targeted microRNAs (miRNAs), transcription factors (TFs), and small drug molecules. Finally, receiver operating characteristic (ROC) curve analysis was used to validate the hub genes. A total of 958 DEGs were screened out in this study, including 479 up regulated genes and 479 down regulated genes. Most of the DEGs were significantly enriched in response to stimulus, GPCR ligand binding, microtubule-based process, and defective GALNT3 causes HFTC. In combination with the results of the PPI network, miRNA-hub gene regulatory network and TF-hub gene regulatory network, hub genes including LRRK2, BMI1, EBP, MNDA, KBTBD7, KRT15, OTX1, TEKT4, SPAG8, and EFHC2 were selected. Cyclothiazide and rotigotinethe are predicted small drug molecules for IPF treatment. Our findings will contribute to identification of potential biomarkers and novel strategies for the treatment of IPF, and provide a novel strategy for clinical therapy.
Collapse
Affiliation(s)
- Muttanagouda Giriyappagoudar
- Department of Radiation Oncology, Karnataka Institute of Medical Sciences (KIMS), Hubballi 580022, Karnataka, India;
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. Socitey’s College of Pharmacy, Gadag 582101, Karnataka, India;
| | - Rajeshwari Horakeri
- Department of Computer Science, Govt First Grade College, Hubballi 580032, Karnataka, India;
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
| |
Collapse
|
5
|
Su MC, Nethi SK, Dhanyamraju PK, Prabha S. Nanomedicine Strategies for Targeting Tumor Stroma. Cancers (Basel) 2023; 15:4145. [PMID: 37627173 PMCID: PMC10452920 DOI: 10.3390/cancers15164145] [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: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The tumor stroma, or the microenvironment surrounding solid tumors, can significantly impact the effectiveness of cancer therapies. The tumor microenvironment is characterized by high interstitial pressure, a consequence of leaky vasculature, and dense stroma created by excessive deposition of various macromolecules such as collagen, fibronectin, and hyaluronic acid (HA). In addition, non-cancerous cells such as cancer-associated fibroblasts (CAFs) and the extracellular matrix (ECM) itself can promote tumor growth. In recent years, there has been increased interest in combining standard cancer treatments with stromal-targeting strategies or stromal modulators to improve therapeutic outcomes. Furthermore, the use of nanomedicine, which can improve the delivery and retention of drugs in the tumor, has been proposed to target the stroma. This review focuses on how different stromal components contribute to tumor progression and impede chemotherapeutic delivery. Additionally, this review highlights recent advancements in nanomedicine-based stromal modulation and discusses potential future directions for developing more effective stroma-targeted cancer therapies.
Collapse
Affiliation(s)
- Mei-Chi Su
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Susheel Kumar Nethi
- Nanovaccine Institute, Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA;
| | - Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Swayam Prabha
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Temple University, Philadelphia, PA 19111, USA
| |
Collapse
|
6
|
Ma HW, Kim JM, Kim DH, Park IS, Kim JH, Park KC, Seo DH, Kim JH, Che X, Kim TI, Cheon JH, Kim SW. Olfactomedin 4 produces dysplasia but suppresses metastasis of colon cancer. Cancer Gene Ther 2023; 30:694-703. [PMID: 36577836 DOI: 10.1038/s41417-022-00585-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022]
Abstract
Development of colorectal cancer (CRC) is regulated by a series of genetic and microenvironmental alterations. Olfactomedin 4 (OLFM4) is a secreted glycoprotein that is highly expressed in the gastrointestinal tract and modulates inflammation. However, the role of OLFM4 in CRC is uncertain. Here we aimed to explore the function of OLFM4 in CRC in vivo and in vitro. The mRNA expression of OLFM4 was up-regulated in precursor lesions with dysplasia or ulcerative colitis but was reduced in CRC. OLFM4 neutralizing antibody suppressed inflammation-mediated early-stage CRC formation in an AOM/DSS colitis-associated cancer model. OLFM4 knockdown cells exhibited increased cell proliferation and motility in vitro and in vivo. Ablation of OLFM4 increased tumor growth and metastasis in xenograft experiments. In addition, OLFM4 knockdown cells showed elevated expression of colon cancer stem cell markers including CD133, resulting in increased metastasis via epithelial-mesenchymal transition signaling. This study demonstrated that OLFM4 regulates inflammation and cancer progression differently; ablation of OLFM4 promotes cancer metastasis via stemness and epithelial-mesenchymal transition. These results suggest a new route for controlling cancer progression and metastasis.
Collapse
Affiliation(s)
- Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Min Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Da Hye Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ki Cheong Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hyuk Seo
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hyeon Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Xiumei Che
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Il Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
7
|
Guan D, Zhao L, Shi X, Ma X, Chen Z. Copper in cancer: From pathogenesis to therapy. Biomed Pharmacother 2023; 163:114791. [PMID: 37105071 DOI: 10.1016/j.biopha.2023.114791] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023] Open
Abstract
One of the basic trace elements for the structure and metabolism of human tissue is copper. However, as a heavy metal, excessive intake or abnormal accumulation of copper in the body can cause inevitable damage to the organism because copper can result in direct injury to various cell components or disruption of the redox balance, eventually leading to cell death. Interestingly, a growing body of research reports that diverse cancers have raised serum and tumor copper levels. Tumor cells depend on more copper for their metabolism than normal cells, and a decrease in copper or copper overload can have a detrimental effect on tumor cells. New modalities for identifying and characterizing copper-dependent signals offer translational opportunities for tumor therapy, but their mechanisms remain unclear. Therefore, this article summarizes what we currently know about the correlation between copper and cancer and describes the characteristics of copper metabolism in tumor cells and the prospective application of copper-derived therapeutics.
Collapse
Affiliation(s)
- Defeng Guan
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Lihui Zhao
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Xin Shi
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Xiaoling Ma
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China.
| | - Zhou Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Chen Z, Han F, Du Y, Shi H, Zhou W. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:70. [PMID: 36797231 PMCID: PMC9935926 DOI: 10.1038/s41392-023-01332-8] [Citation(s) in RCA: 452] [Impact Index Per Article: 226.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/20/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Having a hypoxic microenvironment is a common and salient feature of most solid tumors. Hypoxia has a profound effect on the biological behavior and malignant phenotype of cancer cells, mediates the effects of cancer chemotherapy, radiotherapy, and immunotherapy through complex mechanisms, and is closely associated with poor prognosis in various cancer patients. Accumulating studies have demonstrated that through normalization of the tumor vasculature, nanoparticle carriers and biocarriers can effectively increase the oxygen concentration in the tumor microenvironment, improve drug delivery and the efficacy of radiotherapy. They also increase infiltration of innate and adaptive anti-tumor immune cells to enhance the efficacy of immunotherapy. Furthermore, drugs targeting key genes associated with hypoxia, including hypoxia tracers, hypoxia-activated prodrugs, and drugs targeting hypoxia-inducible factors and downstream targets, can be used for visualization and quantitative analysis of tumor hypoxia and antitumor activity. However, the relationship between hypoxia and cancer is an area of research that requires further exploration. Here, we investigated the potential factors in the development of hypoxia in cancer, changes in signaling pathways that occur in cancer cells to adapt to hypoxic environments, the mechanisms of hypoxia-induced cancer immune tolerance, chemotherapeutic tolerance, and enhanced radiation tolerance, as well as the insights and applications of hypoxia in cancer therapy.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yan Du
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaqing Shi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China. .,Lanzhou University Sencond Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
9
|
Wu Q, You L, Nepovimova E, Heger Z, Wu W, Kuca K, Adam V. Hypoxia-inducible factors: master regulators of hypoxic tumor immune escape. J Hematol Oncol 2022; 15:77. [PMID: 35659268 PMCID: PMC9166526 DOI: 10.1186/s13045-022-01292-6] [Citation(s) in RCA: 221] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia, a common feature of the tumor microenvironment in various types of cancers, weakens cytotoxic T cell function and causes recruitment of regulatory T cells, thereby reducing tumoral immunogenicity. Studies have demonstrated that hypoxia and hypoxia-inducible factors (HIFs) 1 and 2 alpha (HIF1A and HIF2A) are involved in tumor immune escape. Under hypoxia, activation of HIF1A induces a series of signaling events, including through programmed death receptor-1/programmed death ligand-1. Moreover, hypoxia triggers shedding of complex class I chain-associated molecules through nitric oxide signaling impairment to disrupt immune surveillance by natural killer cells. The HIF-1-galactose-3-O-sulfotransferase 1-sulfatide axis enhances tumor immune escape via increased tumor cell-platelet binding. HIF2A upregulates stem cell factor expression to recruit tumor-infiltrating mast cells and increase levels of cytokines interleukin-10 and transforming growth factor-β, resulting in an immunosuppressive tumor microenvironment. Additionally, HIF1A upregulates expression of tumor-associated long noncoding RNAs and suppresses immune cell function, enabling tumor immune escape. Overall, elucidating the underlying mechanisms by which HIFs promote evasion of tumor immune surveillance will allow for targeting HIF in tumor treatment. This review discusses the current knowledge of how hypoxia and HIFs facilitate tumor immune escape, with evidence to date implicating HIF1A as a molecular target in such immune escape. This review provides further insight into the mechanism of tumor immune escape, and strategies for tumor immunotherapy are suggested.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| | - Li You
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China. .,Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic. .,Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic.
| |
Collapse
|
10
|
Chen Z, Zhang X, Xing Z, Lv S, Huang L, Liu J, Ye S, Li X, Chen M, Zuo S, Tao Y, He Y. OLFM4 deficiency delays the progression of colitis to colorectal cancer by abrogating PMN-MDSCs recruitment. Oncogene 2022; 41:3131-3150. [PMID: 35487976 DOI: 10.1038/s41388-022-02324-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 11/09/2022]
Abstract
Chronic inflammatory bowel disease (IBD) is strongly associated with the development of colitis-associated tumorigenesis (CAT). Despite recent advances in the understanding of polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) responses in cancer, the mechanisms of these cells during this process remain largely uncharacterized. Here, we discovered a glycoprotein, olfactomedin-4 (OLFM4), was highly expressed in PMN-MDSCs from colitis to colorectal cancer (CRC), and its expression level and PMN-MDSC population positively correlated with the progression of IBD to CRC. Moreover, mice lacking OLFM4 in myeloid cells showed poor recruitment of PMN-MDSCs, impaired intestinal homeostasis, and delayed development from IBD to CRC, and increased response to anti-PD1 therapy. The main mechanism of OLFM4-mediated PMN-MDSC activity involved the NF-κB/PTGS2 pathway, through the binding of LGALS3, a galactoside-binding protein expressed on PMN-MDSCs. Our results showed that the OLFM4/NF-κB/PTGS2 pathway promoted PMN-MDSC recruitment, which played an essential role in the maintenance of intestinal homeostasis, but showed resistance to anti-PD1 therapy in CRC.
Collapse
Affiliation(s)
- Ziyang Chen
- Department of Neurosurgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaogang Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuaijun Lv
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Linxuan Huang
- Dongguan Institute of Clinical Cancer Research, Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Jingping Liu
- Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Shubiao Ye
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinyao Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiqi Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaowen Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yingxu Tao
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yumei He
- Department of Neurosurgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China. .,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Gamal-Eldeen AM, Alrehaili AA, Alharthi A, Raafat BM. Perftoran® Inhibits Hypoxia-Associated Resistance in Lung Cancer Cells to Carboplatin. Front Pharmacol 2022; 13:860898. [PMID: 35401227 PMCID: PMC8987772 DOI: 10.3389/fphar.2022.860898] [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: 01/23/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022] Open
Abstract
Perftoran® (perfluorodecalin) is an oxygen carrier, and carboplatin is a common chemotherapy drug used worldwide for lung cancer treatment. Hypoxia is one of the factors that induce resistance of lung cancer cells to carboplatin. This study explored the role of Perftoran®, as an oxygen carrier, in lowering the resistance of lung cancer cells to carboplatin through suppression of hypoxia pathway mediators. The effect of Perftoran® on the resistance of human lung cancer A549 cells to carboplatin was investigated through the evaluation of cytotoxicity by MTT, cell death mode by dual DNA staining, DNA damage by comet assay, DNA platination (DNA/carboplatin adducts) by atomic absorption spectroscopy, hypoxia degree by pimonidazole, HIF-1α/HIF-2α concentrations by ELISA, expression of miRNAs (hypoxamiRs miR-210, miR-21, and miR-181a) by qRT-PCR, and the content of drug resistance transporter MRP-2 by immunocytochemical staining. Results indicated that compared to carboplatin, Perftoran®/carboplatin decreased cell resistance to carboplatin by potentiating its cytotoxicity using only 45% of carboplatin IC50 and inducing apoptosis. Perftoran® induced DNA platination and DNA damage index in cells compared to carboplatin alone. Moreover, compared to treatment with carboplatin alone, co-treatment of cells with Perftoran® and carboplatin inhibited cellular pimonidazole hypoxia adducts, diminished HIF-1α/HIF-2α concentrations, suppressed hypoxamiR expression, and decreased MRP-2. In conclusion, Perftoran® inhibited resistance of lung cancer cells to carboplatin through the inhibition of both hypoxia pathway mediators and the drug resistance transporter MRP-2 and through the induction of DNA/carboplatin adduct formation.
Collapse
Affiliation(s)
- Amira M. Gamal-Eldeen
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- *Correspondence: Amira M. Gamal-Eldeen,
| | - Amani A. Alrehaili
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Afaf Alharthi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Bassem M. Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| |
Collapse
|
12
|
Wang T, Peng B, Luo T, Tian D, Zhao Z, Fu Z, Li Q. ZEB1 recruit Brg1 to regulate airway remodeling epithelial-mesenchymal transition in asthma. Exp Physiol 2022; 107:515-526. [PMID: 35138000 DOI: 10.1113/ep090212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/21/2022] [Indexed: 11/08/2022]
Abstract
What is the central question of this study? The aim was to investigate the function of Brg1 in airway remodeling epithelial-mesenchymal transition (EMT) of asthma and identify the transcription factor (TF) of Brg1 which bind to the protomer of E-cadherin. What is the main finding and its importance? This study highlighted an important molecular mechanism involving chromatin remodeling factor Brahma-related gene-1 (Brg1) that played a critical role in airway remodeling EMT of asthma and demonstrated ZEB1 was the key TF recruiting Brg1. This finding might offer new insights into gene-based therapy for asthma. ABSTRACT: Epithelial-mesenchymal transition (EMT) of airway remodeling happens in children with asthma. Reduction of epithelial marker E-cadherin is reported to be one of the initiating factors of EMT. Our previous study shows that chromatin remodeling factor Brahma-related gene-1 (Brg1) could regulate the expression of E-cadherin indirectly, but the transcription factor (TF) involved in the recruitment of Brg1 in asthma is unknown. Here, we studied the function of Brg1 in an ovalbumin (OVA)-induced asthma model (lung-specific conditional Brg1 (Brg1-/- ) knockdown mice) and human bronchial epithelial 16HBE cells stably expressing Brg1 shRNA. Our results showed that Brg1 was involved in epithelial-mesenchymal transition in asthmatic mice by detecting the expression of EMT markers. Meanwhile, we identified that Brg1 participated in the TGF-β induced EMT of 16HBE cells. We observed that Zinc Finger E-Box Binding Homeobox 1 (ZEB1) and Brg1 colocalized in the EMT of TGF-β induced 16HBE cells. Further results revealed that ZEB1 recruited Brg1 and bound to the promoter region (+3563/3715) to regulate E-cadherin expression. Thus, ZEB1 might be the key TF to recruit Brg1 in airway remodeling EMT of asthma and might be a novel therapeutic target. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Bingming Peng
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Tingting Luo
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Daiyin Tian
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhihua Zhao
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhou Fu
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qubei Li
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| |
Collapse
|
13
|
Ouyang W, Jiang Y, Bu S, Tang T, Huang L, Chen M, Tan Y, Ou Q, Mao L, Mai Y, Yao H, Yu Y, Lin X. A Prognostic Risk Score Based on Hypoxia-, Immunity-, and Epithelialto-Mesenchymal Transition-Related Genes for the Prognosis and Immunotherapy Response of Lung Adenocarcinoma. Front Cell Dev Biol 2022; 9:758777. [PMID: 35141229 PMCID: PMC8819669 DOI: 10.3389/fcell.2021.758777] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Lung adenocarcinoma (LUAD), the most common subtype of non-small cell lung cancer (NSCLC), is associated with poor prognosis. However, current stage-based clinical methods are insufficient for survival prediction and decision-making. This study aimed to establish a novel model for evaluating the risk of LUAD based on hypoxia, immunity, and epithelial-mesenchymal transition (EMT) gene signatures. Methods: In this study, we used data from TCGA-LUAD for the training cohort and GSE68465 and GSE72094 for the validation cohorts. Immunotherapy datasets GSE135222, GSE126044, and IMvigor210 were obtained from a previous study. Using bioinformatic and machine algorithms, we established a risk model based on hypoxia, immune, and EMT gene signatures, which was then used to divide patients into the high and low risk groups. We analyzed differences in enriched pathways between the two groups, following which we investigated whether the risk score was correlated with stemness scores, genes related to m6A, m5C, m1A and m7G modification, the immune microenvironment, immunotherapy response, and multiple anti-cancer drug sensitivity. Results: Overall survival differed significantly between the high-risk and low-risk groups (HR = 4.26). The AUCs for predicting 1-, 3-, and 5-year survival were 0.763, 0.766, and 0.728, respectively. In the GSE68465 dataset, the HR was 2.03, while the AUCs for predicting 1-, 3-, and 5-year survival were 0.69, 0.651, and 0.618, respectively. The corresponding values in the GSE72094 dataset were an HR of 2.36 and AUCs of 0.653, 0.662, and 0.749, respectively. The risk score model could independently predict OS in patients with LUAD, and highly correlated with stemness scores and numerous m6A, m5C, m1A and m7G modification-related genes. Furthermore, the risk model was significantly correlated with multiple immune microenvironment characteristics. In the GSE135222 dataset, the HR was 4.26 and the AUC was 0.702. Evaluation of the GSE126044 and IMvigor210 cohorts indicated that PD-1/PD-LI inhibitor treatment may be indicated in patients with low risk scores, while anti-cancer therapy with various drugs may be indicated in patients with high risk scores. Conclusion: Our novel risk model developed based on hypoxia, immune, and EMT gene signatures can aid in predicting clinical prognosis and guiding treatment in patients with LUAD.
Collapse
Affiliation(s)
- Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yupeng Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shiyi Bu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tiantian Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Linjie Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ming Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiyun Ou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Ultrasound in Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Luhui Mao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yingjie Mai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Artificial Intelligence and Digital Media Programme, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Hong Kong Baptist University, Zhuhai, China
| | - Xiaoling Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Lin Z, Yang S, Zhou Y, Hou Z, Li L, Meng M, Ge C, Zeng B, Lai J, Gao H, Zhao Y, Xie Y, He S, Tang W, Li R, Tan J, Wang W. OLFM4 depletion sensitizes gallbladder cancer cells to cisplatin through the ARL6IP1/caspase-3 axis. Transl Oncol 2022; 16:101331. [PMID: 34974280 PMCID: PMC8728528 DOI: 10.1016/j.tranon.2021.101331] [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: 12/02/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022] Open
Abstract
OLFM4 is involved in development of gallbladder cancer. Depletion of OLFM4 sensitizes gallbladder cancer cells to cisplatin by regulating apoptosis. Low expression of OLFM4 in GBC patients indicates longer survival.
Background Gallbladder cancer (GBC) is a highly lethal malignancy that carries an extremely poor prognosis due to its chemoresistant nature. Cisplatin (CDDP) is a first-line chemotherapeutic for GBC; however, patients experienced no benefit when treated with CDDP alone. The underlying mechanisms of CDDP resistance in GBC remain largely unknown. Methods Agilent mRNA microarray analysis was performed between paired GBC and paracarcinoma to explore differentially expressed genes that might underlie drug resistance. Gene Set Enrichment Analysis (GSEA) was employed to identify key genes mediating CDDP resistance in GBC, and immunohistochemistry was performed to validate protein expression and test correlations with clinicopathological features. In vitro and in vivo functional assays were performed to investigate the proteins’ roles in CDDP resistance. Results Olfactomedin 4 (OLFM4) was differentially expressed between GBC and paracarcinoma and had the highest rank metric score in the GSEA. OLFM4 expression was increasingly upregulated from chronic cholecystitis to GBC in clinical tissue samples, and OLFM4 depletion decreased GBC cell proliferation and invasion. Interestingly, downregulation of OLFM4 reduced ARL6IP1 (antiapoptotic factor) expression and sensitized GBC cells to CDDP both in vitro and in vivo. The evidence indicated that CDDP could significantly increase Bax and Bad expression and activate caspase-3 cascade in OLFM4-depleted GBC cells through ARL6IP1. Clinically, lower OLFM4 expression was associated with good prognosis of GBC patients. Conclusions Our results suggest that OLFM4 is an essential gene that contributes to GBC chemoresistance and could serve as a prognostic biomarker for GBC. Importantly, OLFM4 could be a potential chemotherapeutic target.
Collapse
Affiliation(s)
- Zhuying Lin
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Songlin Yang
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Yong Zhou
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Zongliu Hou
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Lin Li
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Mingyao Meng
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Chunlei Ge
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Baozhen Zeng
- Department of Pathology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, 106, Zhongshan Road II, Guangzhou 510000, China
| | - Jinbao Lai
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China
| | - Hui Gao
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Yiyi Zhao
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Yanhua Xie
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Shan He
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Weiwei Tang
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China
| | - Ruhong Li
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China.
| | - Jing Tan
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China.
| | - Wenju Wang
- Yan'an Hospital Affiliated to Kunming Medical University/Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan 650051, China; Yunnan Cell Biology and Clinical Translational Research Center, Kunming, Yunnan 650051, China; Kunming Key Laboratory of Biotherapy, Kunming, Yunnan 650051, China.
| |
Collapse
|
15
|
Olfm4 Is Highly Expressed in HCC Patients and as a Biomarker and Therapeutic Target for HCC. Can J Gastroenterol Hepatol 2021; 2021:5601678. [PMID: 34912753 PMCID: PMC8668352 DOI: 10.1155/2021/5601678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the primary types of cancer that claims many lives worldwide, and its incidence continues to increase. Conventional therapies against liver cancer are inadequate, and the pathogenesis of HCC remains unclear. Thus, not only are more effective therapies to treat HCC required but also identification of the key genes involved in its pathogenesis is important for developing such therapies. This study found that olfactomedin 4 (OLFM4) level is higher in HCC patients than in healthy individuals. Furthermore, HCC patients also have higher messenger ribonucleic acid (mRNA) expression level in HCC tissues than in liver paracancerous tissues. OLFM4 has high predictive capacity as a biomarker for HCC and closely correlates to tumor size. It is confirmed that OLFM4 contributes to cancer cell proliferation, and HIF1α is involved in this process. Thus, the OLFM4/HIF-1α axis might be a target signaling pathway for developing novel drugs to treat HCC.
Collapse
|
16
|
Ren X, Geng M, Xu K, Lu C, Cheng Y, Kong L, Cai Y, Hou W, Lu Y, Aihaiti Y, Xu P. Quantitative Proteomic Analysis of Synovial Tissue Reveals That Upregulated OLFM4 Aggravates Inflammation in Rheumatoid Arthritis. J Proteome Res 2021; 20:4746-4757. [PMID: 34496567 DOI: 10.1021/acs.jproteome.1c00399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tandem mass tag (TMT)-coupled liquid chromatography coupled with tandem mass spectrometry is a powerful method to investigate synovial tissue protein profiles in patients with rheumatoid arthritis (RA) and osteoarthritis (OA). Protein was isolated from synovial tissue samples of 22 patients and labeled with a TMT kit. Over 500 proteins were identified as the differential expression protein on comparing RA and OA synovial tissue, including 239 upregulated and 271 downregulated proteins. Data are available via ProteomeXchange with identifier PXD027703. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis showed that the majority participated in the developmental processes and protein processing in the endoplasmic reticulum. Olfactomedin 4 (OLFM4), a secreted glycoprotein, in joint inflammation of RA was explored. OLFM4 was upregulated in RA synovial tissue samples. In fibroblast-like synoviocytes (FLS), inflammation cytokines, TNF-α, interleukin (IL)-1β, and LPS can upregulate OLFM4. After OLFM4 knockdown under TNF-α stimulation, RA FLS proliferation was inhibited and the expression of CXCL9, CXCL11, and MMP-1 was decreased. Overall, the RA synovial tissue protein expression profile by proteomic analysis shows some unique targets in RA pathophysiology, and OLFM4 in FLS plays an important role in RA joint inflammation. OLFM4 can be a promising therapeutic target in RA synovial tissue.
Collapse
Affiliation(s)
- Xiaoyu Ren
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Manman Geng
- Precision Medicine Institute, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an 710061, P. R. China.,National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an 710004, Shaanxi, P. R. China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Chao Lu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Yuanyuan Cheng
- Precision Medicine Institute, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an 710061, P. R. China.,National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an 710004, Shaanxi, P. R. China
| | - Linbo Kong
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Weikun Hou
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Yufeng Lu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Yirixiati Aihaiti
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P. R. China
| |
Collapse
|
17
|
Regulation of olfactomedin 4 by Porphyromonas gingivalis in a community context. THE ISME JOURNAL 2021; 15:2627-2642. [PMID: 33731837 PMCID: PMC8397782 DOI: 10.1038/s41396-021-00956-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023]
Abstract
At mucosal barriers, the virulence of microbial communities reflects the outcome of both dysbiotic and eubiotic interactions with the host, with commensal species mitigating or potentiating the action of pathogens. We examined epithelial responses to the oral pathogen Porphyromonas gingivalis as a monoinfection and in association with a community partner, Streptococcus gordonii. RNA-Seq of oral epithelial cells showed that the Notch signaling pathway, including the downstream effector olfactomedin 4 (OLFM4), was differentially regulated by P. gingivalis alone; however, regulation was overridden by S. gordonii. OLFM4 was required for epithelial cell migratory, proliferative and inflammatory responses to P. gingivalis. Activation of Notch signaling was induced through increased expression of the Notch1 receptor and the Jagged1 (Jag1) agonist. In addition, Jag1 was released in response to P. gingivalis, leading to paracrine activation. Following Jag1-Notch1 engagement, the Notch1 extracellular domain was cleaved by P. gingivalis gingipain proteases. Antagonism by S. gordonii involved inhibition of gingipain activity by secreted hydrogen peroxide. The results establish a novel mechanism by which P. gingivalis modulates epithelial cell function which is dependent on community context. These interrelationships have relevance for innate inflammatory responses and epithelial cell fate decisions in oral health and disease.
Collapse
|
18
|
Xu R, Luo X, Ye X, Li H, Liu H, Du Q, Zhai Q. SIRT1/PGC-1α/PPAR-γ Correlate With Hypoxia-Induced Chemoresistance in Non-Small Cell Lung Cancer. Front Oncol 2021; 11:682762. [PMID: 34381712 PMCID: PMC8351465 DOI: 10.3389/fonc.2021.682762] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Resistance is the major cause of treatment failure and disease progression in non-small cell lung cancer (NSCLC). There is evidence that hypoxia is a key microenvironmental stress associated with resistance to cisplatin, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), and immunotherapy in solid NSCLCs. Numerous studies have contributed to delineating the mechanisms underlying drug resistance in NSCLC; nevertheless, the mechanisms involved in the resistance associated with hypoxia-induced molecular metabolic adaptations in the microenvironment of NSCLC remain unclear. Studies have highlighted the importance of posttranslational regulation of molecular mediators in the control of mitochondrial function in response to hypoxia-induced metabolic adaptations. Hypoxia can upregulate the expression of sirtuin 1 (SIRT1) in a hypoxia-inducible factor (HIF)-dependent manner. SIRT1 is a stress-dependent metabolic sensor that can deacetylate some key transcriptional factors in both metabolism dependent and independent metabolic pathways such as HIF-1α, peroxisome proliferator-activated receptor gamma (PPAR-γ), and PPAR-gamma coactivator 1-alpha (PGC-1α) to affect mitochondrial function and biogenesis, which has a role in hypoxia-induced chemoresistance in NSCLC. Moreover, SIRT1 and HIF-1α can regulate both innate and adaptive immune responses through metabolism-dependent and -independent ways. The objective of this review is to delineate a possible SIRT1/PGC-1α/PPAR-γ signaling-related molecular metabolic mechanism underlying hypoxia-induced chemotherapy resistance in the NSCLC microenvironment. Targeting hypoxia-related metabolic adaptation may be an attractive therapeutic strategy for overcoming chemoresistance in NSCLC.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China
| | - Xin Luo
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuan Ye
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyue Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Lv P, Man S, Xie L, Ma L, Gao W. Pathogenesis and therapeutic strategy in platinum resistance lung cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188577. [PMID: 34098035 DOI: 10.1016/j.bbcan.2021.188577] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/25/2021] [Accepted: 05/30/2021] [Indexed: 12/20/2022]
Abstract
Platinum compounds (cisplatin and carboplatin) represent the most active anticancer agents in clinical use both of lung cancer in mono-and combination therapies. However, platinum resistance limits its clinical application. It is necessary to understand the molecular mechanism of platinum resistance, identify predictive markers, and develop newer, more effective and less toxic agents to treat platinum resistance in lung cancer. Here, it summarizes the main molecular mechanisms associated with platinum resistance in lung cancer and the development of new approaches to tackle this clinically relevant problem. Moreover, it could lead to the development of more effective treatment for refractory lung cancer in future.
Collapse
Affiliation(s)
- Panpan Lv
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Lu Xie
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
20
|
Mo F, Xu Y, Zhang J, Zhu L, Wang C, Chu X, Pan Y, Bai Y, Shao C, Zhang J. Effects of Hypoxia and Radiation-Induced Exosomes on Migration of Lung Cancer Cells and Angiogenesis of Umbilical Vein Endothelial Cells. Radiat Res 2020; 194:71-80. [PMID: 32352864 DOI: 10.1667/rr15555.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/27/2020] [Indexed: 11/03/2022]
Abstract
Numerous studies have shown that exosomes play important roles in tumor biology development. However, the function of exosomal protein in cancer progression under different oxygen condition after irradiation is poorly understood. In this study, non-small cell lung cancer (NSCLC) A549 cells were γ-ray irradiated under normoxic or hypoxic conditions, then the exosomes released from the irradiated cells were collected and co-cultured with nonirradiated A549 cells or human umbilical vein endothelial cells (HUVECs). It was found that the exosomes significantly promoted the proliferation, migration and invasion of A549 cells as well as the proliferation and angiogenesis of HUVECs. Moreover, the exosomes released from hypoxic cells and/or irradiated cells had more powerful driving force in tumor progression compared to that generated from normoxia cells. Meanwhile, the proteins contained in the exosomes derived from A549 cells under different conditions were detected using tandem mass tag (TMT), and their expression profiles were analyzed. It was found that the exosome-derived protein of angiopoietin-like 4 (ANGPTL4) contributed to the migration of A549 cells as well as the angiogenesis of HUVECs, suggesting its potential as an effective diagnostic biomarker of metastasis and even a therapeutic target of lung cancer.
Collapse
Affiliation(s)
- Fang Mo
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yanwu Xu
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junling Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Lin Zhu
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chen Wang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Xiaofei Chu
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|