251
|
Wang X, Xue X, Pang M, Yu L, Qian J, Li X, Tian M, Lyu A, Lu C, Liu Y. Epithelial-mesenchymal plasticity in cancer: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e659. [PMID: 39092293 PMCID: PMC11292400 DOI: 10.1002/mco2.659] [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: 01/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Currently, cancer is still a leading cause of human death globally. Tumor deterioration comprises multiple events including metastasis, therapeutic resistance and immune evasion, all of which are tightly related to the phenotypic plasticity especially epithelial-mesenchymal plasticity (EMP). Tumor cells with EMP are manifest in three states as epithelial-mesenchymal transition (EMT), partial EMT, and mesenchymal-epithelial transition, which orchestrate the phenotypic switch and heterogeneity of tumor cells via transcriptional regulation and a series of signaling pathways, including transforming growth factor-β, Wnt/β-catenin, and Notch. However, due to the complicated nature of EMP, the diverse process of EMP is still not fully understood. In this review, we systematically conclude the biological background, regulating mechanisms of EMP as well as the role of EMP in therapy response. We also summarize a range of small molecule inhibitors, immune-related therapeutic approaches, and combination therapies that have been developed to target EMP for the outstanding role of EMP-driven tumor deterioration. Additionally, we explore the potential technique for EMP-based tumor mechanistic investigation and therapeutic research, which may burst vigorous prospects. Overall, we elucidate the multifaceted aspects of EMP in tumor progression and suggest a promising direction of cancer treatment based on targeting EMP.
Collapse
Affiliation(s)
- Xiangpeng Wang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoxia Xue
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Mingshi Pang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Liuchunyang Yu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Jinxiu Qian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoyu Li
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Meng Tian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Aiping Lyu
- School of Chinese MedicineHong Kong Baptist UniversityKowloonHong KongChina
| | - Cheng Lu
- Institute of Basic Research in Clinical MedicineChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuanyan Liu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
252
|
Yan P, Wang G, Huang M, Liu Z, Dai C, Hu B, Gu M, Deng Z, Liu R, Wang X, Liu T. Combinatorial Biosynthesis Creates a Novel Aglycone Polyether with High Potency and Low Side Effects Against Bladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404668. [PMID: 38935027 PMCID: PMC11348059 DOI: 10.1002/advs.202404668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Polyethers play a crucial role in the development of anticancer drugs. To enhance the anticancer efficacy and reduce the toxicity of these compounds, thereby advancing their application in cancer treatment, herein, guided by the structure-activity relationships of aglycone polyethers, novel aglycone polyethers are rationally redesigned with potentially improved efficacy and reduced toxicity against tumors. To realize the biosynthesis of the novel aglycone polyethers, the gene clusters and the post-polyketide synthase tailoring pathways for aglycone polyethers endusamycin and lenoremycin are identified and subjected to combinatorial biosynthesis studies, resulting in the creation of a novel aglycone polyether termed End-16, which demonstrates significant potential for treating bladder cancer (BLCA). End-16 demonstrates the ability to suppress the proliferation, migration, invasion, and cellular protrusions formation of BLCA cells, as well as induce cell cycle arrest in the G1 phase in vitro. Notably, End-16 exhibits superior inhibitory activity and fewer side effects against BLCA compared to the frontline anti-BLCA drug cisplatin in vivo, thereby warranting further preclinical studies. This study highlights the significant potential of integrating combinatorial biosynthesis strategies with rational design to create unnatural products with enhanced pharmacological properties.
Collapse
Affiliation(s)
- Pan Yan
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Gang Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Biological RepositoriesHuman Genetic Resource Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
- Medical Research InstituteFrontier Science Center of Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Minjian Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Wuhan Hesheng Technology Co., LtdWuhan430074China
| | - Zhen Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Chong Dai
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Ben Hu
- Precision Cancer Diagnostic CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Meijia Gu
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Zixin Deng
- State Key Laboratory of Microbial MetabolismSchool of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
| | - Ran Liu
- State Key Laboratory of Microbial MetabolismSchool of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
- Key Laboratory of Quantitative Synthetic BiologyShenzhen Institute of Synthetic BiologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Xinghuan Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Biological RepositoriesHuman Genetic Resource Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
- Medical Research InstituteFrontier Science Center of Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Tiangang Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryMinistry of Education and School of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Wuhan Hesheng Technology Co., LtdWuhan430074China
- State Key Laboratory of Microbial MetabolismSchool of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
- Department of UrologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| |
Collapse
|
253
|
Wang L, Wang P, Liu B, Zhang H, Wei CC, Xiong M, Luo G, Wang M. LncRNA MEG3 Inhibits the Epithelial-mesenchymal Transition of Bladder Cancer Cells through the Snail/E-cadherin Axis. Curr Med Sci 2024; 44:726-734. [PMID: 38990449 DOI: 10.1007/s11596-024-2895-x] [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: 07/26/2023] [Accepted: 04/28/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE This study aimed to investigate the role of the long noncoding RNA (lncRNA) maternally expressed gene 3 (MEG3) in the epithelial-mesenchymal transition (EMT) of bladder cancer cells and the potential mechanisms. METHODS Cell invasion, migration, and wound healing assays were conducted to assess the effects of MEG3 on the invasive and migratory capabilities of bladder cancer cells. The expression levels of E-cadherin were measured using Western blotting, RT-qPCR, and dual luciferase reporter assays. RNA immunoprecipitation and pull-down assays were performed to investigate the interactions between MEG3 and its downstream targets. RESULTS MEG3 suppressed the invasion and migration of bladder cancer cells and modulated the transcription of E-cadherin. The binding of MEG3 to the zinc finger region of the transcription factor Snail prevented its ability to transcriptionally repress E-cadherin. Additionally, MEG3 suppressed the phosphorylation of extracellular regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), and P38, thereby decreasing the expression of Snail and stimulating the expression of E-cadherin. CONCLUSION MEG3 plays a vital role in suppressing the EMT in bladder cancer cells, indicating its potential as a promising therapeutic target for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bing Liu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Cheng Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Luo
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China.
| | - Miao Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
254
|
Chang CY, Pearce G, Betaneli V, Kapustsenka T, Hosseini K, Fischer-Friedrich E, Corbeil D, Karbanová J, Taubenberger A, Dahncke B, Rauner M, Furesi G, Perner S, Rost F, Jessberger R. The F-actin bundler SWAP-70 promotes tumor metastasis. Life Sci Alliance 2024; 7:e202302307. [PMID: 38760173 PMCID: PMC11101836 DOI: 10.26508/lsa.202302307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
Dynamic rearrangements of the F-actin cytoskeleton are a hallmark of tumor metastasis. Thus, proteins that govern F-actin rearrangements are of major interest for understanding metastasis and potential therapies. We hypothesized that the unique F-actin binding and bundling protein SWAP-70 contributes importantly to metastasis. Orthotopic, ectopic, and short-term tail vein injection mouse breast and lung cancer models revealed a strong positive dependence of lung and bone metastasis on SWAP-70. Breast cancer cell growth, migration, adhesion, and invasion assays revealed SWAP-70's key role in these metastasis-related cell features and the requirement for SWAP-70 to bind F-actin. Biophysical experiments showed that tumor cell stiffness and deformability are negatively modulated by SWAP-70. Together, we present a hitherto undescribed, unique F-actin modulator as an important contributor to tumor metastasis.
Collapse
Affiliation(s)
- Chao-Yuan Chang
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Glen Pearce
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Viktoria Betaneli
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tatsiana Kapustsenka
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kamran Hosseini
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | | | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Taubenberger
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Björn Dahncke
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Giulia Furesi
- Department of Medicine III and Center for Healthy Aging, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sven Perner
- Institute of Pathology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
- Institute of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Rolf Jessberger
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
255
|
Chang Y, Fu Q, Lu Z, Jin Q, Jin T, Zhang M. Ginsenoside Rg3 combined with near-infrared photothermal reversal of multidrug resistance in breast cancer MCF-7/ADR cells. Food Sci Nutr 2024; 12:5750-5761. [PMID: 39139957 PMCID: PMC11317707 DOI: 10.1002/fsn3.4205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 08/15/2024] Open
Abstract
Adriamycin (ADR) is a frequently employed chemotherapeutic agent for the management of breast cancer. Nevertheless, multidrug resistance (MDR) can impair its therapeutic efficacy in breast cancer. MDR is characterized by increased expression of the P-glycoprotein (P-gp) efflux pump, up-regulation of anti-apoptotic proteins, and downregulation of pro-apoptotic proteins. Consequently, inhibition of ATP-binding cassette (ABC) transporter proteins has been deemed the most efficacious approach to overcome MDR. In this study, we used MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide), Western blots, flow cytometry, immunofluorescence, and constructed xenograft tumors to investigate whether ginsenoside Rg3-near-infrared photothermal (Rg3-NIR) combination reversed multidrug resistance in MCF-7/ADR breast cancer. In vivo and in vitro experiments, the results showed that Rg3-NIR co-treatment was effective in inducing the apoptosis of MCF-7/ADR breast cancer cells. This was achieved by reversing the expression of drug resistance-associated proteins, while also inhibiting cell proliferation, migration, and epithelial-mesenchymal transition (EMT) processes via attenuation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway transduction. Ginsenoside Rg3 combined with near-infrared photothermal therapy (NIR) effectively reverses multidrug resistance in breast cancer MCF-7/ADR cells, providing a new therapeutic strategy for breast cancer drug resistance.
Collapse
Affiliation(s)
- Ying Chang
- Department of Ultrasound MedicineAffiliated Hospital of Yanbian UniversityYanjiChina
- Department of Pathology and Cancer Research CenterYanbian University Medical CollegeYanjiChina
- Key Laboratory of the Science and Technology, Department of Jilin ProvinceYanjiChina
| | - Qiang Fu
- Department of Ultrasound MedicineAffiliated Hospital of Yanbian UniversityYanjiChina
- Department of Pathology and Cancer Research CenterYanbian University Medical CollegeYanjiChina
- Key Laboratory of the Science and Technology, Department of Jilin ProvinceYanjiChina
| | - Zhongqi Lu
- Department of Ultrasound MedicineAffiliated Hospital of Yanbian UniversityYanjiChina
- Department of Pathology and Cancer Research CenterYanbian University Medical CollegeYanjiChina
- Key Laboratory of the Science and Technology, Department of Jilin ProvinceYanjiChina
| | - Quanxin Jin
- Department of Immunology and Pathogenic BiologyYanbian University Medical CollegeYanjiChina
| | - Tiefeng Jin
- Department of Pathology and Cancer Research CenterYanbian University Medical CollegeYanjiChina
- Key Laboratory of the Science and Technology, Department of Jilin ProvinceYanjiChina
| | - Meihua Zhang
- Department of Ultrasound MedicineAffiliated Hospital of Yanbian UniversityYanjiChina
- Department of Pathology and Cancer Research CenterYanbian University Medical CollegeYanjiChina
- Key Laboratory of the Science and Technology, Department of Jilin ProvinceYanjiChina
| |
Collapse
|
256
|
Liu X, Gao X, Yang Y, Yang D, Guo Q, Li L, Liu S, Cong W, Lu S, Hou L, Wang B, Li N. EVA1A reverses lenvatinib resistance in hepatocellular carcinoma through regulating PI3K/AKT/p53 signaling axis. Apoptosis 2024; 29:1161-1184. [PMID: 38743191 DOI: 10.1007/s10495-024-01967-0] [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] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
Lenvatinib is a commonly used first-line drug for the treatment of advanced hepatocellular carcinoma (HCC). However, its clinical efficacy is limited due to the drug resistance. EVA1A was a newly identified tumor suppressor, nevertheless, the impact of EVA1A on resistance to lenvatinib treatment in HCC and the potential molecular mechanisms remain unknown. In this study, the expression of EVA1A in HCC lenvatinib-resistant cells is decreased and its low expression was associated with a poor prognosis of HCC. Overexpression of EVA1A reversed lenvatinib resistance in vitro and in vivo, as demonstrated by its ability to promote cell apoptosis and inhibit cell proliferation, invasion, migration, EMT, and tumor growth. Silencing EVA1A in lenvatinib-sensitive parental HCC cells exerted the opposite effect and induced resistance to lenvatinib. Mechanistically, upregulated EVA1A inhibited the PI3K/AKT/MDM2 signaling pathway, resulting in a reduced interaction between MDM2 and p53, thereby stabilizing p53 and enhancing its antitumor activity. In addition, upregulated EVA1A suppressed the PI3K/AKT/mTOR signaling pathway and promoted autophagy, leading to the degradation of mutant p53 and attenuating its oncogenic impact. On the contrary, loss of EVA1A activated the PI3K/AKT/MDM2 signaling pathway and inhibited autophagy, promoting p53 proteasomal degradation and mutant p53 accumulation respectively. These findings establish a crucial role of EVA1A loss in driving lenvatinib resistance involving a mechanism of modulating PI3K/AKT/p53 signaling axis and suggest that upregulating EVA1A is a promising therapeutic strategy for alleviating resistance to lenvatinib, thereby improving the efficacy of HCC treatment.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Quinolines/pharmacology
- Quinolines/therapeutic use
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Signal Transduction/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Animals
- Cell Line, Tumor
- Mice
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Mice, Nude
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Xenograft Model Antitumor Assays
- Mice, Inbred BALB C
- Proto-Oncogene Proteins c-mdm2/metabolism
- Proto-Oncogene Proteins c-mdm2/genetics
- Female
Collapse
Affiliation(s)
- Xiaokun Liu
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Xiao Gao
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Yuling Yang
- Department of Infectious Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Di Yang
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Qingming Guo
- Clinical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Lianhui Li
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Shunlong Liu
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wanxin Cong
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Sen Lu
- Department of Medical Laboratory, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Lin Hou
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Bin Wang
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Ning Li
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China.
| |
Collapse
|
257
|
Yuan L, Meng Y, Xiang J. KLF4 Induces Colorectal Cancer by Promoting EMT via STAT3 Activation. Dig Dis Sci 2024; 69:2841-2855. [PMID: 38816600 DOI: 10.1007/s10620-024-08473-y] [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: 02/11/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Krüppel-like factor 4 (KLF4) has been demonstrated to exert a pro-carcinogenic effect in solid tissues. However, the precise biological function and underlying mechanisms in colorectal cancer (CRC) remains elucidated. AIMS To investigate whether KLF4 participates in the proliferation and invasion of CRC. METHODS The expression of KLF4 was investigated using immunohistochemistry and immunoblotting. The clinical significance of KLF4 was evaluated. Furthermore, the effect of inhibiting or overexpressing KLF4 on tumor was examined. Immunoblotting and qPCR were used to detect Epithelial-mesenchymal transition-related proteins levels. Additionally, the molecular function of KLF4 is related to the STAT3 signaling pathway and was determined through JASPAR, GSEA analysis, and in vitro experiments. RESULTS KLF4 exhibits down-regulated expression in CRC and is part of the vessel invasion, TNM stage, and worse prognosis. In vitro studies have shown that KLF4 promotes cellular proliferation and invasion, as well as EMT processes. Xenograft tumor models confirmed the oncogenic role of KLF4 in nude mice. Furthermore, GSEA and JASPAR databases analysis reveal that the binding of KLF4 to the signal transducer and activator of transcription 3 (STAT3) promoter site induces activation of p-STAT3 signaling. Subsequent targeting of STAT3 confirmed its pivotal role in mediating the oncogenic effects exerted by KLF4. CONCLUSION The study suggests that KLF4 activates STAT3 signaling, inducing epithelial-mesenchymal transition, thereby promoting CRC progression.
Collapse
Affiliation(s)
- Lebin Yuan
- Department of Nail and Breast Surgery, Affiliated Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang Center Hospital, Xiangyang, Hubei, China
| | - Yanqiu Meng
- Oncology Department, First Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiajia Xiang
- Molecular Centre Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
258
|
Wang S, Zheng C, Zhao L, Jiang H, Zheng X. Benefits of adjuvant treatment with the Pingxiao capsule in patients with early breast cancer: A single‑center retrospective cohort study. Oncol Lett 2024; 28:366. [PMID: 38933812 PMCID: PMC11200151 DOI: 10.3892/ol.2024.14499] [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: 01/19/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Early breast cancer (EBC) is cancer that has not spread beyond the breast or the axillary lymph nodes. The present retrospective cohort study investigated the efficacy and safety of the Pingxiao capsule (PXC), which contains a formula of traditional Chinese herbs, as adjuvant therapy in patients with EBC in a single Chinese academic medical center. Patients with EBC who had received surgery and chemotherapy were analyzed and divided into the PXC and non-PXC groups. Disease-free survival (DFS) time, overall survival (OS) time, demographic characteristics and adverse events were examined. Kaplan-Meier survival curves were used to compare the differences in DFS and OS. A total of 371 participants with a median age of 54 years were included in this study. The median DFS time of all patients was 101 months. The overall DFS rate was 72.1% in the PXC group compared with 63.6% in the non-PXC group. For women with hormone receptor-negative tumors, the DFS rate in the PXC group was significantly higher than that in the non-PXC group, irrespective of node status. Adjuvant treatment with PXC for ≥3 months was associated with significantly longer median DFS time compared with that in the non-PXC group. In addition, the incidence of neutropenia rated to be grade 2 or higher was significantly lower in the PXC group compared with that in the control group, and a markedly, but non-significantly, lower prevalence of nausea was observed in PXC group (0 vs. 4.1%). In conclusion, PXC as an adjuvant therapy along with chemotherapy is associated with prolonged DFS times in patients with EBC when compared with chemotherapy alone. The therapeutic value of combined PXC and systemic chemotherapy should be further elucidated by rigorous prospective clinical trials.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Caiwei Zheng
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Lei Zhao
- Science Experiment Center, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Haiyang Jiang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinyu Zheng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Lab 1, Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
259
|
Zhang D, Luo Q, Xiao L, Chen X, Yang S, Zhang S. Exosomes derived from gastric cancer cells promote phenotypic transformation of hepatic stellate cells and affect the malignant behavior of gastric cancer cells. J Cancer Res Ther 2024; 20:1157-1164. [PMID: 39206977 DOI: 10.4103/jcrt.jcrt_749_23] [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: 04/03/2023] [Accepted: 03/01/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE This study aimed to evaluate the effect of exosomes derived from gastric cancer cells on the phenotypic transformation of hepatic stellate cells (HSCs) and the effect of HSC activation on the malignant behavior of gastric cancer cells, including its molecular mechanism. METHODS Exosomes derived from the human gastric adenocarcinoma cell line AGS were extracted and purified by polymer precipitation and ultrafiltration, respectively. The exosomes' morphologic characteristics were observed using transmission electron microscopy, particle size was determined through nanoparticle-tracking analysis, and marker proteins were detected using western blotting. Exosome uptake by LX-2 HSCs was observed through fluorescence-based tracing. Reverse transcription quantitative PCR (RT-qPCR) was used to detect the messenger RNA (mRNA) expression of alpha-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP). Using functional assays, the effects of LX-2 HSC activation on the biological behavior of malignant gastric cancer cells were evaluated. The effects of LX-2 HSC activation on the protein expression of epithelial-mesenchymal transition (EMT)-related genes and β-catenin were evaluated via western blotting. RESULTS The extracted particles conformed to the definitions of exosomes and were thus considered gastric cancer cell-derived exosomes. Fluorescence-based tracing successfully demonstrated that exosomes were enriched in LX-2 HSCs. RT-qPCR revealed that the mRNA expression of the cancer-associated fibroblast markers α-SMA and FAP was significantly increased. LX-2 HSC activation considerably enhanced gastric cancer cell proliferation, invasion, and migration. Western blotting showed that the expression of the EMT-related epithelial marker E-cadherin was significantly downregulated, whereas the expression of interstitial markers (N-cadherin and vimentin) and β-catenin was remarkably upregulated in gastric cancer cells. CONCLUSION Exosomes derived from gastric cancer cells promoted phenotypic transformation of HSCs and activated HSCs to become tumor-associated fibroblasts. Gastric cancer cell-derived cells significantly enhanced gastric cancer cell proliferation, invasion, and migration after HSC activation, which may promote EMT of gastric cancer cells through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Donghuan Zhang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Department of Oncology Medicine, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Qiong Luo
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Lirong Xiao
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiangqi Chen
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Sheng Yang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Suyun Zhang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
260
|
Nian Z, Wang D, Wang H, Liu W, Ma Z, Yan J, Cao Y, Li J, Zhao Q, Liu Z. Single-cell RNA-seq reveals the transcriptional program underlying tumor progression and metastasis in neuroblastoma. Front Med 2024; 18:690-707. [PMID: 39014137 DOI: 10.1007/s11684-024-1081-7] [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: 01/08/2024] [Accepted: 04/18/2024] [Indexed: 07/18/2024]
Abstract
Neuroblastoma (NB) is one of the most common childhood malignancies. Sixty percent of patients present with widely disseminated clinical signs at diagnosis and exhibit poor outcomes. However, the molecular mechanisms triggering NB metastasis remain largely uncharacterized. In this study, we generated a transcriptomic atlas of 15 447 NB cells from eight NB samples, including paired samples of primary tumors and bone marrow metastases. We used time-resolved analysis to chart the evolutionary trajectory of NB cells from the primary tumor to the metastases in the same patient and identified a common 'starter' subpopulation that initiates tumor development and metastasis. The 'starter' population exhibited high expression levels of multiple cell cycle-related genes, indicating the important role of cell cycle upregulation in NB tumor progression. In addition, our evolutionary trajectory analysis demonstrated the involvement of partial epithelial-to-mesenchymal transition (p-EMT) along the metastatic route from the primary site to the bone marrow. Our study provides insights into the program driving NB metastasis and presents a signature of metastasis-initiating cells as an independent prognostic indicator and potential therapeutic target to inhibit the initiation of NB metastasis.
Collapse
Affiliation(s)
- Zhe Nian
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hao Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wenxu Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhenyi Ma
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jie Yan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanna Cao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jie Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Zhe Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
261
|
Campos-Fernández E, Alqualo NO, Vaz ER, Rodrigues CM, Alonso-Goulart V. Unveiling the characteristics of D4 and R4 aptamers for their future use in prostate cancer clinical practice. Biophys Chem 2024; 311:107259. [PMID: 38763045 DOI: 10.1016/j.bpc.2024.107259] [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: 02/25/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
The DNA and RNA aptamers D4 and R4, respectively, emerged from the modification of PC-3 cell-binding aptamer A4. Our objective was to characterize the aptamers in silico and in vitro and begin to identify their target molecules. We represented their structures using computational algorithms; evaluated their binding to several prostate cell lines and their effects on the viability and migration of these cells; and determined their dissociation constant by flow cytometry. We analyzed circulating prostate tumor cells from patients using D4, R4, anti-CD133 and anti-CD44. Finally, the target proteins of both aptamers were precipitated and identified by mass spectrometry to simulate their in silico docking. The aptamers presented similar structures and bound to prostate tumor cells without modifying the cellular parameters studied, but with different affinities. The ligand cells for both aptamers were CD44+, indicating that they could identify cells in the mesenchymal stage of the metastatic process. The possible target proteins NXPE1, ADAM30, and MUC6 need to be further studied to better understand their interaction with the aptamers. These results support the development of new assays to determine the clinical applications of D4 and R4 aptamers in prostate cancer.
Collapse
Affiliation(s)
- Esther Campos-Fernández
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Nathalia Oliveira Alqualo
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Emília Rezende Vaz
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Cláudia Mendonça Rodrigues
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Vivian Alonso-Goulart
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
262
|
Tang J, Chen Y, Wang C, Xia Y, Yu T, Tang M, Meng K, Yin L, Yang Y, Shen L, Xing H, Mao X. The role of mesenchymal stem cells in cancer and prospects for their use in cancer therapeutics. MedComm (Beijing) 2024; 5:e663. [PMID: 39070181 PMCID: PMC11283587 DOI: 10.1002/mco2.663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are recruited by malignant tumor cells to the tumor microenvironment (TME) and play a crucial role in the initiation and progression of malignant tumors. This role encompasses immune evasion, promotion of angiogenesis, stimulation of cancer cell proliferation, correlation with cancer stem cells, multilineage differentiation within the TME, and development of treatment resistance. Simultaneously, extensive research is exploring the homing effect of MSCs and MSC-derived extracellular vesicles (MSCs-EVs) in tumors, aiming to design them as carriers for antitumor substances. These substances are targeted to deliver antitumor drugs to enhance drug efficacy while reducing drug toxicity. This paper provides a review of the supportive role of MSCs in tumor progression and the associated molecular mechanisms. Additionally, we summarize the latest therapeutic strategies involving engineered MSCs and MSCs-EVs in cancer treatment, including their utilization as carriers for gene therapeutic agents, chemotherapeutics, and oncolytic viruses. We also discuss the distribution and clearance of MSCs and MSCs-EVs upon entry into the body to elucidate the potential of targeted therapies based on MSCs and MSCs-EVs in cancer treatment, along with the challenges they face.
Collapse
Affiliation(s)
- Jian Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Yu Chen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Medical Affairs, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Chunhua Wang
- Department of Clinical LaboratoryXiangyang No. 1 People's HospitalHubei University of MedicineXiangyangHubei ProvinceChina
| | - Ying Xia
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Tingyu Yu
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Mengjun Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Kun Meng
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Lijuan Yin
- State Key Laboratory of Food Nutrition and SafetyKey Laboratory of Industrial MicrobiologyMinistry of EducationTianjin Key Laboratory of Industry MicrobiologyNational and Local United Engineering Lab of Metabolic Control Fermentation TechnologyChina International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal ChemistryCollege of BiotechnologyTianjin University of Science & TechnologyTianjinChina
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and ImmunityNational Clinical Research Center for Infectious DiseaseState Key Discipline of Infectious DiseaseShenzhen Third People's HospitalSecond Hospital Affiliated to Southern University of Science and TechnologyShenzhenChina
| | - Liang Shen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Hui Xing
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| | - Xiaogang Mao
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| |
Collapse
|
263
|
Shi R, Yu R, Lian F, Zheng Y, Feng S, Li C, Zheng X. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35:623-637. [PMID: 38718070 DOI: 10.1097/cad.0000000000001612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Heat shock protein 47 (HSP47) serves as an endoplasmic reticulum residing collagen-specific chaperone and plays an important role in collagen biosynthesis and structural assembly. HSP47 is encoded by the SERPINH1 gene, which is located on chromosome 11q13.5, one of the most frequently amplified regions in human cancers. The expression of HSP47 is regulated by multiple cellular factors, including cytokines, transcription factors, microRNAs, and circular RNAs. HSP47 is frequently upregulated in a variety of cancers and plays an important role in tumor progression. HSP47 promotes tumor stemness, angiogenesis, growth, epithelial-mesenchymal transition, and metastatic capacity. HSP47 also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Inhibition of HSP47 expression has antitumor effects, suggesting that targeting HSP47 is a feasible strategy for cancer treatment. In this review, we highlight the function and expression of regulatory mechanisms of HSP47 in cancer progression and point out the potential development of therapeutic strategies in targeting HSP47 in the future.
Collapse
Affiliation(s)
- Run Shi
- School of Medicine, Pingdingshan University, Pingdingshan, China
| | | | | | | | | | | | | |
Collapse
|
264
|
Mincy C, Revelt L, Carter K, Reed D, Joy A. Unique Cohorts of Salivary Gland Cancer Cells as an in-vitro Model of Circulating Tumor Cells. J Maxillofac Oral Surg 2024; 23:896-908. [PMID: 39118911 PMCID: PMC11303642 DOI: 10.1007/s12663-024-02250-0] [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: 02/06/2024] [Accepted: 06/09/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction The characterization of circulating tumor cells (CTC) and circulating tumor microemboli (CTM) has emerged as both a challenge to the standard view of metastasis, and as a valuable means for understanding genotypic and phenotypic variability shown even within the same cancer type. However, in the case of salivary gland neoplasms, limited data are available for the role that CTCs and CTMs play in metastasis and secondary tumor formation.ru.AQ1 In response to this, we propose that similarities between in vitro clusters of cultured salivary gland cancer cells may act as a surrogate model for in vivo CTCs and CTMs isolated from patients. Materials and Methods Using techniques in immunofluorescence, immunoblotting, and 2-dimensional migration, we isolated and characterized a group of cohort cells from a commercially available cell line (HTB-41). Results: Here, cells exhibited a hybrid phenotype with simultaneous expression of both epithelial and mesenchymal markers (E-cadherin, vimentin, and α-SMA). Cohort cells also exhibited increased migration in comparison to parental cells. Conclusion Data suggest that these isolated cell clusters may fucntion as a potential in vitro model of CTCs and CTMs.
Collapse
Affiliation(s)
- Callie Mincy
- Department of Growth, Development and Structure, Southern Illinois University School of Dental Medicine, Alton, IL USA
- Department of Biological Sciences, College of Arts and Sciences, Southern Illinois University Edwardsville, Edwardsville, IL USA
| | - Luke Revelt
- Department of Growth, Development and Structure, Southern Illinois University School of Dental Medicine, Alton, IL USA
| | - Kathryn Carter
- Department of Growth, Development and Structure, Southern Illinois University School of Dental Medicine, Alton, IL USA
| | - Donald Reed
- Department of Growth, Development and Structure, Southern Illinois University School of Dental Medicine, Alton, IL USA
| | - Anita Joy
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Houston School of Dentistry, 7500 Cambridge Ave., Houston, TX USA
| |
Collapse
|
265
|
Yueh PF, Chiang CS, Tsai IJ, Tseng YL, Chen HR, Lan KL, Hsu FT. A multifunctional PEGylated liposomal-encapsulated sunitinib enhancing autophagy, immunomodulation, and safety in renal cell carcinoma. J Nanobiotechnology 2024; 22:459. [PMID: 39085911 PMCID: PMC11293195 DOI: 10.1186/s12951-024-02664-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Sunitinib is a multikinase inhibitor used to treat patients with advanced renal cell carcinoma (RCC). However, sunitinib toxicity makes it a double-edged sword. Potent immune modulation by sunitinib extends to nuclear interactions. To address these issues, there is an urgent need for delivery vectors suitable for sunitinib treatment. METHODS We developed PEGylated liposomes as delivery vectors to precisely target sunitinib (lipo-sunitinib) to RCC tumors. Further investigations, including RNA sequencing (RNA-seq), were performed to evaluate transcriptomic changes in these pathways. DiI/DiR-labeled lipo-sunitinib was used for the biodistribution analysis. Flow cytometry and immunofluorescence (IF) were used to examine immune modulation in orthotopic RCC models. RESULTS The evaluation of results indicated that lipo-sunitinib precisely targeted the tumor site to induce autophagy and was readily taken up by RCC tumor cells. In addition, transcriptomic assays revealed that following lipo-sunitinib treatment, autophagy, antigen presentation, cytokine, and chemokine production pathways were upregulated, whereas the epithelial-mesenchymal transition (EMT) pathway was downregulated. In vivo data provided evidence supporting the inhibitory effect of lipo-sunitinib on RCC tumor progression and metastasis. Flow cytometry further demonstrated that liposunitinib increased the infiltration of effector T cells (Teffs) and conventional type 1 dendritic cells (cDC1s) into the tumor. Furthermore, systemic immune organs such as the tumor-draining lymph nodes, spleen, and bone marrow exhibited upregulated anticancer immunity following lipo-sunitinib treatment. CONCLUSION Our findings demonstrated that lipo-sunitinib is distributed at the RCC tumor site, concurrently inducing potent autophagy, elevating antigen presentation, activating cytokine and chemokine production pathways, and downregulating EMT in RCC cells. This comprehensive approach significantly enhanced tumor inhibition and promoted anticancer immune modulation.
Collapse
Affiliation(s)
- Po-Fu Yueh
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 6th Floor, Shouren Building, No. 155, Section 2, Linong Street, Beitou District, Taipei, 112, Taiwan, ROC
| | - Chih-Sheng Chiang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | - I-Jung Tsai
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | | | - He-Ru Chen
- Taiwan Liposome Company, Ltd., Taipei, Taiwan, ROC
| | - Keng-Li Lan
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 6th Floor, Shouren Building, No. 155, Section 2, Linong Street, Beitou District, Taipei, 112, Taiwan, ROC.
- Department of Heavy Ion and Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Fei-Ting Hsu
- Department of Biology Science and Technology, China Medical University, 7F, Research Building, No. 100, Jingmao 1st Rd., Beitun Dist., Taichung City, 406, Taiwan, ROC.
| |
Collapse
|
266
|
Li Y, Wang M, Jiang L, Jia J, Pan F, Li W, Wang B, Huang K, Luo J. SIPA1 promotes epithelial-mesenchymal transition in colorectal cancer through STAT3 activation. Heliyon 2024; 10:e34527. [PMID: 39130435 PMCID: PMC11315193 DOI: 10.1016/j.heliyon.2024.e34527] [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: 01/05/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Colorectal cancer (CRC) is the third leading cancer type worldwide and accounts for the second highest rate of cancer-related mortality. Liver metastasis significantly contributes to the mortality associated with CRC, but the fundamental mechanisms behind it remain unclear. Signal-induced proliferation-associated protein 1 (SIPA1), a GTPase activating protein, has been shown to promote metastasis in breast cancer. In this study, our objective was to explore the role of SIPA1 in regulating epithelial-mesenchymal transition (EMT) in CRC. The analysis of The Cancer Genome Atlas (TCGA) database revealed that the expression level of SIPA1 mRNA was notably upregulated and exhibited a positively correlated with EMT and STAT3 signaling pathways in CRC. Knockdown of SIPA1 impairs CRC cell proliferation and migration. Further studies on the reliance of SIPA1 on STAT3 signaling for EMT regulation have shown that SIPA1 stimulates the activation of STAT3, resulting in its nuclear translocation. The co-treatment of overexpressed SIPA1 with the STAT3 inhibitor STTITA has shown that SIPA1 regulates the expression of EMT-related markers through STAT3. Our study indicate that SIPA1 promotes CRC metastasis by activating the STAT3 signaling pathway, underscoring the potential of SIPA1 as a therapeutic target for metastatic CRC patients.
Collapse
Affiliation(s)
- Youjian Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Mengjie Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Jiang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Jiehong Jia
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Fei Pan
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wen Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Bochu Wang
- Biomedical and Health Engineering Laboratory, Chongqing University, Chongqing, China
| | - Ke Huang
- College of Landscape Architecture and Life Science/Institute of Special Plants, Chongqing University of Arts and Sciences, Yongchuan, Chongqing, China
| | - Jie Luo
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
- Biomedical and Health Engineering Laboratory, Chongqing University, Chongqing, China
| |
Collapse
|
267
|
Liu F, Chen J, Li K, Li H, Zhu Y, Zhai Y, Lu B, Fan Y, Liu Z, Chen X, Jia X, Dong Z, Liu K. Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches. Mol Cancer 2024; 23:148. [PMID: 39048965 PMCID: PMC11270804 DOI: 10.1186/s12943-024-02046-3] [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: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
Collapse
Affiliation(s)
- Fangfang Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jingyu Chen
- Department of Pediatric Medicine, School of Third Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Kai Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Haochen Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiyi Zhu
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yubo Zhai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanle Fan
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Ziyue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojie Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xuechao Jia
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Zigang Dong
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Kangdong Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
268
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Zhong J. LINC00518: a key player in tumor progression and clinical outcomes. Front Immunol 2024; 15:1419576. [PMID: 39108268 PMCID: PMC11300200 DOI: 10.3389/fimmu.2024.1419576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/08/2024] [Indexed: 09/17/2024] Open
Abstract
Long non-coding RNAs (lncRNAs), defined as RNA molecules exceeding 200 nucleotides in length, have been implicated in the regulation of various biological processes and the progression of tumors. Among them, LINC00518, a recently identified lncRNA encoded by a gene located on chromosome 6p24.3, consists of three exons and is predicted to positively regulate the expression of specific genes. LINC00518 has emerged as a key oncogenic lncRNA in multiple cancer types. It exerts its tumor-promoting effects by modulating the expression of several target genes, primarily through acting as a sponge for microRNAs (miRNAs). Additionally, LINC00518 influences critical signaling pathways, including the Wnt/β-catenin, JAK/STAT, and integrin β3/FAK pathways. Elevated levels of LINC00518 in tumor tissues are associated with increased tumor size, advanced clinical stage, metastasis, and poor survival prognosis. This review provides a comprehensive summary of the genetic characteristics, expression patterns, biological functions, and underlying mechanisms of LINC00518 in human diseases.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
269
|
Subbalakshmi AR, Ramisetty S, Mohanty A, Pareek S, Do D, Shrestha S, Khan A, Talwar N, Tan T, Vishnubhotla P, Singhal SS, Salgia R, Kulkarni P. Phenotypic Plasticity and Cancer: A System Biology Perspective. J Clin Med 2024; 13:4302. [PMID: 39124569 PMCID: PMC11313222 DOI: 10.3390/jcm13154302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a major axis of phenotypic plasticity not only in diseased conditions such as cancer metastasis and fibrosis but also during normal development and wound healing. Yet-another important axis of plasticity with metastatic implications includes the cancer stem cell (CSCs) and non-CSC transitions. However, in both processes, epithelial (E) and mesenchymal (M) phenotypes are not merely binary states. Cancer cells acquire a spectrum of phenotypes with traits, properties, and markers of both E and M phenotypes, giving rise to intermediary hybrid (E/M) phenotypes. E/M cells play an important role in tumor initiation, metastasis, and disease progression in multiple cancers. Furthermore, the hybrid phenotypes also play a major role in causing therapeutic resistance in cancer. Here, we discuss how a systems biology perspective on the problem, which is implicit in the 'Team Medicine' approach outlined in the theme of this Special Issue of The Journal of Clinical Medicine and includes an interdisciplinary team of experts, is more likely to shed new light on EMT in cancer and help us to identify novel therapeutics and strategies to target phenotypic plasticity in cancer.
Collapse
Affiliation(s)
- Ayalur Raghu Subbalakshmi
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Sravani Ramisetty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Siddhika Pareek
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Dana Do
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Sagun Shrestha
- Department of Medical Oncology and Therapeutics Research, City of Hope Phoenix, Goodyear, AZ 85338, USA
| | - Ajaz Khan
- Department of Medical Oncology and Therapeutics Research, City of Hope Chicago, Zion, IL 60099, USA
| | - Neel Talwar
- Department of Medical Oncology and Therapeutics Research, City of Hope San Bernardino Road, Upland, CA 91786, USA
| | - Tingting Tan
- Department of Medical Oncology and Therapeutics Research, City of Hope Avocado Avenue, Newport Beach, CA 92660, USA
| | - Priya Vishnubhotla
- Department of Medical Oncology and Therapeutics Research, City of Hope Atlanta, Newnan, GA 30265, USA
| | - Sharad S. Singhal
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.R.S.)
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
270
|
Liang Y, Wu G, Tan J, Xiao X, Yang L, Saw PE. Targeting NETosis: nature's alarm system in cancer progression. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:28. [PMID: 39143953 PMCID: PMC11322967 DOI: 10.20517/cdr.2024.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024]
Abstract
Neutrophils are recognized active participants in inflammatory responses and are intricately linked to cancer progression. In response to inflammatory stimuli, neutrophils become activated, releasing neutrophils extracellular traps (NETs) for the capture and eradication of pathogens, a phenomenon termed NETosis. With a deeper understanding of NETs, there is growing evidence supporting their role in cancer progression and their involvement in conferring resistance to various cancer therapies, especially concerning tumor reactions to chemotherapy, radiation therapy (RT), and immunotherapy. This review summarizes the roles of NETs in the tumor microenvironment (TME) and their mechanisms of neutrophil involvement in the host defense. Additionally, it elucidates the mechanisms through which NETs promote tumor progression and their role in cancer treatment resistance, highlighting their potential as promising therapeutic targets in cancer treatment and their clinical applicability.
Collapse
Affiliation(s)
- Yixia Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
- Authors contributed equally
| | - Guo Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
- Authors contributed equally
| | - Jiabao Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Xiaoyun Xiao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China
| | - Linbin Yang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
| |
Collapse
|
271
|
Tian H, Tang L, Yang Z, Xiang Y, Min Q, Yin M, You H, Xiao Z, Shen J. Current understanding of functional peptides encoded by lncRNA in cancer. Cancer Cell Int 2024; 24:252. [PMID: 39030557 PMCID: PMC11265036 DOI: 10.1186/s12935-024-03446-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/09/2024] [Indexed: 07/21/2024] Open
Abstract
Dysregulated gene expression and imbalance of transcriptional regulation are typical features of cancer. RNA always plays a key role in these processes. Human transcripts contain many RNAs without long open reading frames (ORF, > 100 aa) and that are more than 200 bp in length. They are usually regarded as long non-coding RNA (lncRNA) which play an important role in cancer regulation, including chromatin remodeling, transcriptional regulation, translational regulation and as miRNA sponges. With the advancement of ribosome profiling and sequencing technologies, increasing research evidence revealed that some ORFs in lncRNA can also encode peptides and participate in the regulation of multiple organ tumors, which undoubtedly opens a new chapter in the field of lncRNA and oncology research. In this review, we discuss the biological function of lncRNA in tumors, the current methods to evaluate their coding potential and the role of functional small peptides encoded by lncRNA in cancers. Investigating the small peptides encoded by lncRNA and understanding the regulatory mechanisms of these functional peptides may contribute to a deeper understanding of cancer and the development of new targeted anticancer therapies.
Collapse
Affiliation(s)
- Hua Tian
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, China
| | - Lu Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Zihan Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China, 646000
| | - Yanxi Xiang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Qi Min
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Mengshuang Yin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Huili You
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China.
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China.
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, China.
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China.
| |
Collapse
|
272
|
Moro M, Balestrero FC, Grolla AA. Pericytes: jack-of-all-trades in cancer-related inflammation. Front Pharmacol 2024; 15:1426033. [PMID: 39086395 PMCID: PMC11288921 DOI: 10.3389/fphar.2024.1426033] [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: 04/30/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Pericytes, recognized as mural cells, have long been described as components involved in blood vessel formation, playing a mere supporting role for endothelial cells (ECs). Emerging evidence strongly suggests their multifaceted roles in tissues and organs. Indeed, pericytes exhibit a remarkable ability to anticipate endothelial cell behavior and adapt their functions based on the specific cells they interact with. Pericytes can be activated by pro-inflammatory stimuli and crosstalk with immune cells, actively participating in their transmigration into blood vessels. Moreover, they can influence the immune response, often sustaining an immunosuppressive phenotype in most of the cancer types studied. In this review, we concentrate on the intricate crosstalk between pericytes and immune cells in cancer, highlighting the primary evidence regarding pericyte involvement in primary tumor mass dynamics, their contributions to tumor reprogramming for invasion and migration of malignant cells, and their role in the formation of pre-metastatic niches. Finally, we explored recent and emerging pharmacological approaches aimed at vascular normalization, including novel strategies to enhance the efficacy of immunotherapy through combined use with anti-angiogenic drugs.
Collapse
Affiliation(s)
| | | | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
273
|
林 雅, 颜 崇, 洪 文, 蔡 成, 龚 小. [Role and mechanism of epithelial-mesenchymal transition in a rat model of bronchopulmonary dysplasia induced by hyperoxia exposure]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:765-773. [PMID: 39014955 PMCID: PMC11562037 DOI: 10.7499/j.issn.1008-8830.2312112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/31/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVES To investigate the role and mechanism of epithelial-mesenchymal transition (EMT) in a rat model of bronchopulmonary dysplasia (BPD). METHODS The experiment consisted of two parts. (1) Forty-eight preterm rats were randomly divided into a normoxia group and a hyperoxia group, with 24 rats in each group. The hyperoxia group was exposed to 85% oxygen to establish a BPD model, while the normoxia group was kept in room air at normal pressure. Lung tissue samples were collected on days 1, 4, 7, and 14 of the experiment. (2) Rat type II alveolar epithelial cells (RLE-6TN) were randomly divided into a normoxia group (cultured in air) and a hyperoxia group (cultured in 95% oxygen), and cell samples were collected 12, 24, and 48 hours after hyperoxia exposure. Hematoxylin-eosin staining was used to observe alveolarization in preterm rat lungs, and immunofluorescence was used to detect the co-localization of surfactant protein C (SPC) and α-smooth muscle actin (α-SMA) in preterm rat lung tissue and RLE-6TN cells. Quantitative real-time polymerase chain reaction and protein immunoblotting were used to detect the expression levels of EMT-related mRNA and proteins in preterm rat lung tissue and RLE-6TN cells. RESULTS (1) Compared with the normoxia group, the hyperoxia group showed blocked alveolarization and simplified alveolar structure after 7 days of hyperoxia exposure. Co-localization of SPC and α-SMA was observed in lung tissue, with decreased SPC expression and increased α-SMA expression in the hyperoxia group at 7 and 14 days of hyperoxia exposure compared to the normoxia group. In the hyperoxia group, the mRNA and protein levels of TGF-β1, α-SMA, and N-cadherin were increased, while the mRNA and protein levels of SPC and E-cadherin were decreased at 7 and 14 days of hyperoxia exposure compared to the normoxia group (P<0.05). (2) SPC and α-SMA was observed in RLE-6TN cells, with decreased SPC expression and increased α-SMA expression in the hyperoxia group at 24 and 48 hours of hyperoxia exposure compared to the normoxia group. Compared to the normoxia group, the mRNA and protein levels of SPC and E-cadherin in the hyperoxia group were decreased, while the mRNA and protein levels of TGF-β1, α-SMA, and E-cadherin in the hyperoxia group increased at 48 hours of hyperoxia exposure (P<0.05). CONCLUSIONS EMT disrupts the tight connections between alveolar epithelial cells in a preterm rat model of BPD, leading to simplified alveolar structure and abnormal development, and is involved in the development of BPD. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 765-773.
Collapse
|
274
|
Young KA, Wojdyla K, Lai T, Mulholland KE, Aldaz Casanova S, Antrobus R, Andrews SR, Biggins L, Mahler-Araujo B, Barton PR, Anderson KR, Fearnley GW, Sharpe HJ. The receptor protein tyrosine phosphatase PTPRK promotes intestinal repair and catalysis-independent tumour suppression. J Cell Sci 2024; 137:jcs261914. [PMID: 38904097 PMCID: PMC11298714 DOI: 10.1242/jcs.261914] [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: 12/18/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
PTPRK is a receptor tyrosine phosphatase that is linked to the regulation of growth factor signalling and tumour suppression. It is stabilized at the plasma membrane by trans homophilic interactions upon cell-cell contact. PTPRK regulates cell-cell adhesion but is also reported to regulate numerous cancer-associated signalling pathways. However, the signalling mechanism of PTPRK remains to be determined. Here, we find that PTPRK regulates cell adhesion signalling, suppresses invasion and promotes collective, directed migration in colorectal cancer cells. In vivo, PTPRK supports recovery from inflammation-induced colitis. In addition, we confirm that PTPRK functions as a tumour suppressor in the mouse colon and in colorectal cancer xenografts. PTPRK regulates growth factor and adhesion signalling, and suppresses epithelial to mesenchymal transition (EMT). Contrary to the prevailing notion that PTPRK directly dephosphorylates EGFR, we find that PTPRK regulation of both EGFR and EMT is independent of its catalytic function. This suggests that additional adaptor and scaffold functions are important features of PTPRK signalling.
Collapse
Affiliation(s)
| | | | - Tiffany Lai
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | - Robin Antrobus
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | | | - Laura Biggins
- Bioinformatics, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Philippa R. Barton
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Keith R. Anderson
- Molecular biology department, Genentech, South San Francisco, CA 94080, USA
| | | | - Hayley J. Sharpe
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
275
|
Hsu CY, Pallathadka H, Gupta J, Ma H, Al-Shukri HHK, Kareem AK, Zwamel AH, Mustafa YF. Berberine and berberine nanoformulations in cancer therapy: Focusing on lung cancer. Phytother Res 2024. [PMID: 38994919 DOI: 10.1002/ptr.8255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/02/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024]
Abstract
Lung cancer is the second most prevalent cancer and ranks first in cancer-related death worldwide. Due to the resistance development to conventional cancer therapy strategies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, various natural products and their extracts have been revealed as alternatives. Berberine (BBR), which is present in the stem, root, and bark of various trees, could exert anticancer activities by regulating tumor cell proliferation, apoptosis, autophagy, metastasis, angiogenesis, and immune responses via modulating several signaling pathways within the tumor microenvironment. Due to its poor water solubility, poor pharmacokinetics/bioavailability profile, and extensive p-glycoprotein-dependent efflux, BBR application in (pre) clinical studies is restricted. To overcome these limitations, BBR can be encapsulated in nanoparticle (NP)-based drug delivery systems, as monotherapy or combinational therapy, and improve BBR therapeutic efficacy. Nanoformulations also facilitate the selective delivery of BBR into lung cancer cells. In addition to the anticancer activities of BBR, especially in lung cancer, here we reviewed the BBR nanoformulations, including polymeric NPs, metal-based NPs, carbon nanostructures, and others, in the treatment of lung cancer.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Haowei Ma
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - A K Kareem
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Hillah, Iraq
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
276
|
Yang Y, Gong Y, Ding Y, Sun S, Bai R, Zhuo S, Zhang Z. LINC01133 promotes pancreatic ductal adenocarcinoma epithelial-mesenchymal transition mediated by SPP1 through binding to Arp3. Cell Death Dis 2024; 15:492. [PMID: 38987572 PMCID: PMC11237081 DOI: 10.1038/s41419-024-06876-3] [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: 12/12/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with limited treatment methods. Long non-coding RNAs (lncRNAs) have been found involved in tumorigenic and progression. The present study revealed that LINC01133, a fewly reported lncRNA, was one of 16 hub genes that could predict PDAC patients' prognosis. LINC01133 was over-expressed in PDAC tumors compared to adjacent pancreas and could promote PDAC proliferation and metastasis in vitro and in vivo, as well as inhibit PDAC apoptosis. LINC01133 expression positively correlated to secreted phosphoprotein 1 (SPP1) expression, leading to an enhanced epithelial-mesenchymal transition (EMT) process. LINC01133 bound with actin-related protein 3 (Arp3), the complex reduced SPP1 mRNA degradation which increased SPP1 mRNA level, ultimately leading to PDAC proliferation. This research revealed a novel mechanism of PDAC development and provided a potential prognosis indicator that may benefit PDAC patients.
Collapse
Affiliation(s)
- Yefan Yang
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yuxi Gong
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ying Ding
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Shuning Sun
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Rumeng Bai
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Shuaishuai Zhuo
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhihong Zhang
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
277
|
Chang WL, Peng JY, Hong CL, Li PC, Lu FJ, Chen CH. Parecoxib and 5-Fluorouracil Synergistically Inhibit EMT and Subsequent Metastasis in Colorectal Cancer by Targeting PI3K/Akt/NF-κB Signaling. Biomedicines 2024; 12:1526. [PMID: 39062099 PMCID: PMC11274433 DOI: 10.3390/biomedicines12071526] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer is one of the most common causes of cancer mortality worldwide, and innovative drugs for the treatment of colorectal cancer are continually being developed. 5-Fluorouracil (5-FU) is a common clinical chemotherapeutic drug. Acquired resistance to 5-FU is a clinical challenge in colorectal cancer treatment. Parecoxib is a selective COX-2-specific inhibitor that was demonstrated to inhibit metastasis in colorectal cancers in our previous study. This study aimed to investigate the synergistic antimetastatic activities of parecoxib to 5-FU in human colorectal cancer cells and determine the underlying mechanisms. Parecoxib and 5-FU synergistically suppressed metastasis in colorectal cancer cells. Treatment with the parecoxib/5-FU combination induced an increase in E-cadherin and decrease in β-catenin expression. The parecoxib/5-FU combination inhibited MMP-9 activity, and the NF-κB pathway was suppressed as well. Mechanistic analysis denoted that the parecoxib/5-FU combination hindered the essential molecules of the PI3K/Akt route to obstruct metastatic colorectal cancer. Furthermore, the parecoxib/5-FU combination could inhibit reactive oxygen species. Our work showed the antimetastatic capacity of the parecoxib/5-FU combination for treating colorectal cancers via the targeting of the PI3K/Akt/NF-κB pathway.
Collapse
Affiliation(s)
- Wan-Ling Chang
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Jyun-Yu Peng
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Chain-Lang Hong
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Pei-Ching Li
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Fung-Jou Lu
- Institute of Medicine, Chung Shan Medical University, No. 110, Section 1, Jianguo North Road, Taichung City 402306, Taiwan;
| | - Ching-Hsein Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, A25-303 Room, Life Sciences Hall, No. 300, Syuefu Road, National Chiayi University, Chiayi City 600355, Taiwan
| |
Collapse
|
278
|
Hassan A, Roy S, Das A, Wahed SA, Bairagi A, Mondal S, Chatterjee N, Das N. Covalent Organic Frameworks as Potential Drug Carriers and Chemotherapeutic Agents for Ovarian Cancers. ACS Biomater Sci Eng 2024; 10:4227-4236. [PMID: 38848308 DOI: 10.1021/acsbiomaterials.4c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Anticancer drugs are often associated with limitations such as poor stability in aqueous solutions, limited cell membrane permeability, nonspecific targeting, and irregular drug release when taken orally. One possible solution to these problems is the use of nanocarriers of drug molecules, particularly those with targeting ability, stimuli-responsive properties, and high drug loading capacity. These nanocarriers can improve drug stability, increase cellular uptake, allow specific targeting of cancer cells, and provide controlled drug release. While improving the therapeutic efficacy of cancer drugs, contemporary researchers also aim to reduce their associated side effects, such that cancer patients are offered with a more effective and targeted treatment strategy. Herein, a set of nine porous covalent organic frameworks (COFs) were tested as drug delivery nanocarriers. Among these, paclitaxel loaded in COF-3 was most effective against the proliferation of ovarian cancer cells. This study highlights the emerging potential of COFs in the field of therapeutic drug delivery. Due to their biocompatibility, these porous COFs provide a viable substrate for controlled drug release, making them attractive candidates for improving drug delivery systems. This work also demonstrates the potential of COFs as efficient drug delivery agents, thereby opening up new opportunities in the field of sarcoma therapy.
Collapse
Affiliation(s)
- Atikur Hassan
- Department of Chemistry, Indian Institute of Technology Patna, Patna 801106, Bihar, India
- Functional Materials Group, Gulf University for Science and Technology, Mubarak Al-Abdullah 32093, Kuwait
| | - Sraddhya Roy
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, 37, S P Mukherjee Road, Kolkata 700 026, India
| | - Ananya Das
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, 37, S P Mukherjee Road, Kolkata 700 026, India
| | - Sk Abdul Wahed
- Department of Chemistry, Indian Institute of Technology Patna, Patna 801106, Bihar, India
| | - Aparajita Bairagi
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, 37, S P Mukherjee Road, Kolkata 700 026, India
| | - Subhadip Mondal
- Department of Chemistry, Indian Institute of Technology Patna, Patna 801106, Bihar, India
| | - Nabanita Chatterjee
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, 37, S P Mukherjee Road, Kolkata 700 026, India
| | - Neeladri Das
- Department of Chemistry, Indian Institute of Technology Patna, Patna 801106, Bihar, India
| |
Collapse
|
279
|
He L, Chen H, Ruan B, He L, Luo M, Fu Y, Zou R. UBQLN4 promotes the proliferation and invasion of non-small cell lung cancer cell by regulating PI3K/AKT pathway. J Cancer Res Clin Oncol 2024; 150:335. [PMID: 38969831 PMCID: PMC11226510 DOI: 10.1007/s00432-024-05862-8] [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: 05/06/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Ubiquilin-4 (UBQLN4), a member of the ubiquilin family, has received limited attention in cancer research to date. Here, we investigated for the first time the functional role and mechanism of UBQLN4 in non-small cell lung cancer (NSCLC). METHODS The Cancer Genome Atlas (TCGA) database was employed to validate UBQLN4 as a differentially expressed gene. Expression differences of UBQLN4 in NSCLC cells and tissues were assessed using immunohistochemistry (IHC) experiment and western blotting (WB) experiment. Kaplan-Meier analysis was conducted to examine the association between UBQLN4 expression and NSCLC prognosis. Functional analyses of UBQLN4 were performed through cell counting kit-8 (CCK-8), colony formation, and transwell invasion assays. The impact of UBQLN4 on tumor-associated signaling pathways was assessed using the path scan intracellular signaling array. In vivo tumorigenesis experiments were conducted to further investigate the influence of UBQLN4 on tumor formation. RESULTS UBQLN4 exhibited up-regulation in both NSCLC tissues and cells. Additionally, over-expression of UBQLN4 was associated with an unfavorable prognosis in NSCLC patients. Functional loss analyses demonstrated that inhibiting UBQLN4 could suppress the proliferation and invasion of NSCLC cells in both in vitro and in vivo settings. Conversely, functional gain experiments yielded opposite results. Path scan intracellular signaling array results suggested that the role of UBQLN4 is associated with the PI3K/AKT pathway, a correlation substantiated by in vitro and in vivo tumorigenesis experiments. CONCLUSION We validated that UBQLN4 promotes proliferation and invasion of NSCLC cells by activating the PI3K/AKT pathway, thereby facilitating the progression of NSCLC. These findings underscore the potential of targeting UBQLN4 as a therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Li He
- Department of Oncology, The People's Hospital of Xinyu City, Xinyu, Jiangxi, 338099, People's Republic of China
| | - Heng Chen
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical Collge, Nanchang University, 17 Yongwai Street, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Bin Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical Collge, Nanchang University, 17 Yongwai Street, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Li He
- Department of Pathology, Jingdezhen First People's Hospital, Jingdezhen, Jiangxi, 333000, People's Republic of China
| | - Ming Luo
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical Collge, Nanchang University, 17 Yongwai Street, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Yulun Fu
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical Collge, Nanchang University, 17 Yongwai Street, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Rui Zou
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical Collge, Nanchang University, 17 Yongwai Street, Nanchang, Jiangxi, 330006, People's Republic of China.
| |
Collapse
|
280
|
Diao Y, Huang S, Liu F, Liao S, Guan C, Xiong X, Zhang P, Li J, Zhang W, Ying Y. CCL2 promotes EGFR-TKIs resistance in non-small cell lung cancer via the AKT-EMT pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1549-1560. [PMID: 38961814 PMCID: PMC11532253 DOI: 10.3724/abbs.2024106] [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: 01/12/2024] [Accepted: 03/11/2024] [Indexed: 07/05/2024] Open
Abstract
Acquired resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) represents a primary cause of treatment failure in non-small cell lung cancer (NSCLC) patients. Chemokine (C-C motif) ligand 2 (CCL2) is recently found to play a pivotal role in determining anti-cancer treatment response. However, the role and mechanism of CCL2 in the development of EGFR-TKIs resistance have not been fully elucidated. In the present study, we focus on the function of CCL2 in the development of acquired resistance to EGFR-TKIs in NSCLC cells. Our results show that CCL2 is aberrantly upregulated in EGFR-TKIs-resistant NSCLC cells and that CCL2 overexpression significantly diminishes sensitivity to EGFR-TKIs. Conversely, CCL2 suppression by CCL2 synthesis inhibitor, bindarit, or CCL2 knockdown can reverse this resistance. CCL2 upregulation can also lead to enhanced migration and increased expressions of epithelial-mesenchymal transition (EMT) markers in EGFR-TKI-resistant NSCLC cells, which could also be rescued by CCL2 knockdown or inhibition. Furthermore, our findings suggest that CCL2-dependent EGFR-TKIs resistance involves the AKT-EMT signaling pathway; inhibition of this pathway effectively attenuates CCL2-induced cell migration and EMT marker expression. In summary, CCL2 promotes the development of acquired EGFR-TKIs resistance and EMT while activating AKT signaling in NSCLC. These insights suggest a promising avenue for the development of CCL2-targeted therapies that prevent EGFR-TKIs resistance in NSCLC.
Collapse
Affiliation(s)
- Yunlian Diao
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Shibo Huang
- The Clinical Trial Research Centerthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Fangpeng Liu
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Shu Liao
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Chenxi Guan
- Department of PhysiologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Xiaojian Xiong
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Ping Zhang
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Junyao Li
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Wei Zhang
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Ying Ying
- Jiangxi Provincial Key Laboratory of Prevention and Treatment of Infectious DiseasesJiangxi Medical Center for Major Public Health Eventsthe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| |
Collapse
|
281
|
Ma S, Pu C. miR-129-2-3p binds SEMA4C to regulate HCC development and inhibit the EMT. Mutat Res 2024; 829:111872. [PMID: 39018715 DOI: 10.1016/j.mrfmmm.2024.111872] [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: 03/02/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Among primary liver cancers, HCC is the most prevalent. Small noncoding RNAs called miRNAs control the expression of downstream target genes to take part in a variety of physiological and pathological processes, including those related to cancer. METHODS miR-129-2-3p and SEMA4C expression levels were assessed using RT-qPCR. The CCK-8, invasion, and wound healing assays were used to confirm the capacity of HCC cells for proliferation, invasion and migration respectively. Serum SEMA4C levels were detected via ELISA. The RIP and dual-luciferase reporter assays were used to confirm the existence of intergenic binding sites. Cell apoptosis assay and cell cycle assay were performed to detect the apoptosis rate and cycle distribution of cells, and WB was performed to detect the protein expression of SEMA4C, RhoA, ROCK1, E-cadherin, N-cadherin, and vimentin. Furthermore, cancer-inhibiting role of miR-129-2-3p were further confirmed by animal tests. RESULTS miR-129-2-3p expression was reduced in HCC tissues and cells. Overexpression of miR-129-2-3p decreased the proliferation, invasion, migration, and EMT in HCC cells, whereas inhibition of miR-129-2-3p had the opposite effects. Our research also showed that SEMA4C was increased in HCC tissues, serum and cells, and that SEMA4C knockdown prevented HCC cell invasion, migration, proliferation, and EMT. Overexpression of SEMA4C reversed the inhibitory effect of miR-129-2-3p on HCC. CONCLUSIONS Overall, we discovered that through binding to SEMA4C, miR-129-2-3p regulates HCC cell proliferation, invasion, migration, and EMT.
Collapse
Affiliation(s)
- Siyuan Ma
- School of Laboratory Medicine, Wannan Medical College, China.
| | - Chun Pu
- School of Laboratory Medicine, Wannan Medical College, China.
| |
Collapse
|
282
|
Peng Z, Yi Y, Nie Y, Wang T, Tang J, Hong S, Liu Y, Huang W, Sun S, Tan H, Wu M. Softening the tumor matrix through cholesterol depletion breaks the physical barrier for enhanced antitumor therapy. J Control Release 2024; 371:29-42. [PMID: 38763389 DOI: 10.1016/j.jconrel.2024.05.027] [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: 01/23/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
The tumor develops defense tactics, including conversing the mechanical characteristics of tumor cells and their surrounding environment. A recent study reported that cholesterol depletion stiffens tumor cells, which could enhance adaptive T-cell immunotherapy. However, it remains unclear whether reducing the cholesterol in tumor cells contributes to re-educating the stiff tumor matrix, which serves as a physical barrier against drug penetration. Herein, we found that depleting cholesterol from tumor cells can demolish the intratumor physical barrier by disrupting the mechanical signal transduction between tumor cells and the extracellular matrix through the destruction of lipid rafts. This disruption allows nanoparticles (H/S@hNP) to penetrate deeply, resulting in improved photodynamic treatment. Our research also indicates that cholesterol depletion can inhibit the epithelial-mesenchymal transition and repolarize tumor-associated macrophages from M2 to M1, demonstrating the essential role of cholesterol in tumor progression. Overall, this study reveals that a cholesterol-depleted, softened tumor matrix reduces the difficulty of drug penetration, leading to enhanced antitumor therapeutics.
Collapse
Affiliation(s)
- Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yunfei Yi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yichu Nie
- Department of Translational Medicine Research Institute, First People's Hospital of Foshan, Foshan 528000, China
| | - Tianqi Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Jia Tang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yuanqi Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Wenxin Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Hui Tan
- Center for Child Care and Mental Health (CCCMH), Shenzhen Children's Hospital, Shenzhen 518038, China.
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
283
|
Ito M, Yajima S, Suzuki T, Oshima Y, Nanami T, Sumazaki M, Shiratori F, Takizawa H, Li SY, Zhang BS, Yoshida Y, Matsutani T, Hiwasa T, Shimada H. Combination of high anti-SKI and low anti-TMED5 antibody levels is preferable prognostic factor in esophageal carcinoma. Cancer Sci 2024; 115:2209-2219. [PMID: 38634426 PMCID: PMC11247554 DOI: 10.1111/cas.16185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Given that esophageal cancer is highly malignant, the discovery of novel prognostic markers is eagerly awaited. We performed serological identification of antigens by recombinant cDNA expression cloning (SEREX) and identified SKI proto-oncogene protein and transmembrane p24 trafficking protein 5 (TMED5) as antigens recognized by serum IgG antibodies in patients with esophageal carcinoma. SKI and TMED5 proteins were expressed in Escherichia coli, purified by affinity chromatography, and used as antigens. The serum anti-SKI antibody (s-SKI-Ab) and anti-TMED5 antibody (s-TMED5-Ab) levels were significantly higher in 192 patients with esophageal carcinoma than in 96 healthy donors. The presence of s-SKI-Abs and s-TMED5-Abs in the patients' sera was confirmed by western blotting. Immunohistochemical staining showed that the TMED5 protein was highly expressed in the cytoplasm and nuclear compartments of the esophageal squamous cell carcinoma tissues, whereas the SKI protein was localized predominantly in the nuclei. Regarding the overall survival in 91 patients who underwent radical surgery, the s-SKI-Ab-positive and s-TMED5-Ab-negative statuses were significantly associated with a favorable prognosis. Additionally, the combination of s-SKI-Ab-positive and s-TMED5-Ab-negative cases showed an even clearer difference in overall survival as compared with that of s-SKI-Ab-negative and s-TMED5-Ab-positive cases. The s-SKI-Ab and s-TMED5-Ab biomarkers are useful for diagnosing esophageal carcinoma and distinguishing between favorable and poor prognoses.
Collapse
Affiliation(s)
- Masaaki Ito
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Yajima
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Takashi Suzuki
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Yoko Oshima
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Tatsuki Nanami
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Makoto Sumazaki
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Fumiaki Shiratori
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, Japan
| | - Shu-Yang Li
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Bo-Shi Zhang
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takaki Hiwasa
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
284
|
Bracken CP, Goodall GJ, Gregory PA. RNA regulatory mechanisms controlling TGF-β signaling and EMT in cancer. Semin Cancer Biol 2024; 102-103:4-16. [PMID: 38917876 DOI: 10.1016/j.semcancer.2024.06.001] [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: 12/15/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a major contributor to metastatic progression and is prominently regulated by TGF-β signalling. Both EMT and TGF-β pathway components are tightly controlled by non-coding RNAs - including microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) - that collectively have major impacts on gene expression and resulting cellular states. While miRNAs are the best characterised regulators of EMT and TGF-β signaling and the miR-200-ZEB1/2 feedback loop plays a central role, important functions for lncRNAs and circRNAs are also now emerging. This review will summarise our current understanding of the roles of non-coding RNAs in EMT and TGF-β signaling with a focus on their functions in cancer progression.
Collapse
Affiliation(s)
- Cameron P Bracken
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Gregory J Goodall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
285
|
Zhang J, Miao G, Ta MH, Zhao B, Wang W, Xing Y, Qian H, Huang D, Chen W, Zhong Y. Photothermal-controlled NO-releasing Nanogels reverse epithelial-mesenchymal transition and restore immune surveillance against cancer metastasis. J Control Release 2024; 371:16-28. [PMID: 38763388 DOI: 10.1016/j.jconrel.2024.05.028] [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: 12/07/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Metastasis leads to high mortality among cancer patients. It is a complex, multi-step biological process that involves the dissemination of cancer cells from the primary tumor and their systemic spread throughout the body, primarily through the epithelial-mesenchymal transition (EMT) program and immune evasion mechanisms. It presents a challenge in how to comprehensively treat metastatic cancer cells throughout the entire stage of the metastatic cascade using a simple system. Here, we fabricate a nanogel (HNO-NG) by covalently crosslinking a macromolecular nitric oxide (NO) donor with a photothermal IR780 iodide-containing hyaluronic acid derivative via a click reaction. This enables stable storage and tumor-targeted, photothermia-triggered release of NO to combat tumor metastasis throughout all stages. Upon laser irradiation (HNO-NG+L), the surge in NO production within tumor cells impairs the NF-κB/Snail/RKIP signaling loop that promotes the EMT program through S-nitrosylation, thus inhibiting cell dissemination from the primary tumor. On the other hand, it induces immunogenic cell death (ICD) and thereby augments anti-tumor immunity, which is crucial for killing both the primary tumor and systemically distributed tumor cells. Therefore, HNO-NG+L, by fully leveraging EMT reversal, ICD induction, and the lethal effect of NO, achieved impressive eradication of the primary tumor and significant prevention of lung metastasis in a mouse model of orthotropic 4T1 breast tumor that spontaneously metastasizes to the lungs, extending the NO-based therapeutic approach against tumor metastasis.
Collapse
Affiliation(s)
- Junmei Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Guizhi Miao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - My Hanh Ta
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yanran Xing
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Hongliang Qian
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
286
|
Wang Z, Lei Z, Wang Y, Wang S, Wang JP, Jin E, Liu X, Sun R, Zhang HT. Bone-metastatic lung adenocarcinoma cells bearing CD74-ROS1 fusion interact with macrophages to promote their dissemination. Oncogene 2024; 43:2215-2227. [PMID: 38802647 DOI: 10.1038/s41388-024-03072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Approximately 40% of patients with lung adenocarcinoma (LUAD) often develop bone metastases during the course of their disease. However, scarcely any in vivo model of LUAD bone metastasis has been established, leading to a poor understanding of the mechanisms underlying LUAD bone metastasis. Here, we established a multiorgan metastasis model via the left ventricular injection of luciferase-labeled LUAD cells into nude mice and then screened out lung metastasis (LuM) and bone metastasis (BoM) cell subpopulations. BoM cells exhibited greater stemness and epithelial-mesenchymal transition (EMT) plasticity than LuM cells and initially colonized the bone and subsequently disseminated to distant organs after being reinjected into mice. Moreover, a CD74-ROS1 fusion mutation (C6; R34) was detected in BoM cells but not in LuM cells. Mechanistically, BoM cells bearing the CD74-ROS1 fusion highly secrete the C-C motif chemokine ligand 5 (CCL5) protein by activating STAT3 signaling, recruiting macrophages in tumor microenvironment and strongly inducing M2 polarization of macrophages. BoM cell-activated macrophages produce a high level of TGF-β1, thereby facilitating EMT and invasion of LUAD cells via TGF-β/SMAD2/3 signaling. Targeting the CD74-ROS1/CCL5 axis with Crizotinib (a ROS1 inhibitor) and Maraviroc (a CCL5 receptor inhibitor) in vivo strongly impeded bone metastasis and secondary metastasis of BoM cells. Our findings reveal the critical role of the CD74-ROS1/STAT3/CCL5 axis in the interaction between LUAD bone metastasis cells and macrophages for controlling LUAD cell dissemination, highlighting the significance of the bone microenvironment in LUAD bone metastasis and multiorgan secondary metastasis, and suggesting that targeting CD74-ROS1 and CCL5 is a promising therapeutic strategy for LUAD bone metastasis.
Collapse
Affiliation(s)
- Zhao Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Zhe Lei
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yong Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Shengjie Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu Province, China
| | - Jia-Ping Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Donghai County People's Hospital, Lianyungang, 222000, Jiangsu Province, China
| | - Ersuo Jin
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Xia Liu
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Runfeng Sun
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China.
- Donghai County People's Hospital, Lianyungang, 222000, Jiangsu Province, China.
| | - Hong-Tao Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Collaborative Innovation Center of Molecular Medicine between Soochow University and Donghai County People's Hospital, Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China.
- Department of Genetics, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China.
- Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, 215123, Jiangsu Province, China.
| |
Collapse
|
287
|
Man S, Cui Y, Shi D, Lv P, Ma L, Gao W. Formosanin C inhibits pulmonary metastasis by targeting stearyl CoA desaturase-1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155689. [PMID: 38728921 DOI: 10.1016/j.phymed.2024.155689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Cisplatin (DDP) as the first-line drug has been used in cancer therapy. However, side effects and drug resistance are the challenges of DDP. Disordered lipid metabolism is related to DDP resistance. STUDY DESIGN In this study, formosanin C (FC) as the main compound of Rhizoma Paridis saponins (RPS) inhibits pulmonary metastasis by targeting stearyl CoA desaturase-1. METHODS AND RESULTS RPS prolonged the survival period of mice, reduced pulmonary metastases and alleviated colon toxicity caused by DDP. FC as the main compound of RPS enhanced the anti-tumor and anti-metastatic effects of DDP. FC decreased the mRNA level of SCD1 and the content of lipid droplets (LDs) in lung cancer cells. Molecular dynamics and isothermal titration calorimetry verified the binding stability and spontaneously between FC and SCD1. SiSCD1 reduced the content of LDs in cell lines and increased mitochondria (mtROS), which was consistent with the results of FC treatment. The combination group decreased DNA repair associated protein as well as DDP resistance markers such as ERCC1 and 53bp1, and increased DNA damage marker like γH2AX, which indirectly confirmed the occurrence of mtROS. In addition, FC combination with DDP also affected epithelial-mesenchymal transition-related protein like VIM and CDH1 in vivo experiments, and thereby inhibited pulmonary metastasis. CONCLUSION Our research indicated that the FC as the main compound of RPS targeted the CY2 domain of SCD1, inhibited lipid metabolism in mice, and thereby suppressed cancer metastases. This provided support for use of FC to treat cancer based on lipid metabolism pathway.
Collapse
Affiliation(s)
- 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.
| | - Yingfang Cui
- 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
| | - Dandan Shi
- 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
| | - 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
| | - 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
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
288
|
Acero-Riaguas L, Griso-Acevedo AB, SanLorenzo-Vaquero A, Ibáñez-Herrera B, Fernandez-Diaz SM, Mascaraque M, Sánchez-Siles R, López-García I, Benítez-Buelga C, Bravo-Burguillos ER, Castelo B, Cebrián-Carretero JL, Perona R, Sastre L, Sastre-Perona A. DUSP1 and SOX2 expression determine squamous cell carcinoma of the salivary gland progression. Sci Rep 2024; 14:15007. [PMID: 38951654 PMCID: PMC11217270 DOI: 10.1038/s41598-024-65945-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Salivary gland squamous cell carcinomas (SG-SCCs) constitute a rare type of head and neck cancer which is linked to poor prognosis. Due to their low frequency, the molecular mechanisms responsible for their aggressiveness are poorly understood. In this work we studied the role of the phosphatase DUSP1, a negative regulator of MAPK activity, in controlling SG-SCC progression. We generated DUSP1 KO clones in A253 human cells. These clones showed a reduced ability to grow in 2D, self-renew in ECM matrices and to form tumors in immunodeficient mice. This was caused by an overactivation of the stress and apoptosis kinase JNK1/2 in DUSP1-/+ clones. Interestingly, RNAseq analysis revealed that the expression of SOX2, a well-known self-renewal gene was decreased at the mRNA and protein levels in DUSP1-/+ cells. Unexpectedly, CRISPR-KO of SOX2 did not recapitulate DUSP1-/+ phenotype, and SOX2-null cells had an enhanced ability to self-renew and to form tumors in mice. Gene expression analysis demonstrated that SOX2-null cells have a decreased squamous differentiation profile -losing TP63 expression- and an increased migratory phenotype, with an enhanced epithelial to mesenchymal transition signature. In summary, our data indicates that DUSP1 and SOX2 have opposite functions in SG-SCC, being DUSP1 necessary for tumor growth and SOX2 dispensable showing a tumor suppressor function. Our data suggest that the combined expression of SOX2 and DUSP1 could be a useful biomarker to predict progression in patients with SG-SCCs.
Collapse
Affiliation(s)
- Lucía Acero-Riaguas
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Ana Belén Griso-Acevedo
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Alejandro SanLorenzo-Vaquero
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Blanca Ibáñez-Herrera
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Sara María Fernandez-Diaz
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Marta Mascaraque
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Rocío Sánchez-Siles
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Iván López-García
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Carlos Benítez-Buelga
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Elena Ruiz Bravo-Burguillos
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Beatriz Castelo
- Medical Oncology Department, University Hospital La Paz, 28046, Madrid, Spain
| | - José Luis Cebrián-Carretero
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
- Oral and Maxillofacial Surgery Department, University Hospital La Paz, 28046, Madrid, Spain
| | - Rosario Perona
- Instituto de Salud Carlos III and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Leandro Sastre
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Ana Sastre-Perona
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain.
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain.
| |
Collapse
|
289
|
Cai Q, He Y, Zhou Y, Zheng J, Deng J. Nanomaterial-Based Strategies for Preventing Tumor Metastasis by Interrupting the Metastatic Biological Processes. Adv Healthc Mater 2024; 13:e2303543. [PMID: 38411537 DOI: 10.1002/adhm.202303543] [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: 10/17/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Tumor metastasis is the primary cause of cancer-related deaths. The prevention of tumor metastasis has garnered notable interest and interrupting metastatic biological processes is considered a potential strategy for preventing tumor metastasis. The tumor microenvironment (TME), circulating tumor cells (CTCs), and premetastatic niche (PMN) play crucial roles in metastatic biological processes. These processes can be interrupted using nanomaterials due to their excellent physicochemical properties. However, most studies have focused on only one aspect of tumor metastasis. Here, the hypothesis that nanomaterials can be used to target metastatic biological processes and explore strategies to prevent tumor metastasis is highlighted. First, the metastatic biological processes and strategies involving nanomaterials acting on the TME, CTCs, and PMN to prevent tumor metastasis are briefly summarized. Further, the current challenges and prospects of nanomaterials in preventing tumor metastasis by interrupting metastatic biological processes are discussed. Nanomaterial-and multifunctional nanomaterial-based strategies for preventing tumor metastasis are advantageous for the long-term fight against tumor metastasis and their continued exploration will facilitate rapid progress in the prevention, diagnosis, and treatment of tumor metastasis. Novel perspectives are outlined for developing more effective strategies to prevent tumor metastasis, thereby improving the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijia He
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
290
|
Ma Y, Zhou X. Accurate and efficient integrative reference-informed spatial domain detection for spatial transcriptomics. Nat Methods 2024; 21:1231-1244. [PMID: 38844627 PMCID: PMC11831598 DOI: 10.1038/s41592-024-02284-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 04/18/2024] [Indexed: 06/23/2024]
Abstract
Spatially resolved transcriptomics (SRT) studies are becoming increasingly common and large, offering unprecedented opportunities in mapping complex tissue structures and functions. Here we present integrative and reference-informed tissue segmentation (IRIS), a computational method designed to characterize tissue spatial organization in SRT studies through accurately and efficiently detecting spatial domains. IRIS uniquely leverages single-cell RNA sequencing data for reference-informed detection of biologically interpretable spatial domains, integrating multiple SRT slices while explicitly considering correlations both within and across slices. We demonstrate the advantages of IRIS through in-depth analysis of six SRT datasets encompassing diverse technologies, tissues, species and resolutions. In these applications, IRIS achieves substantial accuracy gains (39-1,083%) and speed improvements (4.6-666.0) in moderate-sized datasets, while representing the only method applicable for large datasets including Stereo-seq and 10x Xenium. As a result, IRIS reveals intricate brain structures, uncovers tumor microenvironment heterogeneity and detects structural changes in diabetes-affected testis, all with exceptional speed and accuracy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Biostatistics, Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
291
|
Ni D, Qi Z, Wang Y, Man Y, Pang J, Tang W, Chen J, Li J, Li G. KLF15-activated MARCH2 boosts cell proliferation and epithelial-mesenchymal transition and presents diagnostic significance for hepatocellular carcinoma. Exp Cell Res 2024; 440:114117. [PMID: 38848952 DOI: 10.1016/j.yexcr.2024.114117] [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: 01/22/2024] [Revised: 05/07/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
PURPOSE Membrane associated ubiquitin ligase MARCH2 majorly involves in inflammation response and protein trafficking. However, its comprehensive role in hepatocellular carcinoma (HCC) is largely unknown. METHODS Firstly, multiple bioinformatic analyses were applied to determine MARCH2 mRNA level, its expression comparison in diverse molecular and immune subtypes, and diagnostic value in HCC. Subsequently, RNA-seq, real-time quantitative PCR, immunohistochemistry and cell proliferation assay are used to explore the epithelial-mesenchymal transition (EMT) and proliferation by gene-silencing or overexpressing in cultured HCC cells or in vivo xenograft. Moreover, dual luciferase reporter assay and immunoblotting are delved into verify the transcription factor that activating MARCH2 promoter. RESULTS Multiple bioinformatic analyses demonstrate that MARCH2 is upregulated in multiple cancer types and exhibits startling diagnostic value as well as distinct molecular and immune subtypes in HCC. RNA-seq analysis reveals MARCH2 may promote EMT, cell proliferation and migration in HepG2 cells. Furthermore, overexpression of MARCH2 triggers EMT and significantly enhances HCC cell migration, proliferation and colony formation in a ligase activity-dependent manner. Additionally, above observations are validated in the HepG2 mice xenografts. For up-stream mechanism, transcription factor KLF15 is highly expressed in HCC and activates MARCH2 expression. CONCLUSION KLF15 activated MARCH2 triggers EMT and serves as a fascinating biomarker for precise diagnosis of HCC. Consequently, MARCH2 emerges as a promising candidate for target therapy in cancer management.
Collapse
Affiliation(s)
- Dongsheng Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Graduate School of Peking Union Medical College, Beijing, 100730, PR China
| | - Zhaolai Qi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Graduate School of Peking Union Medical College, Beijing, 100730, PR China
| | - Yuefeng Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Yong Man
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Jing Pang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, PR China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Guoping Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Graduate School of Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
292
|
Biswas P, Kaium MA, Islam Tareq MM, Tauhida SJ, Hossain MR, Siam LS, Parvez A, Bibi S, Hasan MH, Rahman MM, Hosen D, Islam Siddiquee MA, Ahmed N, Sohel M, Azad SA, Alhadrami AH, Kamel M, Alamoudi MK, Hasan MN, Abdel-Daim MM. The experimental significance of isorhamnetin as an effective therapeutic option for cancer: A comprehensive analysis. Biomed Pharmacother 2024; 176:116860. [PMID: 38861855 DOI: 10.1016/j.biopha.2024.116860] [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: 04/06/2024] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Isorhamnetin (C16H12O7), a 3'-O-methylated derivative of quercetin from the class of flavonoids, is predominantly present in the leaves and fruits of several plants, many of which have traditionally been employed as remedies due to its diverse therapeutic activities. The objective of this in-depth analysis is to concentrate on Isorhamnetin by addressing its molecular insights as an effective anticancer compound and its synergistic activity with other anticancer drugs. The main contributors to Isorhamnetin's anti-malignant activities at the molecular level have been identified as alterations of a variety of signal transduction processes and transcriptional agents. These include ROS-mediated cell cycle arrest and apoptosis, inhibition of mTOR and P13K pathway, suppression of MEK1, PI3K, NF-κB, and Akt/ERK pathways, and inhibition of Hypoxia Inducible Factor (HIF)-1α expression. A significant number of in vitro and in vivo research studies have confirmed that it destroys cancerous cells by arresting cell cycle at the G2/M phase and S-phase, down-regulating COX-2 protein expression, PI3K, Akt, mTOR, MEK1, ERKs, and PI3K signaling pathways, and up-regulating apoptosis-induced genes (Casp3, Casp9, and Apaf1), Bax, Caspase-3, P53 gene expression and mitochondrial-dependent apoptosis pathway. Its ability to suppress malignant cells, evidence of synergistic effects, and design of drugs based on nanomedicine are also well supported to treat cancer patients effectively. Together, our findings establish a crucial foundation for understanding Isorhamnetin's underlying anti-cancer mechanism in cancer cells and reinforce the case for the requirement to assess more exact molecular signaling pathways relating to specific cancer and in vivo anti-cancer activities.
Collapse
Affiliation(s)
- Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh; ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh
| | - Md Abu Kaium
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Sadia Jannat Tauhida
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Ridoy Hossain
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Labib Shahriar Siam
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Anwar Parvez
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad 41000, Pakistan
| | - Md Hasibul Hasan
- Department of Food Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj 8100, Bangladesh
| | - Md Moshiur Rahman
- Department of Information Systems Security, Faculty of Science & Technology, Bangladesh University of Professionals, Mirpur 1216, Bangladesh
| | - Delwar Hosen
- Department of Electrical and Computer Engineering, North South University, Dhaka 1229, Bangladesh
| | | | - Nasim Ahmed
- Department of Pharmacy, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Sohel
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Salauddin Al Azad
- Immunoinformatics and Vaccinomics Research Unit, RPG Interface Lab, Jashore 7400, Bangladesh
| | - Albaraa H Alhadrami
- Faculty of Medicine, King Abdulaziz University, P.O.Box 80402, Jeddah 21589, Saudi Arabia
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Mariam K Alamoudi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Md Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
293
|
Zeng Z, Li Y, Zhou H, Li M, Ye J, Li D, Zhu Y, Zhang Y, Zhang X, Deng Y, Li J, Gu L, Wu J. System-wide identification of novel de-ubiquitination targets for USP10 in gastric cancer metastasis through multi-omics screening. BMC Cancer 2024; 24:773. [PMID: 38937694 PMCID: PMC11209979 DOI: 10.1186/s12885-024-12549-3] [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: 02/21/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
OBJECTIVE Ubiquitin-specific peptidase 10 (USP10), a typical de-ubiquitinase, has been found to play a double-edged role in human cancers. Previously, we reported that the expression of USP10 was negatively correlated with the depth of gastric wall invasion, lymph node metastasis, and prognosis in gastric cancer (GC) patients. However, it remains unclear whether USP10 can regulate the metastasis of GC cells through its de-ubiquitination function. METHODS In this study, proteome, ubiquitinome, and transcriptome analyses were conducted to comprehensively identify novel de-ubiquitination targets for USP10 in GC cells. Subsequently, a series of validation experiments, including in vitro cell culture studies, in vivo metastatic tumor models, and clinical sample analyses, were performed to elucidate the regulatory mechanism of USP10 and its de-ubiquitination targets in GC metastasis. RESULTS After overexpression of USP10 in GC cells, 146 proteins, 489 ubiquitin sites, and 61 mRNAs exhibited differential expression. By integrating the results of multi-omics, we ultimately screened 9 potential substrates of USP10, including TNFRSF10B, SLC2A3, CD44, CSTF2, RPS27, TPD52, GPS1, RNF185, and MED16. Among them, TNFRSF10B was further verified as a direct de-ubiquitination target for USP10 by Co-IP and protein stabilization assays. The dysregulation of USP10 or TNFRSF10B affected the migration and invasion of GC cells in vitro and in vivo models. Molecular mechanism studies showed that USP10 inhibited the epithelial-mesenchymal transition (EMT) process by increasing the stability of TNFRSF10B protein, thereby regulating the migration and invasion of GC cells. Finally, the retrospective clinical sample studies demonstrated that the downregulation of TNFRSF10B expression was associated with poor survival among 4 of 7 GC cohorts, and the expression of TNFRSF10B protein was significantly negatively correlated with the incidence of distant metastasis, diffuse type, and poorly cohesive carcinoma. CONCLUSIONS Our study established a high-throughput strategy for screening de-ubiquitination targets for USP10 and further confirmed that inhibiting the ubiquitination of TNFRSF10B might be a promising therapeutic strategy for GC metastasis.
Collapse
Affiliation(s)
- Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yina Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Heng Zhou
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mingyang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juan Ye
- Department of Pharmacy, Huazhong University of Science and Technology Hospital, Wuhan, Hubei, China
| | - Dan Li
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuxi Zhu
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yunchao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juan Li
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Lijuan Gu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Jie Wu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
294
|
Farina A, Viggiani V, Cortese F, Moretti M, Tartaglione S, Angeloni A, Anastasi E. Combined PIVKA II and Vimentin-Guided EMT Tracking in Pancreatic Adenocarcinoma Combined Biomarker-Guided EMT Tracking in PDAC. Cancers (Basel) 2024; 16:2362. [PMID: 39001424 PMCID: PMC11240554 DOI: 10.3390/cancers16132362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
"Background/Aim": the current inability to diagnose Pancreatic Cancer Adenocarcinoma (PDAC) at an early stage strongly influences therapeutic strategies. Protein Induced by Vitamin K Absence (PIVKA II) showed an accurate diagnostic performance for PDAC. Since circulating PIVKA II has been recently associated with pancreatic origin cells with Vimentin, an epithelial-to-mesenchymal transition (EMT) early activation marker, the aim of this study was to investigate in vivo the combination between the two proteins. "Materials and Methods": we assayed the presence of PIVKA II and Vimentin proteins by using different diagnostic methods. A total of 20 PDAC patients and 10 healthy donors were tested by Western Blot analysis; 74 PDAC patient and 46 healthy donors were assayed by ECLIA and Elisa. "Results": Western Blot analysis showed the concomitant expression of PIVKA II and Vimentin in PDAC patient sera. Immunometric assay performed on a larger cohort of patients demonstrated that 72% of PIVKA II-positive PDAC patients were Vimentin-positive. Additionally, in a group of PDAC patients with PIVKA II levels ≥2070 ng/mL, the percentage of Vimentin-positive subjects reached 84%. "Conclusion": the association between PIVKA II protein and the EMT suggests that this molecule could be considered a marker of the acquisition of an aggressive phenotype.
Collapse
Affiliation(s)
- Antonella Farina
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Valentina Viggiani
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Francesca Cortese
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Marta Moretti
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Sara Tartaglione
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Emanuela Anastasi
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
295
|
Li Q, Wang T, Wang X, Ge X, Yang T, Wang W. DDX56 promotes EMT and cancer stemness via MELK-FOXM1 axis in hepatocellular carcinoma. iScience 2024; 27:109827. [PMID: 38827395 PMCID: PMC11141150 DOI: 10.1016/j.isci.2024.109827] [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: 09/01/2023] [Revised: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 06/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global cause of death, with epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC)-like properties contributing to its metastasis. DEAD box helicase 56 (DDX56) is involved in carcinogenesis, but its role in EMT induction and stem phenotype maintenance is unclear. This study assessed the impact of DDX56 absence on HCC cell stemness and EMT. DDX56 was found to be overexpressed in HCC tissues, correlating with disease stage and prognosis. In vitro, DDX56 stimulated tumor cell proliferation, migration, invasion, EMT, and stemness. It also enhanced maternal embryonic leucine-zipper kinase (MELK)-mediated forkhead box protein M1 (FOXM1) expression, regulating cancer stemness and malignant traits. In vivo, DDX56 knockdown in tumor-bearing mice reduced tumorigenicity and lung metastasis by modulating the MELK-FOXM1 signaling pathway. Collectively, DDX56 initiates stem cell-like traits in HCC and promotes EMT via MELK-FOXM1 activation, shedding light on HCC pathogenesis and suggesting a potential anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Qing Li
- Department of Internal Medicine, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, Liaoning Province, China
| | - Tianyi Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Ximin Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - XinYu Ge
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Tao Yang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| |
Collapse
|
296
|
Zhao X, Shao X, Huang X, Dang C, Wang R, Li H. The H2Valdien derivatives regulate the epithelial-mesenchymal transition of hepatoma carcinoma cells through the Hedgehog signaling pathway. Open Med (Wars) 2024; 19:20240954. [PMID: 38911252 PMCID: PMC11193360 DOI: 10.1515/med-2024-0954] [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: 10/31/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 06/25/2024] Open
Abstract
This research delves into the influence of H2Valdien derivatives on the proliferation, migration, and apoptosis induction in hepatoma carcinoma cells (HepG2, Huh-7, and SMMC-7721), with a specific emphasis on inhibiting epithelial-mesenchymal transition (EMT) through modulation of the Hedgehog (Hh) signaling pathway. Utilizing the cell counting kit-8 method, flow cytometry, TUNEL assay, wound healing, and transwell assays, we observed a dose-dependent growth arrest and apoptosis induction in HepG2, Huh-7, and SMMC-7721 cells. Notably, H2Valdien derivatives exhibited a capacity to reduce migration and invasion, impacting the expression of EMT-associated proteins such as N-cadherin, vimentin, and E-cadherin. Mechanistically, these derivatives demonstrated the inhibition of the Hh signaling pathway by inactivating Sonic Hh (Shh) and smoothened proteins. This study underscores the robust antiproliferative and apoptosis-inducing effects of H2Valdien derivatives on hepatoma carcinoma cells and elucidates their regulatory role in EMT through modulation of the Hh signaling pathway, providing valuable insights for potential therapeutic interventions.
Collapse
Affiliation(s)
- Xuhui Zhao
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, 730000, Lanzhou, China
| | - Xiangxiang Shao
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, 730000, Lanzhou, China
| | - Xiaomin Huang
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, 730000, Lanzhou, China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, 730000, Lanzhou, China
| | - Ruilin Wang
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, 730000, Lanzhou, China
| | - Hongling Li
- Department of Oncology, Gansu Provincial Hospital, 730000, Lanzhou, China
| |
Collapse
|
297
|
Yan W, Rao D, Fan F, Liang H, Zhang Z, Dong H. Hepatitis B virus X protein and TGF-β: partners in the carcinogenic journey of hepatocellular carcinoma. Front Oncol 2024; 14:1407434. [PMID: 38962270 PMCID: PMC11220127 DOI: 10.3389/fonc.2024.1407434] [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: 03/26/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Hepatitis B infection is substantially associated with the development of liver cancer globally, with the prevalence of hepatocellular carcinoma (HCC) cases exceeding 50%. Hepatitis B virus (HBV) encodes the Hepatitis B virus X (HBx) protein, a pleiotropic regulatory protein necessary for the transcription of the HBV covalently closed circular DNA (cccDNA) microchromosome. In previous studies, HBV-associated HCC was revealed to be affected by HBx in multiple signaling pathways, resulting in genetic mutations and epigenetic modifications in proto-oncogenes and tumor suppressor genes. In addition, transforming growth factor-β (TGF-β) has dichotomous potentials at various phases of malignancy as it is a crucial signaling pathway that regulates multiple cellular and physiological processes. In early HCC, TGF-β has a significant antitumor effect, whereas in advanced HCC, it promotes malignant progression. TGF-β interacts with the HBx protein in HCC, regulating the pathogenesis of HCC. This review summarizes the respective and combined functions of HBx and TGB-β in HCC occurrence and development.
Collapse
Affiliation(s)
- Wei Yan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
| | - Dean Rao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
| | - Feimu Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission (NHC), Chinese Academy of Medical Sciences, Wuhan, China
| | - Zunyi Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
| | - Hanhua Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
| |
Collapse
|
298
|
Jin L, Zhou S, Zhao S, Long J, Huang Z, Zhou J, Zhang Y. Early short-term hypoxia promotes epidermal cell migration by activating the CCL2-ERK1/2 pathway and epithelial-mesenchymal transition during wound healing. BURNS & TRAUMA 2024; 12:tkae017. [PMID: 38887221 PMCID: PMC11182653 DOI: 10.1093/burnst/tkae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/05/2024] [Indexed: 06/20/2024]
Abstract
Background Due to vasculature injury and increased oxygen consumption, the early wound microenvironment is typically in a hypoxic state. We observed enhanced cell migration ability under early short-term hypoxia. CCL2 belongs to the CC chemokine family and was found to be increased in early hypoxic wounds and enriched in the extracellular signal-regulated kinase (ERK)1/2 pathway in our previous study. However, the underlying mechanism through which the CCL2-ERK1/2 pathway regulates wound healing under early short-term hypoxia remains unclear. Activation of epithelial-mesenchymal transition (EMT) is a key process in cancer cell metastasis, during which epithelial cells acquire the characteristics of mesenchymal cells and enhance cell motility and migration ability. However, the relationship between epithelial cell migration and EMT under early short-term hypoxia has yet to be explored. Methods HaCaT cells were cultured to verify the effect of early short-term hypoxia on migration through cell scratch assays. Lentiviruses with silenced or overexpressed CCL2 were used to explore the relationship between CCL2 and migration under short-term hypoxia. An acute full-thickness cutaneous wound rat model was established with the application of an ERK inhibitor to reveal the hidden role of the ERK1/2 pathway in the early stage of wound healing. The EMT process was verified in all the above experiments through western blotting. Results In our study, we found that short-term hypoxia promoted cell migration. Mechanistically, hypoxia promoted cell migration through mediating CCL2. Overexpression of CCL2 via lentivirus promoted cell migration, while silencing CCL2 via lentivirus inhibited cell migration and the production of related downstream proteins. In addition, we found that CCL2 was enriched in the ERK1/2 pathway, and the application of an ERK inhibitor in vivo and in vitro verified the upstream and downstream relationships between the CCL2 pathway and ERK1/2. Western blot results both in vivo and in vitro demonstrated that early short-term hypoxia promotes epidermal cell migration by activating the CCL2-ERK1/2 pathway and EMT during wound healing. Conclusions Our work demonstrated that hypoxia in the early stage serves as a stimulus for triggering wound healing through activating the CCL2-ERK1/2 pathway and EMT, which promote epidermal cell migration and accelerate wound closure. These findings provide additional detailed insights into the mechanism of wound healing and new targets for clinical treatment.
Collapse
Affiliation(s)
- Linbo Jin
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shiqi Zhou
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shihan Zhao
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Junhui Long
- Department of Dermatology, Southwest Hospital Jiangbei Area (The 958th hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Zhidan Huang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Junli Zhou
- Department of Burn and Plastic Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Yiming Zhang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| |
Collapse
|
299
|
Wu Z, Ke Q, Jiang L, Hong H, Pan W, Chen W, Abudukeremu X, She F, Chen Y. TGF-β1 facilitates gallbladder carcinoma metastasis by regulating FOXA1 translation efficiency through m 6A modification. Cell Death Dis 2024; 15:422. [PMID: 38886389 PMCID: PMC11183149 DOI: 10.1038/s41419-024-06800-9] [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: 10/13/2023] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
TGF-β1 plays a pivotal role in the metastatic cascade of malignant neoplasms. N6-methyladenosine (m6A) stands as one of the most abundant modifications on the mRNA transcriptome. However, in the metastasis of gallbladder carcinoma (GBC), the effect of TGF-β1 with mRNA m6A modification, especially the effect of mRNA translation efficiency associated with m6A modification, remains poorly elucidated. Here we demonstrated a negative correlation between FOXA1 and TGF-β1 expression in GBC. Overexpression of FOXA1 inhibited TGF-β1-induced migration and epithelial-mesenchymal transition (EMT) in GBC cells. Mechanistically, we confirmed that TGF-β1 suppressed the translation efficiency of FOXA1 mRNA through polysome profiling analysis. Importantly, both in vivo and in vitro experiments showed that TGF-β1 promoted m6A modification on the coding sequence (CDS) region of FOXA1 mRNA, which was responsible for the inhibition of FOXA1 mRNA translation by TGF-β1. We demonstrated through MeRIP and RIP assays, dual-luciferase reporter assays and site-directed mutagenesis that ALKBH5 promoted FOXA1 protein expression by inhibiting m6A modification on the CDS region of FOXA1 mRNA. Moreover, TGF-β1 inhibited the binding capacity of ALKBH5 to the FOXA1 CDS region. Lastly, our study confirmed that overexpression of FOXA1 suppressed lung metastasis and EMT in a nude mice lung metastasis model. In summary, our research findings underscore the role of TGF-β1 in regulating TGF-β1/FOXA1-induced GBC EMT and metastasis by inhibiting FOXA1 translation efficiency through m6A modification.
Collapse
Affiliation(s)
- Zhenheng Wu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Qiming Ke
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Lei Jiang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Haijie Hong
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Wei Pan
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Wen Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Xiahenazi Abudukeremu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Feifei She
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China.
| | - Yanling Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China.
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
300
|
Chen H, Zhang W, Shi J, Tang Y, Chen X, Li J, Yao X. Study on the mechanism of S100A4-mediated cancer oncogenesis in uveal melanoma cells through the integration of bioinformatics and in vitro experiments. Gene 2024; 911:148333. [PMID: 38431233 DOI: 10.1016/j.gene.2024.148333] [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: 12/14/2023] [Revised: 02/13/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND The elevated metastasis rate of uveal melanoma (UM) is intricately correlated with patient prognosis, significantly affecting the quality of life. S100 calcium-binding protein A4 (S100A4) has tumorigenic properties; therefore, the present study investigated the impact of S100A4 on UM cell proliferation, apoptosis, migration, and invasion using bioinformatics and in vitro experiments. METHODS Bioinformatic analysis was used to screen S100A4 as a hub gene and predict its possible mechanism in UM cells, and the S100A4 silencing cell line was constructed. The impact of S100A4 silencing on the proliferative ability of UM cells was detected using the Cell Counting Kit-8 and colony formation assays. Annexin V-FITC/PI double fluorescence and Hoechst 33342 staining were used to observe the effects of apoptosis on UM cells. The effect of S100A4 silencing on the migratory and invasive capabilities of UM cells was assessed using wound healing and Transwell assays. Western blotting was used to detect the expression of related proteins. RESULTS The present study found that S100A4 is a biomarker of UM, and its high expression is related to poor prognosis. After constructing the S100A4 silencing cell line, cell viability, clone number, proliferating cell nuclear antigen, X-linked inhibitor of apoptosis protein, and survivin expression were decreased in UM cells. The cell apoptosis rate and relative fluorescence intensity increased, accompanied by increased levels of Bax and caspase-3 and decreased levels of Bcl-2. Additionally, a decrease in the cell migration index and relative invasion rate was observed with increased E-cadherin expression and decreased N-cadherin and vimentin protein expression. CONCLUSION S100A4 silencing can inhibit the proliferation, migration, and invasion and synchronously induces apoptosis in UM cells.
Collapse
Affiliation(s)
- Huimei Chen
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenqing Zhang
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Jian Shi
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Yu Tang
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiong Chen
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiangwei Li
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiaolei Yao
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|