1
|
Li X, Wu Y, Xie B, Xu M, Xie T, Yue W, Lin M, Lin Y, Chen Y. SPP1 Promotes NSCLC Brain Metastasis Via Sequestration of Ubiquitin Ligase RNF114 to Facilitate P85α Ubiquitination. Mol Carcinog 2025; 64:829-841. [PMID: 39918025 DOI: 10.1002/mc.23866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 04/12/2025]
Abstract
Brain metastasis (BM) is a significant factor contributing to the poor prognosis of patients with non-small cell lung cancer (NSCLC). Secreted phosphoprotein 1 (SPP1) is implicated in the progression and metastasis of several cancers. The role of SPP1 in NSCLC remains unclear, especially in NSCLC BM. This study aimed to identify genes associated with NSCLC BM and to investigate the involvement of SPP1 in NSCLC BM. Integrated genomic analysis was utilized to identify candidate genes in NSCLC. The expression levels of SPP1 were evaluated in NSCLC tissues and cell lines. In vitro and in vivo experiments were conducted to assess the effect of SPP1 on NSCLC cell behavior and BM. The potential mechanisms of SPP1 were demonstrated by CO-IP and liquid chromatography-mass spectrometry (LC-MS). The underlying mechanism involving the PI3K/AKT/mTOR pathway was explored. The results showed that SPP1 expression was upregulated in NSCLC tissues and cell lines. Depletion of SPP1 using shRNA inhibited cell proliferation, migration, and invasion in vitro and suppressed BM in vivo. Mechanistically, SPP1 facilitates the ubiquitination of P85α by interacting with the ubiquitin ligase RNF114, thus playing a role in regulating NSCLC BM through the PI3K/AKT/mTOR signaling pathway. Moreover, immunohistochemistry staining confirmed higher expression of SPP1 in NSCLC tissues with BM compared to those without BM. In summary, elevated SPP1 expression was associated with poor clinical outcomes in NSCLC patients. This study highlights the role of SPP1 as a regulator of cell metastasis and suggests its potential as a novel therapeutic target for BM in NSCLC.
Collapse
Affiliation(s)
- Xiaoqin Li
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Yun Wu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Baosong Xie
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Tianjian Xie
- Xiapu County Hospital of Fujian Province, Ningde, China
| | - Wenxiang Yue
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ming Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Ying Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, China
| | - Yusheng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Fujian Provincial Researching Laboratory of Respiratory Diseases, Fuzhou, China
| |
Collapse
|
2
|
Deng L, Ren J, Liu D, Li H, Yang G, Wang K, Song Y, Su H. Ran drives pancreatic cancer metastasis by activating the osteopontin-PI3K/AKT-androgen receptor signaling cascade. Toxicol Appl Pharmacol 2025; 499:117328. [PMID: 40187660 DOI: 10.1016/j.taap.2025.117328] [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: 10/30/2024] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The small GTPase Ran has emerged as a key player in cancer metastasis. Our previous studies demonstrated that Ran drives pancreatic cancer metastasis by modulating androgen receptor (AR) expression. However, the detailed mechanisms by which Ran regulates AR expression remain unclear. This study aimed to elucidate the regulatory mechanisms through which Ran influences AR expression in the context of pancreatic cancer metastasis. We observed elevated levels of Ran, osteopontin (OPN), and AR in metastatic lymph node tissues, with OPN positively correlated with either Ran or AR expression. Ran silencing led to decreased levels of OPN and AR, whereas Ran upregulation increased their expression. Notably, OPN overexpression restored AR levels in Ran-silenced cells, whereas OPN knockdown diminished the inductive effect of Ran on AR expression. Additionally, OPN knockdown decreased AR expression and was associated with reduced activation of the PI3K/AKT signaling pathway. Functional assays revealed that silencing OPN significantly impaired the mobility and invasion of pancreatic cancer cells and restricted hepatic metastasis. Conversely, OPN overexpression restored the impaired metastasis caused by Ran knockdown. Furthermore, inhibiting PI3K/AKT signaling abolished the promoting effects of either Ran or OPN on pancreatic cancer metastasis. Importantly, re-expressing AR reversed the inhibitory effects of Ran or OPN silencing on the mobility and invasion of pancreatic cancer cells. In summary, Ran induces AR expression through the regulation of the OPN-PI3K/AKT signaling cascade. The Ran-OPN-PI3K/AKT-AR signaling pathway is crucial for driving pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Lin Deng
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Jingyi Ren
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Dong Liu
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Hong Li
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Guang Yang
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Kairui Wang
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
3
|
Zhou M, Li R, Lian G, Yang M, Li L, Yin Z, Li G, Zhao J, Tan R. Tetrahydrocurcumin alleviates colorectal tumorigenesis by modulating the SPP1/CD44 axis and preventing M2 tumor-associated macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156674. [PMID: 40220425 DOI: 10.1016/j.phymed.2025.156674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Recent studies show that secreted phosphoprotein 1 (SPP1) is linked to the progression of various cancers, including colorectal cancer (CRC). SPP1 also promotes M2 macrophage polarization, contributing to immune evasion in the tumor microenvironment. Tetrahydrocurcumin (THC) has been reported to alleviate CRC, but the mechanism remains unclear. PURPOSE The study aimed to explore how THC modulated the SPP1/CD44 axis to inhibit M2 polarization and suppress CRC development. METHODS Azoxymethane/dextran sulfate sodium (AOM/DSS)-induced mouse model was used to assess the anti-CRC effects of THC. Transcriptome sequencing was conducted to identify the key targets of THC in CRC. The effects of THC on CRC cells were evaluated by CCK-8, colony formation, migration assays, immunofluorescence staining and flow cytometry. Human monocytic cells, THP-1, and colon cancer cell line, HCT116, were co-cultured, both directly or indirectly, to mimic the tumor-macrophage interactions, and investigate the role of SPP1/CD44 axis and the intervention effect of THC. RESULTS THC significantly inhibited CRC carcinogenesis in mice and improved pathologic symptoms, serum inflammatory markers, and intestinal barrier integrity. THC inhibited CRC cell proliferation, migration and colony formation, while promoting apoptosis. Transcriptome analysis identified SPP1 as a key target of THC against CRC. SPP1 facilitated CRC progression by activating the ERK signaling pathway and maintaining the M2-like phenotype of macrophage, which further exacerbated this response. THC inhibited CRC development by targeting the SPP1/CD44 axis, rather than the integrin pathway. CONCLUSIONS SPP1 played a crucial role in maintaining the M2 phenotype of macrophage and promoting CRC cells proliferation. THC inhibited the activation of ERK signals in CRC cells and phenotypic transformation of M2-like macrophages through the SPP1/CD44 axis, thereby regulating the tumor immune microenvironment to exert anti-CRC effect.
Collapse
Affiliation(s)
- Mengting Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rui Li
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China.
| | - Guiyun Lian
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Mengni Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Guiyu Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Junning Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; National Key Laboratory of Drug Regulatory Science, National Medical Products Administration (NMPA), Beijing 100038, China; National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
4
|
Pasha A, Ravinder D, Pawar SC. Andrographolide Mitigates Cisplatin Resistance by Inhibiting SPP1 Regulated NF-kB/iNOS/COX-2 and PI3K/AKT Pathway in Cisplatin Resistant Cervical Carcinoma Cells. Drug Dev Res 2025; 86:e70052. [PMID: 39888044 DOI: 10.1002/ddr.70052] [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: 09/11/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Drug resistance and cancer recurrence are major cause of Cervical cancer (CC) patient mortality. Cisplatin (CDDP) is the major drug that has been extremely used in all stages in treating CC, although relapse and malignant instances have been observed as a result of cisplatin resistance in CC. In the present study, we established Cisplatin resistant CC HeLa cell line model and the cytotoxic effects of Andro as a single agent or in combination with CDDP were investigated to assess its potential as a chemotherapeutic agent in cisplatin-resistant HeLa (CisR-HeLa) cells. Andro enhanced the cytotoxicity of CDDP in CisR-HeLa cells and shown a synergistic effect by reducing cell viability, proliferation, migration, invasion, and inducing apoptosis in cisplatin resistant cells. Furthermore, we evaluated the expression levels of inflammatory and oncogenic proteins, SPP1, NF-kB, iNOS, COX-2, and the PI3K/AKT signaling pathway, which are associated with cisplatin resistance, as well as using Andro to regulate the targeted markers in CisR-HeLa cells to overcome resistance. The results show that suppressing SPP1 and NF-kB by Andro alone or in combination with CDDP regulates iNOS, COX-2, and increases PTEN expression. The addition of Andro to CDDP inhibited PI3K and AKT expression as well as triggered synergistic apoptosis, which could be associated with variations in Bax and Bcl-2 protein levels. The results suggest that Andro in combination with CDDP exhibits synergistic anti-tumor growth efficacy that targets multiple inflammatory markers, resulting in a promising treatment option for individuals with recurrent cancer due to drug resistance and advanced CC.
Collapse
Affiliation(s)
- Akbar Pasha
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Doneti Ravinder
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Smita C Pawar
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad, Telangana, India
| |
Collapse
|
5
|
Liu C, Wu K, Li C, Zhang Z, Zhai P, Guo H, Zhang J. SPP1+ macrophages promote head and neck squamous cell carcinoma progression by secreting TNF-α and IL-1β. J Exp Clin Cancer Res 2024; 43:332. [PMID: 39726047 DOI: 10.1186/s13046-024-03255-w] [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: 07/31/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a very aggressive disease characterized by a heterogeneous tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs) constitute the major innate immune population in the TIME where they facilitate crucial regulatory processes that participate in malignant tumor progression. SPP1 + macrophages (SPP1 + Macs) are found in many cancers, but their effects on HNSCC remain unknown. This study aimed to identify and validate the role and function of SPP1 + Macs in the malignant progression of HNSCC. METHODS In this study, we applied single-cell RNA sequencing (scRNA-seq) analyses of paired tumor and normal tissues from 5 HNSCC patients to identify tumor-specific SPP1 + Macs. RT-qPCR and multiplex immunohistochemical and multiplex immunofluorescence staining were used to verify the presence of SPP1 + Macs in the clinical samples. Gene set variation analysis suggested that SPP1 + Macs were actively involved in cytokine production. Thus, we constructed SPP1-OE macrophages and SPP1-KD macrophages (both differentiated from THP-1 cells), performed a Luminex liquid suspension chip detection assay to detect differential cytokines, and further assessed their biological functions and mechanisms in several HNSCC cell lines and adjacent macrophages. An in vivo experiment was used to verify the function of SPP1 + Macs in HNSCC progression. RESULTS The scRNA-seq results revealed that myeloid cells were heterogeneous and strongly correlated with tumor cells in the TIME in HNSCC and identified tumor-specific SPP1 + Macs, which were positively correlated with poor prognosis of HNSCC patients. Gene set variation analysis (GSVA) suggested that SPP1 + Macs were actively involved in cytokine production. Luminex liquid suspension chip detection assay indicated that SPP1 + Mac-derived TNF-α and IL-1β played important roles. Both in vitro and in vivo experiments and the use of VGX-1027, an inhibitor of macrophage-derived TNF-α and IL-1β, confirmed that SPP1 + Mac-derived TNF-α and IL-1β promoted HNSCC progression by supporting tumor cell proliferation and migration. Mechanistically, we found that TNF-α and IL-1β were upregulated due to NF-kappa B signaling pathway activation in SPP1 + Macs. Moreover, SPP1 + Mac-derived TNF-α and IL-1β promoted the expression of OPN in both tumor cells and other adjacent macrophages through different signaling pathways. CONCLUSIONS SPP1 + Macs increase the secretion of TNF-α and IL-1β via the NF-kappa B pathway to promote HNSCC cell proliferation, and TNF-α and IL-1β in turn upregulate the expression of OPN in tumor cells and macrophages; thus, SPP1 + Macs may be a candidate target through which antitumor efficacy can be enhanced.
Collapse
Affiliation(s)
- Chun Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Wu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral and Maxillofacial Surgery, Second Xiangya Hospital of Central South University, Changsha, China
| | - Chuwen Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyan Guo
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianjun Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Zhao L, Wang Z, Tan Y, Ma J, Huang W, Zhang X, Jin C, Zhang T, Liu W, Yang YG. IL-17A/CEBPβ/OPN/LYVE-1 axis inhibits anti-tumor immunity by promoting tumor-associated tissue-resident macrophages. Cell Rep 2024; 43:115039. [PMID: 39643970 DOI: 10.1016/j.celrep.2024.115039] [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: 06/29/2024] [Revised: 10/18/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a critical component of the immunosuppressive tumor microenvironment, comprising monocyte-derived macrophages (MDM-TAMs) and tissue-resident macrophages (TRM-TAMs). Here, we discovered that TRM-TAMs mediate the pro-tumor effects of interleukin (IL)-17A and that IL-17A-driven tumor progression requires tumor cell production of osteopontin (OPN). Mechanistically, we identified CEBPβ as a transcription factor downstream of IL-17A in tumor cells and LYVE-1 as an OPN receptor on TRM-TAMs. IL-17A stimulates tumor cell production of OPN, and OPN/LYVE-1 signaling activates the JNK/c-Jun pathway, leading to the proliferation of immunosuppressive LYVE-1+ TRM-TAMs. Unlike its effect on LYVE-1+ TRM-TAMs, OPN interacts with α4β1 to promote the chemotaxis of LYVE-1- MDM-TAMs toward tumors. IL-17A neutralization, OPN inactivation in tumor cells, or LYVE-1 deletion in macrophages inhibited TAMs and enhanced anti-tumor immune responses and anti-PDL1 therapy. Thus, the IL-17A/CEBPβ/OPN/LYVE-1 axis offers a mechanism suppressing anti-tumor immune responses and, hence, an effective therapeutic target for cancer.
Collapse
Affiliation(s)
- Lei Zhao
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Zonghan Wang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Yuying Tan
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Jianan Ma
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Wei Huang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Xiaoying Zhang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Chunhui Jin
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China; Department of Pathology, The First Hospital of Jilin University, Changchun, China
| | - Ting Zhang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China; International Center of Future Science, Jilin University, Changchun, China.
| |
Collapse
|
7
|
Yue B, Xiong D, Chen J, Yang X, Zhao J, Shao J, Wei D, Gao F, Huang M, Chen J. SPP1 induces idiopathic pulmonary fibrosis and NSCLC progression via the PI3K/Akt/mTOR pathway. Respir Res 2024; 25:362. [PMID: 39369217 PMCID: PMC11456247 DOI: 10.1186/s12931-024-02989-7] [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: 05/16/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND The prevalence of non-small cell lung cancer (NSCLC) is notably elevated in individuals diagnosed with idiopathic pulmonary fibrosis (IPF). Secreted phosphoprotein 1 (SPP1), known for its involvement in diverse physiological processes, including oncogenesis and organ fibrosis, has an ambiguous role at the intersection of IPF and NSCLC. Our study sought to elucidate the function of SPP1 within the pathogenesis of IPF and its subsequent impact on NSCLC progression. METHODS Four GEO datasets was analyzed for common differential genes and TCGA database was used to analyze the prognosis. The immune infiltration was analyzed by TIMER database. SPP1 expression was examined in human lung tissues, the IPF fibroblasts and the BLM-induced mouse lung fibrosis model. Combined with SPP1 gene gain- and loss-of-function, qRT-PCR, Western blot, EdU and CCK-8 experiments were performed to evaluate the effects and mechanisms of SPP1 in IPF progression. Effect of SPP1 on NSCLC was detected by co-cultured IPF fibroblasts and NSCLC cells. RESULTS Through bioinformatics analysis, we observed a significant overexpression of SPP1 in both IPF and NSCLC patient datasets, correlating with enhanced immune infiltration of cancer-associated fibroblasts in NSCLC. Elevated levels of SPP1 were detected in lung tissue samples from IPF patients and bleomycin-induced mouse models, with partial colocalization observed with α-smooth muscle actin. Knockdown of SPP1 inhibits TGF-β1-induced differentiation of fibroblasts to myofibroblasts and the proliferation of IPF fibroblasts. Conversely, SPP1 overexpression promoted IPF fibroblast proliferation via PI3K/Akt/mTOR pathway. Furthermore, IPF fibroblasts promoted NSCLC cell proliferation and activated the PI3K/Akt/mTOR pathway; these effects were attenuated by SPP1 knockdown in IPF fibroblasts. CONCLUSIONS Our findings suggest that SPP1 functions as a molecule promoting both fibrosis and tumorigenesis, positioning it as a prospective therapeutic target for managing the co-occurrence of IPF and NSCLC.
Collapse
Affiliation(s)
- Bingqing Yue
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Dian Xiong
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Juan Chen
- Department of General Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xiucheng Yang
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jin Zhao
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jingbo Shao
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Dong Wei
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Fei Gao
- Department of Emergency, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Man Huang
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Department of General Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Multiple Organ Failure, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyu Chen
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China.
| |
Collapse
|
8
|
Panda VK, Mishra B, Nath AN, Butti R, Yadav AS, Malhotra D, Khanra S, Mahapatra S, Mishra P, Swain B, Majhi S, Kumari K, Radharani NNV, Kundu GC. Osteopontin: A Key Multifaceted Regulator in Tumor Progression and Immunomodulation. Biomedicines 2024; 12:1527. [PMID: 39062100 PMCID: PMC11274826 DOI: 10.3390/biomedicines12071527] [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: 05/11/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) is composed of various cellular components such as tumor cells, stromal cells including fibroblasts, adipocytes, mast cells, lymphatic vascular cells and infiltrating immune cells, macrophages, dendritic cells and lymphocytes. The intricate interplay between these cells influences tumor growth, metastasis and therapy failure. Significant advancements in breast cancer therapy have resulted in a substantial decrease in mortality. However, existing cancer treatments frequently result in toxicity and nonspecific side effects. Therefore, improving targeted drug delivery and increasing the efficacy of drugs is crucial for enhancing treatment outcome and reducing the burden of toxicity. In this review, we have provided an overview of how tumor and stroma-derived osteopontin (OPN) plays a key role in regulating the oncogenic potential of various cancers including breast. Next, we dissected the signaling network by which OPN regulates tumor progression through interaction with selective integrins and CD44 receptors. This review addresses the latest advancements in the roles of splice variants of OPN in cancer progression and OPN-mediated tumor-stromal interaction, EMT, CSC enhancement, immunomodulation, metastasis, chemoresistance and metabolic reprogramming, and further suggests that OPN might be a potential therapeutic target and prognostic biomarker for the evolving landscape of cancer management.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Ramesh Butti
- Division of Hematology and Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX 75235, USA;
| | - Amit Singh Yadav
- Biomedical Centre, Faculty of Medicine, Lund University, 223 62 Lund, Sweden; (A.S.Y.); (N.N.V.R.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
| | - N. N. V. Radharani
- Biomedical Centre, Faculty of Medicine, Lund University, 223 62 Lund, Sweden; (A.S.Y.); (N.N.V.R.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (A.N.N.); (D.M.); (S.K.); (S.M.); (P.M.); (B.S.); (S.M.); (K.K.)
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Bhubaneswar 751024, India
| |
Collapse
|
9
|
Zhou R, Li R, Ding Q, Zhang Y, Yang H, Han Y, Liu C, Liu J, Wang S. OPN silencing reduces hypoxic pulmonary hypertension via PI3K-AKT-induced protective autophagy. Sci Rep 2024; 14:8670. [PMID: 38622371 PMCID: PMC11018812 DOI: 10.1038/s41598-024-59367-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] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is a pulmonary vascular disease primarily characterized by progressive pulmonary vascular remodeling in a hypoxic environment, posing a significant clinical challenge. Leveraging data from the Gene Expression Omnibus (GEO) and human autophagy-specific databases, osteopontin (OPN) emerged as a differentially expressed gene, upregulated in cardiovascular diseases such as pulmonary arterial hypertension (PAH). Despite this association, the precise mechanism by which OPN regulates autophagy in HPH remains unclear, prompting the focus of this study. Through biosignature analysis, we observed significant alterations in the PI3K-AKT signaling pathway in PAH-associated autophagy. Subsequently, we utilized an animal model of OPNfl/fl-TAGLN-Cre mice and PASMCs with OPN shRNA to validate these findings. Our results revealed right ventricular hypertrophy and elevated mean pulmonary arterial pressure (mPAP) in hypoxic pulmonary hypertension model mice. Notably, these effects were attenuated in conditionally deleted OPN-knockout mice or OPN-silenced hypoxic PASMCs. Furthermore, hypoxic PASMCs with OPN shRNA exhibited increased autophagy compared to those in hypoxia alone. Consistent findings from in vivo and in vitro experiments indicated that OPN inhibition during hypoxia reduced PI3K expression while increasing LC3B and Beclin1 expression. Similarly, PASMCs exposed to hypoxia and PI3K inhibitors had higher expression levels of LC3B and Beclin1 and suppressed AKT expression. Based on these findings, our study suggests that OPNfl/fl-TAGLN-Cre effectively alleviates HPH, potentially through OPN-mediated inhibition of autophagy, thereby promoting PASMCs proliferation via the PI3K-AKT signaling pathway. Consequently, OPN emerges as a novel therapeutic target for HPH.
Collapse
Affiliation(s)
- Rui Zhou
- Qinghai University Medical Department, Xining, 810016, China
| | - Ran Li
- Zhengzhou Medical and Health Vocational College, Zhengzhou, 452385, China
| | - Qi Ding
- Pathology Department of Tianjin Huanghe Hospital, Tianjin, 300110, China
| | - Yuwei Zhang
- Department of Public Health, School of Medical, Qinghai University, Xining, 810016, China
| | - Hui Yang
- Qinghai University Medical Department, Xining, 810016, China
| | - Ying Han
- Qinghai University Medical Department, Xining, 810016, China
| | - Chuanchuan Liu
- Key Laboratory of Hydatid Disease, Qinghai University, Xining, 810001, China
| | - Jie Liu
- Qinghai University Medical Department, Xining, 810016, China
| | - Shenglan Wang
- Qinghai University Medical Department, Xining, 810016, China.
| |
Collapse
|
10
|
Poleboyina PK, Alagumuthu M, Pasha A, Ravinder D, Pasumarthi D, Pawar SC. Entrectinib a Plausible Inhibitor for Osteopontin (SPP1) in Cervical Cancer-Integrated Bioinformatic Approach. Appl Biochem Biotechnol 2023; 195:7766-7795. [PMID: 37086377 DOI: 10.1007/s12010-023-04541-7] [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] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
Cervical cancer is one of the major causes of death in women, especially in developing countries bearing more than a quarter of the global burden. Secreted phosphoprotein-1, also known as OPN (osteopontin), is an integrin-binding glycophosphoprotein that is overexpressed in a variety of tumors. OPN is a chemokine-like calcified ECM-associated protein that plays a crucial role in evaluating the metastatic potential of various cancers. However, the role of SPP1 in the tumor microenvironment and associated signaling pathways in CC is still unclear. In our study, three CC microarray datasets (GSE9750, GSE46857, and GSE67522) were obtained from the GEO database to identify the differentially expressed genes. Enrichment analysis was carried out by Enrichr and ShinyGO and the PPI interaction network was created by using String and Cytoscape. GEPIA datasets were used to validate the top 10 hub genes, and virtual screening, docking, and dynamic simulation studies were used to identify a suitable inhibitor against the OPN protein using MVD, PyRx, and GROMACS respectively. Our results show that a total of 11 DEGs were common for three datasets and gene ontology pathway enrichment analysis revealed that 2 biological processes i.e. programmed cell death and animal organ development commonly affected mechanisms in all three datasets. Docking and dynamic studies revealed that Entrectinib showed excellent binding affinity against OPN protein. Based on the results, we conclude that OPN is one of the most upregulated genes in cervical cancer and Entrectinib emerges to be a promising potential OPN inhibitor to curtail cervical cancer progression. Schematic representation: The schematic representation of methodology steps is illustrated in the graphical abstract. Schematic representation of methodology.
Collapse
Affiliation(s)
- Pavan Kumar Poleboyina
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Manikandan Alagumuthu
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, -632014, Vellore, India
| | - Akbar Pasha
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Doneti Ravinder
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Deepthi Pasumarthi
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India
| | - Smita C Pawar
- Department of Genetics & Biotechnology, University College of Science, Osmania University, -500007, Hyderabad, Telangana, India.
| |
Collapse
|
11
|
Yang L, Zhu Z, Zheng Y, Yang J, Liu Y, Shen T, Li M, He H, Huang H, Dai W. RAB6A functions as a critical modulator of the stem-like subsets in cholangiocarcinoma. Mol Carcinog 2023; 62:1460-1473. [PMID: 37278569 DOI: 10.1002/mc.23589] [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: 02/22/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
RAB6A is a member of RAB GTPase family and plays an important role in the targeted transport of neurotrophic receptors and inflammatory cytokines. RAB6A-mediated secretory pathway is involved in many physiological and pathological processes. Defects in RAB6A-mediated secretory pathway may lead to the development of many diseases, including cancer. However, its role in cholangiocarcinoma (CCA) has not yet been revealed. We explored the regulatory role of RAB6A in the stem-like subsets of CCA. We showed that RAB6A knockdown (KD) impedes cancer stem cells (CSCs) properties and epithelial-mesenchymal transition in vitro and that suppression of RAB6A inhibits tumor growth in vivo. We screened target cargos of RAB6A in CCA cells and identified a extracellular matrix component as the target cargo. RAB6A binds directly to OPN, and RAB6A KD suppressed OPN secretion and inhibited the interaction between OPN and αV integrin receptor. Moreover, RAB6A KD inhibited the AKT signaling pathway, which is a downstream effector of the integrin receptor signaling. In addition, shRNA targeting OPN blocked endogenous expression of OPN and consequently weakened CSCs properties in RAB6A-formed spheres. Similarly, inhibitor of AKT signaling, MK2206 also impedes oncogenic function of RAB6A in the stem-like subsets of CCA cells. In conclusion, our findings showed that RAB6A sustains CSCs phenotype maintenance by modulating the secretion of OPN and consequentially activating the downstream AKT signaling pathway. Targeting the RAB6A/OPN axis may be an effective strategy for CCA therapy.
Collapse
Affiliation(s)
- Liangfang Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhiwen Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yang Zheng
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Yang
- Institution of Plastic Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuxin Liu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tingyun Shen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mingyi Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huijuan He
- Clinical Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haili Huang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Institution of Plastic Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Dai
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
12
|
Yang J, Liu K, Yang L, Ji J, Qin J, Deng H, Wang Z. Identification and validation of a novel cuproptosis-related stemness signature to predict prognosis and immune landscape in lung adenocarcinoma by integrating single-cell and bulk RNA-sequencing. Front Immunol 2023; 14:1174762. [PMID: 37287976 PMCID: PMC10242006 DOI: 10.3389/fimmu.2023.1174762] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/11/2023] [Indexed: 06/09/2023] Open
Abstract
Background Cancer stem cells (CSCs) play vital roles in lung adenocarcinoma (LUAD) recurrence, metastasis, and drug resistance. Cuproptosis has provided a novel insight into the treatment of lung CSCs. However, there is a lack of knowledge regarding the cuproptosis-related genes combined with the stemness signature and their roles in the prognosis and immune landscape of LUAD. Methods Cuproptosis-related stemness genes (CRSGs) were identified by integrating single-cell and bulk RNA-sequencing data in LUAD patients. Subsequently, cuproptosis-related stemness subtypes were classified using consensus clustering analysis, and a prognostic signature was constructed by univariate and least absolute shrinkage operator (LASSO) Cox regression. The association between signature with immune infiltration, immunotherapy, and stemness features was also investigated. Finally, the expression of CRSGs and the functional roles of target gene were validated in vitro. Results We identified six CRSGs that were mainly expressed in epithelial and myeloid cells. Three distinct cuproptosis-related stemness subtypes were identified and associated with the immune infiltration and immunotherapy response. Furthermore, a prognostic signature was constructed to predict the overall survival (OS) of LUAD patients based on eight differently expressed genes (DEGs) with cuproptosis-related stemness signature (KLF4, SCGB3A1, COL1A1, SPP1, C4BPA, TSPAN7, CAV2, and CTHRC1) and confirmed in validation cohorts. We also developed an accurate nomogram to improve clinical applicability. Patients in the high-risk group showed worse OS with lower levels of immune cell infiltration and higher stemness features. Ultimately, further cellular experiments were performed to verify the expression of CRSGs and prognostic DEGs and demonstrate that SPP1 could affect the proliferation, migration, and stemness of LUAD cells. Conclusion This study developed a novel cuproptosis-related stemness signature that can be used to predict the prognosis and immune landscape of LUAD patients, and provided potential therapeutic targets for lung CSCs in the future.
Collapse
Affiliation(s)
- Jia Yang
- *Correspondence: Zhongqi Wang, ; Jia Yang,
| | | | | | | | | | | | | |
Collapse
|
13
|
Xu Z, Xi F, Deng X, Ni Y, Pu C, Wang D, Lou W, Zeng X, Su N, Chen C, Zeng Z, Deng L, Jiang M. Osteopontin Promotes Macrophage M1 Polarization by Activation of the JAK1/STAT1/HMGB1 Signaling Pathway in Nonalcoholic Fatty Liver Disease. J Clin Transl Hepatol 2023; 11:273-283. [PMID: 36643029 PMCID: PMC9817049 DOI: 10.14218/jcth.2021.00474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/12/2022] [Accepted: 05/05/2022] [Indexed: 01/18/2023] Open
Abstract
Background and Aims Osteopontin (OPN) is reported to be associated with the pathogenesis of nonalcoholic fatty liver disease (NAFLD). However, the function of OPN in NAFLD is still inconclusive. Therefore, our aim in this study was to evaluate the role of OPN in NAFLD and clarify the involved mechanisms. Methods We analyzed the expression change of OPN in NAFLD by bioinformatic analysis, qRT-PCR, western blotting and immunofluorescence staining. To clarify the role of OPN in NAFLD, the effect of OPN from HepG2 cells on macrophage polarization and the involved mechanisms were examined by FACS and western blotting. Results OPN was significantly upregulated in NAFLD patients compared with normal volunteers by microarray data, and the high expression of OPN was related with disease stage and progression. OPN level was also significantly increased in liver tissue samples of NAFLD from human and mouse, and in HepG2 cells treated with oleic acid (OA). Furthermore, the supernatants of OPN-treated HepG2 cells promoted the macrophage M1 polarization. Mechanistically, OPN activated the janus kinase 1(JAK1)/signal transducers and activators of transcription 1 (STAT1) signaling pathway in HepG2 cells, and consequently HepG2 cells secreted more high-mobility group box 1 (HMGB1), thereby promoting macrophage M1 polarization. Conclusions OPN promoted macrophage M1 polarization by increasing JAK1/STAT1-induced HMGB1 secretion in hepatocytes.
Collapse
Affiliation(s)
- Zhihao Xu
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Feiyang Xi
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Xinxin Deng
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
- School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Yuqi Ni
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Changqin Pu
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Dan Wang
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Weiming Lou
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Xufang Zeng
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Ning Su
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Chen Chen
- School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi, China
| | - Ziqiang Zeng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Libin Deng
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Meixiu Jiang
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
The Intracellular and Secreted Sides of Osteopontin and Their Putative Physiopathological Roles. Int J Mol Sci 2023; 24:ijms24032942. [PMID: 36769264 PMCID: PMC9917417 DOI: 10.3390/ijms24032942] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Classically, osteopontin (OPN) has been described as a secreted glycophosprotein. Indeed, most data concerning its physiological and pathological roles are mainly related to the secreted OPN (sOPN). However, there are several instances in which intracellular OPN (iOPN) has been described, presenting some specific roles in distinct experimental models, such as in the immune system, cancer cells, and neurological disorders. We herein aimed to highlight and discuss some of these secreted and intracellular roles of OPN and their putative clinical and biological impacts. Moreover, by consolidating data from the OPN protein database, we also analyzed the occurrence of signal peptide (SP) sequences and putative subcellular localization, especially concerning currently known OPN splicing variants (OPN-SV). Comprehending the roles of OPN in its distinct cellular and tissue environments may provide data regarding the additional applications of this protein as biomarkers and targets for therapeutic purposes, besides further describing its pleiotropic roles.
Collapse
|
15
|
An anti-alcoholism drug, disulfiram and copper complex improves radio-resistance of tumor-initiating cells in esophageal squamous cell carcinoma. Esophagus 2023; 20:134-142. [PMID: 36121574 DOI: 10.1007/s10388-022-00948-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/13/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignant cancer with a poor prognosis. Chemoradiotherapy is one of the most important strategies for patients with locally advanced unresectable ESCC; however, its therapeutic effect is unsatisfactory. Tumor-initiating cells (TICs) have been reported to be resistant to conventional chemotherapy and radiotherapy so far. Therefore, we aimed to develop a treatment strategy targeting TICs in ESCC to improve radiosensitivity. METHODS First, we validated aldehyde dehydrogenase 1 (ALDH1) as a TIC marker and investigated its ability to mediate resistance in human ESCC cell lines using flow cytometry, Western blotting, and functional analyses. Then, we focused on disulfiram (DSF), an aldehyde dehydrogenase inhibitor, used to treat alcohol use disorder. We investigated the effect of DSF and copper (II) D-gluconate (Cu) on the radiosensitivity of ESCC in xenograft mouse models. RESULTS ALDH1-positive cells showed an upregulation of SOX2 and Nanog, exhibiting much stronger tumor-initiating properties than ALDH1-negative cells. Furthermore, inhibition of ALDH1 attenuated the tumor-initiating properties of ESCC cell lines. Our results also showed that ALDH1-positive cells were resistant to chemotherapy and radiotherapy, and the inhibition of ALDH1 led to the mitigation of therapeutic resistance. Our in vitro and in vivo studies revealed that the DSF/Cu complex could radiosensitize ALDH1-positive ESCC cells and downregulate the phosphoinositide 3-kinase/Akt pathway. CONCLUSION ALDH1 inhibition by the DSF/Cu complex enhances the radiosensitivity of TICs in ESCC. The drug repositioning approach using disulfiram is a potential treatment option to overcome radioresistance in patients with locally advanced ESCC.
Collapse
|
16
|
Nakajima T, Uehara T, Iwaya M, Matsuda K, Wada M, Nagaya T, Ehara T, Ota H. Osteopontin expression in the invasive front stroma of colorectal adenocarcinoma is associated with tumor budding and prognosis. Pathol Res Pract 2022; 240:154190. [PMID: 36332325 DOI: 10.1016/j.prp.2022.154190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Tumor budding (TB) is an important prognostic factor in colorectal carcinoma (CRC). Osteopontin (OPN) functions in various processes such as immune response, migration and invasion, angiogenesis, epithelial-mesenchymal transition (EMT) and metastasis. However, the involvement of OPN and CD44v6, which is a receptor for OPN, in TB has not been clarified. Therefore, we examined the relationship of OPN with TB in CRC and compared the clinicopathological features. METHODS We investigated the expression of OPN and CD44v6 in 83 cases of CRC by immunostaining and analyzed the clinicopathological features. RESULTS OPN expression was observed mostly in the cytoplasm of stromal cells such as macrophages and fibroblasts, and rarely in cancer cells. There was a significant correlation between OPN positivity and the degree of differentiation at the invasive front and TB grade. CD44v6 was positive in cancer cells in 72 cases (86.7 %) and negative in 11 cases (13.3 %). A statistically significant effect on overall survival (OS) was identified between the OPN-positive group [median OS: 1586 (range, 30-2749) days] and the OPN-negative group [median OS: 1901 (range, 8-2665) days] (log-rank test, p = 0.011). CONCLUSIONS OPN analysis in CRC stromal cells may have prognostic implications.
Collapse
Affiliation(s)
- Tomoyuki Nakajima
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Mai Iwaya
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kazuyuki Matsuda
- Department of Biomedical Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Megumi Wada
- Department of Clinical Laboratory, Nagano Red Cross Hospital, Nagano, Japan
| | - Tadanobu Nagaya
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takehito Ehara
- Department of Surgery, North Alps Medical Center Azumi Hospital, Ikeda, Japan
| | - Hiroyoshi Ota
- Department of Biomedical Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
17
|
Giannos P, Triantafyllidis KK, Giannos G, Kechagias KS. SPP1 in infliximab resistant ulcerative colitis and associated colorectal cancer: an analysis of differentially expressed genes. Eur J Gastroenterol Hepatol 2022; 34:598-606. [PMID: 35102110 DOI: 10.1097/meg.0000000000002349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Infliximab, a tumour necrosis factor-α (TNFα) antagonist, has advanced the management of ulcerative colitis. Although efficacious, considerable percentage of patients are resistant to treatment. Accumulative inflammatory burden in long-term ulcerative colitis patients refractory to therapy increases the risk of developing colorectal cancer (CRC). Our study investigated anti-TNFα-naïve patients with active ulcerative colitis to identify gene biomarkers whose dysregulated expression correlated with resistance to infliximab (IFX) treatment and poor prognosis in CRC. METHODS Differentially expressed genes (DEGs) from two studies (GSE73661 and GSE14580) with colonic mucosal samples were retrieved. Noninflammatory bowel disease controls were compared with those with active ulcerative colitis that either responded or were resistant to IFX before treatment. DEGs from ulcerative colitis samples resistant to IFX were used to construct a protein-protein interaction network, and clustering gene modules were identified. Module DEGs that overlapped with ulcerative colitis samples responsive to IFX were analysed, based on topological closeness and radiality. Hub genes were obtained, and their correlation with CRC progression was evaluated. Their expression in CRC tissues and their tumour microenvironment immune status was estimated. RESULTS Three clusters composed of 582 DEGs from ulcerative colitis samples resistant to IFX were retrieved. Comparative analysis identified 305 overlapping DEGs with ulcerative colitis samples responsive to IFX. Topological analysis revealed a hub gene - SPP1 - whose overexpression in CRC tissues and patients correlated with increased infiltration of immune signatures and poor prognosis. CONCLUSION SPP1 may serve as potential gene biomarker and predictor of resistance to IFX therapy in ulcerative colitis and CRC development.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London
- Society of Meta-research and Biomedical Innovation, London
| | | | - Georgios Giannos
- Second Department of Surgery, Evaggelismos Hospital, Athens
- Department of Medicine, University of Crete Medical School, Heraklion, Crete, Greece
| | - Konstantinos S Kechagias
- Society of Meta-research and Biomedical Innovation, London
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London
- Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital National Health Service (NHS) Foundation Trust, London, UK
| |
Collapse
|
18
|
Amilca-Seba K, Tan TZ, Thiery JP, Louadj L, Thouroude S, Bouygues A, Sabbah M, Larsen AK, Denis JA. Osteopontin (OPN/SPP1), a Mediator of Tumor Progression, Is Regulated by the Mesenchymal Transcription Factor Slug/SNAI2 in Colorectal Cancer (CRC). Cells 2022; 11:cells11111808. [PMID: 35681502 PMCID: PMC9180003 DOI: 10.3390/cells11111808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Expression of the transcription factor Slug/SNAI2 is associated with the epithelial–mesenchymal transition (EMT) and is correlated with poorer disease-free survival in colorectal cancer (CRC). In order to decipher the basis for the Slug-mediated aggressive phenotype, we conducted RNAseq experiments with a panel of HT-29 CRC cells expressing different levels of Slug, both in vitro and in tumor models. Osteopontin (OPN), a mediator associated with tumor progression in different tumor types, was among the top upregulated genes in both cells and tumors and was the most overexpressed gene coding for a secreted protein. We further show that Slug is a direct regulator of osteopontin via binding to the OPN promoter. Interestingly, Slug expression and osteopontin secretion were correlated in vitro, as well as in tumor models, suggesting that liquid biopsies may be useful in estimating the aggressiveness phenotype of the tumor. Abstract In colorectal cancer (CRC), disease-related death is closely linked to tumor aggressiveness and metastasis. Gene expression profiling of patient tumors has suggested that a more mesenchymal phenotype, present in about one-fourth of all patients, is associated with increased aggressiveness. Accordingly, the mesenchymal transcription factor Slug/SNAI2 has been associated with decreased disease-free survival. To decipher the basis for the Slug-mediated phenotype, we conducted RNAseq experiments with a panel of HT-29 CRC cells expressing different levels of Slug, both in vitro and in tumor models. The results show that osteopontin, a secreted pleotropic protein involved in multiple steps of colorectal cancer progression, was highly upregulated by Slug in vitro, as well as in vivo. We further show that Slug is a direct regulator of osteopontin at the promoter level. The levels of secreted osteopontin were correlated with Slug expression, thereby linking the tumor phenotype to a biomarker available by liquid biopsies. The results also suggest that osteopontin neutralization may attenuate at least some of the Slug-mediated functions.
Collapse
Affiliation(s)
- Katyana Amilca-Seba
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
| | - Tuan Zea Tan
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | | | - Lila Louadj
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
| | - Sandrine Thouroude
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
| | - Anaïs Bouygues
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
| | - Michèle Sabbah
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Annette K. Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Jérôme A. Denis
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75571 Paris, France; (K.A.-S.); (L.L.); (S.T.); (A.B.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Sorbonne Université, 75005 Paris, France
- Department of Endocrinology and Oncology Biochemistry, Pitié-Salpetrière Hospital, 075013 Paris, France
- Correspondence:
| |
Collapse
|
19
|
Yang HC, Zhang HW, Yang J, Liu SW, Zhang SJ. Autocrine osteopontin is involved in maintaining the growth and metastasis of Echinococcus multilocularis. Acta Trop 2022; 228:106328. [PMID: 35085512 DOI: 10.1016/j.actatropica.2022.106328] [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: 06/22/2021] [Revised: 01/04/2022] [Accepted: 01/23/2022] [Indexed: 12/01/2022]
Abstract
Alveolar echinococcosis is a zoonotic disease that seriously endangers human health. This study aims to investigate the effects of osteopontin on the growth and intra- or extra-hepatic metastasis of Echinococcus multilocularis. Mice were randomly divided into untreated (control group, n = 25), PBS (n = 25), Lv3-NC (n = 25), and Lv-OPN-734 (n = 25) groups. Knockdown OPN by injecting lentivirus through the intraperitoneal portal vein, the metastatic lesions infected with Echinococcus multilocularis and adjacent liver tissues were observed, and the expression of osteopontin and epidermal growth factor receptor pathway-related molecules were studied. Gross observation of specimens suggested that there was no extra- hepatic metastasis, and mild intrahepatic invasion was observed in the Lv-OPN-734 group after 4 months of infection, and lung metastasis occurred in the Lv3-NC group. Western-blot and immunohistochemical staining results showed that the protein expression of OPN, phosphorylation of epidermal growth factor receptor and downstream molecules of the pathway decreased significantly after osteopontin knockdown, whereas the levels of non-phosphorylated proteins did not change significantly. In human tissues, through western-blot and immunohistochemical staining we found that compared with the control group, the expression of OPN in the liver tissues infected with Echinococcus multilocularis were higher than that in the control group. These findings indicate that osteopontin is involved in maintaining the growth and metastasis of Echinococcus multilocularis, suggesting that osteopontin may be a potential target for the treatment of alveolar echinococcosis.
Collapse
Affiliation(s)
- Hai-Cheng Yang
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Hong-Wei Zhang
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Jian Yang
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Shi-Wen Liu
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Shi-Jie Zhang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, Xinjiang, China; The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
20
|
Hu W, Zhao Y, Su L, Wu Z, Jiang W, Jiang X, Liu M. Silencing the lncRNA NORAD inhibits EMT of head and neck squamous cell carcinoma stem cells via miR‑26a‑5p. Mol Med Rep 2021; 24:743. [PMID: 34435652 PMCID: PMC8430304 DOI: 10.3892/mmr.2021.12383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells are closely associated with tumor metastasis or recurrence. According to previous literature reports, microRNA (miR)‑26a has an inhibitory effect on head and neck squamous cell carcinoma (HNSCC), and the long non‑coding RNA (lncRNA) non‑coding RNA activated by DNA damage (NORAD) has been found to interact with miR‑26a‑5p. The present study aimed to investigate the regulation and mechanism of NORAD and miR‑26a‑5p in the epithelial‑mesenchymal transition (EMT) of HNSCC stem cells. An ALDEFLUOR stem cell detection kit, a flow cytometer, a self‑renewal ability test and western blotting were used to sort and identify HNSCC stem cells. The ENCORI website and a dual‑luciferase assay were used to assess the relationship between genes. The mRNA and protein expression levels of NORAD, miR‑26a‑5p and EMT‑related genes were detected via reverse transcription‑quantitative PCR and western blotting. Functional experiments (MTT assay, flow cytometry, wound healing assay and Transwell assay) were conducted to analyze the effects of NORAD and miR‑26a‑5p on HNSCC stem cells. The successfully sorted aldehyde dehydrogenase (ALDH)+ cells had a self‑renewal capacity and displayed upregulated expression levels of CD44, Oct‑4 and Nanog. NORAD knockdown, achieved using small interfering (si)RNA, downregulated the expression levels of tumor markers in ALDH+ cells. siNORAD inhibited cell vitality, migration and invasion, as well as promoted apoptosis, increased the expression of epithelial cell markers and decreased the expression of interstitial cell markers in HNSCC stem cells. miR‑26a‑5p was a downstream gene of NORAD, and knockdown of miR‑26a‑5p partially offset the regulatory effect of siNORAD on HNSCC stem cells. Collectively, the present study demonstrated that NORAD knockdown attenuated the migration, invasion and EMT of HNSCC stem cells via miR‑26a‑5p.
Collapse
Affiliation(s)
- Weiming Hu
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yong Zhao
- Department of Otorhinolaryngology, XIXI Hospital of Hangzhou, Hangzhou, Zhejiang 310012, P.R. China
| | - Lizhong Su
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zuliang Wu
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Wenjing Jiang
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaoze Jiang
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Ming Liu
- Department of Otorhinolaryngology, Zhejiang Hospital, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
21
|
Bommi PV, Bowen CM, Reyes-Uribe L, Wu W, Katayama H, Rocha P, Parra ER, Francisco-Cruz A, Ozcan Z, Tosti E, Willis JA, Wu H, Taggart MW, Burks JK, Lynch PM, Edelmann W, Scheet PA, Wistuba II, Sinha KM, Hanash SM, Vilar E. The Transcriptomic Landscape of Mismatch Repair-Deficient Intestinal Stem Cells. Cancer Res 2021; 81:2760-2773. [PMID: 34003775 DOI: 10.1158/0008-5472.can-20-2896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 02/16/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
Lynch syndrome is the most common cause of hereditary colorectal cancer and is secondary to germline alterations in one of four DNA mismatch repair (MMR) genes. Here we aimed to provide novel insights into the initiation of MMR-deficient (MMRd) colorectal carcinogenesis by characterizing the expression profile of MMRd intestinal stem cells (ISC). A tissue-specific MMRd mouse model (Villin-Cre;Msh2 LoxP/LoxP ) was crossed with a reporter mouse (Lgr5-EGFP-IRES-creERT2) to trace and isolate ISCs (Lgr5+) using flow cytometry. Three different ISC genotypes (Msh2-KO, Msh2-HET, and Msh2-WT) were isolated and processed for mRNA-seq and mass spectrometry, followed by bioinformatic analyses to identify expression signatures of complete MMRd and haplo-insufficiency. These findings were validated using qRT-PCR, IHC, and whole transcriptomic sequencing in mouse tissues, organoids, and a cohort of human samples, including normal colorectal mucosa, premalignant lesions, and early-stage colorectal cancers from patients with Lynch syndrome and patients with familial adenomatous polyposis (FAP) as controls. Msh2-KO ISCs clustered together with differentiated intestinal epithelial cells from all genotypes. Gene-set enrichment analysis indicated inhibition of replication, cell-cycle progression, and the Wnt pathway and activation of epithelial signaling and immune reaction. An expression signature derived from MMRd ISCs successfully distinguished MMRd neoplastic lesions of patients with Lynch syndrome from FAP controls. SPP1 was specifically upregulated in MMRd ISCs and colocalized with LGR5 in Lynch syndrome colorectal premalignant lesions and tumors. These results show that expression signatures of MMRd ISC recapitulate the initial steps of Lynch syndrome carcinogenesis and have the potential to unveil novel biomarkers of early cancer initiation. SIGNIFICANCE: The transcriptomic and proteomic profile of MMR-deficient intestinal stem cells displays a unique set of genes with potential roles as biomarkers of cancer initiation and early progression.
Collapse
Affiliation(s)
- Prashant V Bommi
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Charles M Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenhui Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pedro Rocha
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Francisco-Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zuhal Ozcan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jason A Willis
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa W Taggart
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick M Lynch
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Paul A Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krishna M Sinha
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
22
|
Butti R, Kumar TVS, Nimma R, Banerjee P, Kundu IG, Kundu GC. Osteopontin Signaling in Shaping Tumor Microenvironment Conducive to Malignant Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:419-441. [PMID: 34664250 DOI: 10.1007/978-3-030-73119-9_20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Context-dependent reciprocal crosstalk between cancer and surrounding stromal cells in the tumor microenvironment is imperative for the regulation of various hallmarks of cancer. A myriad of growth factors, chemokines, and their receptors aids in the interaction between cancer cells and tumor microenvironmental components. Osteopontin is a chemokine-like protein, overexpressed in different types of cancers. Osteopontin plays a crucial role in orchestrating dialogue between cancer and stromal cells. Osteopontin, in tumor microenvironment, is produced in tumor as well as stromal cells. Tumor-derived osteopontin regulates proliferation, migration, activation, and differentiation of different types of stromal cells. Osteopontin secreted from tumor cells regulates the generation of cancer-associated fibroblasts from resident fibroblasts and mesenchymal stem cells. Osteopontin also shapes immunosuppressive tumor microenvironment by controlling regulatory T cells and tumor-associated macrophages. Moreover, secretion of osteopontin from tumor stroma has been highly documented. Stromal cell-derived osteopontin induces epithelial-to-mesenchymal transition, angiogenesis, metastasis, and cancer stem cell enrichment. Tumor- or stroma-derived osteopontin mainly functions through binding with cell surface receptors, integrins and CD44, and activates downstream signaling events like PI-3 kinase/Akt and MAPK pathways. Presumably, disrupting the communication between the tumor cells and surrounding microenvironment by targeting osteopontin-regulated signaling using specific antibodies, small-molecule inhibitors, and chemotherapeutic agents is a novel therapeutic strategy for clinical management of cancer.
Collapse
Affiliation(s)
- Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Totakura V S Kumar
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Ramakrishna Nimma
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Pinaki Banerjee
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Ipsita G Kundu
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Institute of Eminence, Hyderabad, India
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, India. .,School of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Institute of Eminence, Bhubaneswar, India.
| |
Collapse
|
23
|
Osteopontin: A Key Regulator of Tumor Progression and Immunomodulation. Cancers (Basel) 2020; 12:cancers12113379. [PMID: 33203146 PMCID: PMC7698217 DOI: 10.3390/cancers12113379] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anti-PD-1/PD-L1 and anti-CTLA-4-based immune checkpoint blockade (ICB) immunotherapy have recently emerged as a breakthrough in human cancer treatment. Durable efficacy has been achieved in many types of human cancers. However, not all human cancers respond to current ICB immunotherapy and only a fraction of the responsive cancers exhibit efficacy. Osteopontin (OPN) expression is highly elevated in human cancers and functions as a tumor promoter. Emerging data suggest that OPN may also regulate immune cell function in the tumor microenvironment. This review aims at OPN function in human cancer progression and new findings of OPN as a new immune checkpoint. We propose that OPN compensates PD-L1 function to promote tumor immune evasion, which may underlie human cancer non-response to current ICB immunotherapy. Abstract OPN is a multifunctional phosphoglycoprotein expressed in a wide range of cells, including osteoclasts, osteoblasts, neurons, epithelial cells, T, B, NK, NK T, myeloid, and innate lymphoid cells. OPN plays an important role in diverse biological processes and is implicated in multiple diseases such as cardiovascular, diabetes, kidney, proinflammatory, fibrosis, nephrolithiasis, wound healing, and cancer. In cancer patients, overexpressed OPN is often detected in the tumor microenvironment and elevated serum OPN level is correlated with poor prognosis. Initially identified in activated T cells and termed as early T cell activation gene, OPN links innate cells to adaptive cells in immune response to infection and cancer. Recent single cell RNA sequencing revealed that OPN is primarily expressed in tumor cells and tumor-infiltrating myeloid cells in human cancer patients. Emerging experimental data reveal a key role of OPN is tumor immune evasion through regulating macrophage polarization, recruitment, and inhibition of T cell activation in the tumor microenvironment. Therefore, in addition to its well-established direct tumor cell promotion function, OPN also acts as an immune checkpoint to negatively regulate T cell activation. The OPN protein level is highly elevated in peripheral blood of human cancer patients. OPN blockade immunotherapy with OPN neutralization monoclonal antibodies (mAbs) thus represents an attractive approach in human cancer immunotherapy.
Collapse
|
24
|
Liu Y, Li C, Dong L, Chen X, Fan R. Identification and verification of three key genes associated with survival and prognosis of COAD patients via integrated bioinformatics analysis. Biosci Rep 2020; 40:BSR20200141. [PMID: 32936304 PMCID: PMC7522359 DOI: 10.1042/bsr20200141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 08/21/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most lethal malignancy in the world, wherein colon adenocarcinoma (COAD) is the most prevalent type of CRC. Exploring biomarkers is important for the diagnosis, treatment, and prevention of COAD. METHODS We used GEO2R and Venn online software for differential gene screening analysis. Hub genes were screened via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and Cytoscape, following Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, survival analysis and RNA expression validation were performed via UALCAN online software and real-time PCR. Immunohistochemistry (IHC) was performed to verify the protein expression level of hub genes from tissues of COAD patients. RESULTS In the present study, we screened 323 common differentially expressed genes (DEGs) from four GSE datasets. Furthermore, four hub genes were selected for survival correlation analysis and expression level verification, three of which were shown to be statistically significant. CONCLUSION Our study suggests that Serpin Family E Member 1 (SERPINE1), secreted phosphoprotein 1 (SPP1) and tissue inhibitor of metalloproteinase 1 (TIMP1) may be biomarkers closely related to the prognosis of CRC patients.
Collapse
Affiliation(s)
- Yong Liu
- Department of General Surgery, Tianjin Xiqing Hospital, Tianjin 300380, P.R. China
| | - Chao Li
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
| | - Lijin Dong
- Editorial Department of Education and Research Security Centre, Logistic University of Chinese People’s Armed Police Force, Tianjin 300309, P.R. China
| | - Xuewei Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
| | - Rong Fan
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
- Central Laboratory, Tianjin Xiqing Hospital, Tianjin 300380, P.R. China
| |
Collapse
|
25
|
The Crucial Role of CXCL8 and Its Receptors in Colorectal Liver Metastasis. DISEASE MARKERS 2019; 2019:8023460. [PMID: 31827643 PMCID: PMC6886345 DOI: 10.1155/2019/8023460] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022]
Abstract
CXCL8 (also known as IL-8) can produce different biological effects by binding to its receptors: CXCR1, CXCR2, and the Duffy antigen receptor for chemokines (DARC). CXCL8 and its receptors are associated with the development of various tumor types, especially colorectal cancer and its liver metastases. In addition to promoting angiogenesis, proliferation, invasion, migration, and the survival of colorectal cancer (CRC) cells, CXCL8 and its receptors have also been known to induce the epithelial-mesenchymal transition (EMT) of CRC cells, to help them to escape host immunosurveillance as well as to enhance resistance to anoikis, which promotes the formation of circulating tumor cells (CTCs) and their colonization of distant organs. In this paper, we will review the established roles of CXCL8 signaling in CRC and discuss the possible strategies of targeting CXCL8 signaling for overcoming CRC drug resistance and cancer progression, including direct targeting of CXCL8/CXCR1/2 or indirect targeting through the inhibition of CXCL8-CXCR1/2 signaling.
Collapse
|
26
|
Huang TX, Guan XY, Fu L. Therapeutic targeting of the crosstalk between cancer-associated fibroblasts and cancer stem cells. Am J Cancer Res 2019; 9:1889-1904. [PMID: 31598393 PMCID: PMC6780671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/28/2019] [Indexed: 06/10/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in cancer progression and treatment failure. CAFs display extreme phenotypic heterogeneity and functional diversity. Some subpopulations of CAFs have the ability to reconstitute cancer stemness by promoting the expansion of cancer stem cells (CSCs) or by inducing the generation of CSCs from differentiated cancer cells. CAFs regulate cancer stemness in different types of solid tumors by activating a wide array of CSC-related signaling by secreting proteins and exosomes. As feedback, the CSCs can also induce the proliferation and further activation of CAFs to promote their CSC-supporting activities, thus completing the loop of CAF-CSC crosstalk. Current research on targeting CAF-CSC crosstalk could be classified into (i) specific depletion of CAF subpopulations that have CSC-supporting activities and (ii) targeting molecular signaling in CAF-CSC crosstalk, such as the IL6/STAT3, TGF-β/SDF-1/PI3K, WNT/β-catenin, HGF/cMET and SHH/Hh pathways. Strategies targeting CAF-CSC crosstalk may open new avenues for overcoming cancer progression and therapeutic resistance.
Collapse
Affiliation(s)
- Tu-Xiong Huang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen International Cancer Center, Shenzhen University School of MedicineShenzhen, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong KongHong Kong
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen International Cancer Center, Shenzhen University School of MedicineShenzhen, China
| |
Collapse
|