1
|
Huang G, Wang D, Xue J. The role of DDIT3 in modulating proliferation and tamoxifen resistance in luminal A subtype breast cancer through the DDIT3-IRF1 axis. Biochem Biophys Res Commun 2025; 769:151922. [PMID: 40359764 DOI: 10.1016/j.bbrc.2025.151922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND This study investigates the role of DNA Damage Inducible Transcript 3 (DDIT3) in luminal A subtype breast cancer (LABC). DDIT3, a transcription factor linked to various stress responses, has been implicated in tumorigenesis, yet its specific contributions to LABC biology remain poorly understood. METHODS To elucidate these functions, we utilized bioinformatics analyses, including data from TCGA and Kaplan-Meier databases. Furthermore, we performed siRNA-mediated knockdown and overexpression experiments in MCF-7 and T47D cells to assess DDIT3's functional impact on cell proliferation, drug resistance, etc. RNA sequencing analysis identified differentially expressed genes (DEGs) associated with DDIT3 manipulation, and pinpointing the crucial downstream target with rescue experiment. RESULTS Compared to normal breast tissue, DDIT3 is lowly expressed in LABC, and LABC patients with low DDIT3 expression have a lower survival rate, indicating relatively poor prognosis. Furthermore, DDIT3 negatively regulates the proliferation of LABC cells, also negatively correlated with the sensitivity of TAM. RNA-seq result and rescue experiment identified the interferon regulatory factor 1 (IRF1) as a crucial downstream target of DDIT3 to regulating LABC cell proliferation and tamoxifen (TAM) resistance. CONCLUSIONS DDIT3 is negatively correlated with poor prognosis in LABC patients. And DDIT3 may negatively regulate the proliferation and TAM sensitivity in LABC cells through the DDIT3-IRF1 axis.
Collapse
Affiliation(s)
- Guoqing Huang
- Institute of Advanced Technology, Heilongjiang Academy of Sciences, Harbin, Heilongjiang, China.
| | - Dandan Wang
- Institute of Advanced Technology, Heilongjiang Academy of Sciences, Harbin, Heilongjiang, China
| | - Jiaying Xue
- Institute of Advanced Technology, Heilongjiang Academy of Sciences, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Ding S, Hao Y, Qi Y, Wei H, Zhang J, Li H. Molecular mechanism of tumor-infiltrating immune cells regulating endometrial carcinoma. Genes Dis 2025; 12:101442. [PMID: 40083326 PMCID: PMC11904505 DOI: 10.1016/j.gendis.2024.101442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 07/14/2024] [Accepted: 08/14/2024] [Indexed: 03/16/2025] Open
Abstract
Endometrial carcinoma (EC) is a prevalent gynecological cancer, and its interaction with the immune system is pivotal in cancer progression. This comprehensive review explores the molecular mechanisms involved in the regulation of EC by tumor-infiltrating immune cells. This review discusses the composition and functions of various immune cell types within the tumor microenvironment, including T cells, B cells, macrophages, and natural killer cells, and elucidates their specific roles in cancer control. It also delves into the immune evasion strategies employed by EC cells, with a specific focus on immune checkpoint pathways and their influence on tumor development. Signaling pathways, cytokines, and chemokines mediating immune responses within the tumor microenvironment are also detailed. Furthermore, clinical implications and therapeutic strategies, such as immunotherapies, are also reviewed, and relevant clinical trials are discussed. Additionally, this review discusses the existing gaps in our knowledge, suggests potential avenues for future research, and emphasizes the significance of understanding these mechanisms for enhanced EC treatment. This review provides an exhaustive overview of the current knowledge, supporting the ongoing quest for more effective therapeutic interventions on EC.
Collapse
Affiliation(s)
- Silu Ding
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Yingying Hao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Heng Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Hui Li
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning 117004, China
| |
Collapse
|
3
|
Lian C, Liu Y, Lei P. miR-186-5p Down-Regulates PD-L1 Level in Acute Myeloid Leukemia Cells and Inhibits Tumorigenesis and Immune Escape. J Biochem Mol Toxicol 2025; 39:e70278. [PMID: 40285500 DOI: 10.1002/jbt.70278] [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: 10/30/2024] [Revised: 03/07/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Acute myeloid leukemia (AML) is a malignant tumor of blood cells, which seriously interferes with the generation of normal cells. Although miR-186-5p is diminished in AML, its exact mechanism is not well understood. miR-186-5p and PD-L1 levels in AML cells (HL-60, KG-1, TF-1a, MOLT-3) and subcutaneous tumor tissue were discovered through qRT-PCR and Western blot. miR-186-5 p and PD-L1 combining sites were foreseen by the database and verified by dual luciferase and immunoprecipitation experiments. AML cells with miR-186-5p overexpression or knockdown and PD-L1 overexpression were cocultured with CD4+ and CD8+ T cells. The proliferation, migration, invasion and apoptosis of AML cells, CD8+ and CD4+ T cell growth and apoptosis, and activated markers (Perforin and Granzyme B) and secreted cytokines (IFN-γ, IL-4 and TNF-α) levels were detected by CCK8, Transwell, flow cytometry, CFSE, Western blot and ELISA, respectively. Subcutaneous xenograft magnitude and mass in nude mice were measured. Ki67 level was identified through immunohistochemistry. CD4+ and CD8+ T cell level and infiltration were detected by immunofluorescence and flow cytometry. miR-186-5p was downregulated, and PD-L1 was boosted in AML cells and subcutaneous tumor tissues (p < 0.05), while miR-186-5p targeted down-regulate PD-L1. miR-186-5p upregulation hindered AML cell multiplication, migration, invasion and facilitate cell death, and enhanced the proliferation activity, activation markers (Perforin and Granzyme B) and secreted cytokines (IFN-γ, IL-4, TNF-α) of CD8+ and CD4+ T cells, inhibited apoptosis, and inhibited immune escape (p < 0.05). Knockdown of miR-186-5p can promote AML progression, but PD-L1 upregulation weakens the antitumor impact of miR-186-5p overexpression (p < 0.05). Transplanted tumor mice experiments also found that miR-186-5p hindered PD-L1 and tumor growth (p < 0.05). In conclusion, miR-186-5p can target inhibit PD-L1, suppress AML cells multiplication, movement, invasion and immune escape, and then reduce AML, aiming to provide support and basis for the pathological mechanism and prevention and treatment strategy of AML.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/immunology
- MicroRNAs/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Humans
- Animals
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- B7-H1 Antigen/immunology
- Down-Regulation
- Mice
- Mice, Nude
- Carcinogenesis/genetics
- Carcinogenesis/immunology
- Carcinogenesis/pathology
- HL-60 Cells
- Cell Proliferation
- Tumor Escape
- Apoptosis
- Cell Line, Tumor
- RNA, Neoplasm/genetics
- RNA, Neoplasm/immunology
- RNA, Neoplasm/metabolism
- Female
- Male
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/metabolism
- CD8-Positive T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Cheng Lian
- Department of Hematology, Henan provincial People's Hospital, Zhengzhou, Henan, China
| | - Yanhui Liu
- Department of Hematology, Henan provincial People's Hospital, Zhengzhou, Henan, China
| | - Pingchong Lei
- Department of Hematology, Henan provincial People's Hospital, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Chen Y, Jiang L, Zhang L, Chi H, Wang Q. Immune microenvironment and molecular mechanisms in endometrial cancer: implications for resistance and innovative treatments. Discov Oncol 2025; 16:532. [PMID: 40237942 PMCID: PMC12003227 DOI: 10.1007/s12672-025-02169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
This review provides a systematic overview of the molecular mechanisms of endometrial cancer and its drug resistance, particularly involving the aberrant activation of some key signaling pathways. These molecular mechanisms significantly affect the therapeutic outcome of endometrial cancer by promoting tumor cell proliferation, anti-apoptosis, and drug resistance. The article also analyzes the critical role of the immune microenvironment in cancer drug resistance, focusing on the impact of immune cells, immune checkpoints, and hypoxic metabolic reprogramming on anticancer therapies. In recent years, immunotherapy and individualized therapy have shown promising clinical outcomes, especially in advanced endometrial cancer. This article summarizes recent advances in related therapeutic strategies and proposes emerging therapeutic strategies by targeting key pathways and modulating the immune microenvironment to overcome drug resistance and improve patient prognosis.
Collapse
Affiliation(s)
- Yijia Chen
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lanyue Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China.
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
5
|
Zhai F, Li Y, Zheng J, Yan C, Wang S, Yang W, Jin J, Luo X, Zhan Z, Shi J, Wang S, Lin Y, Kong L, Ge Y, Wang H, Ye M, Jin X. SPOP/NOLC1/B4GALT1 signaling axis enhances paclitaxel resistance in endometrial cancer by inducing O-dysglycosylation. Oncogene 2025:10.1038/s41388-025-03347-7. [PMID: 40097806 DOI: 10.1038/s41388-025-03347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/18/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
The effective treatment of paclitaxel-resistant patients remains a major challenge. We found that nucleolar and coiled body phosphoprotein 1 (NOLC1) was highly expressed in the paclitaxel-resistant endometrial cancer (ECa) cells and pathological tissue of ECa patients, which could promote the occurrence and progression of ECa cells. Mechanistically, we confirmed that the E3 ubiquitin ligase substrate-binding adaptor SPOP mediates the ubiquitination and degradation of NOLC1, thereby maintaining normal protein levels. However, ECa-associated SPOP mutants abrogated the binding and ubiquitination of NOLC1, resulting in the accumulation of NOLC1, and ultimately promoting the proliferation, migration, and invasion of ECa cells. In addition, we demonstrated that NOLC1 could act as a transcriptional factor to activate the transcriptional expression of B4GALT1, ultimately leading to abnormal glycosylation metabolism. Moreover, knockdown of B4GALT1 can partly counteract the cancer-promoting effect caused by the overexpression of NOLC1 in vitro and in vivo. Based on these findings, an O-glycosylation inhibitor combined with paclitaxel could effectively improve the sensitivity of paclitaxel-resistant cells. In summary, we found that SPOP can negatively regulate the NOLC1-B4GALT1 signaling axis in ECa, whereas ECa-associated SPOP mutants lead to abnormal activation of this signaling axis, leading to glycosylation metabolism disorders. In addition, paclitaxel combined with B4GALT1-KD or glycosylation inhibitors can significantly inhibit the growth of paclitaxel-resistant endometrial cancer cells.
Collapse
Affiliation(s)
- Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China
| | - Yuxuan Li
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Jingfei Zheng
- Department of Obstetrics and Gynecology, Yinzhou Renmin Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Chunhong Yan
- Department of Obstetrics and Gynecology, Yinzhou Renmin Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Shuyan Wang
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Weili Yang
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Jiabei Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Xia Luo
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Ziqing Zhan
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China
| | - Jiaxin Shi
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Siyuan Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Yan Lin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Lili Kong
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Yidong Ge
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Haoyun Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China.
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
6
|
Ong JY, Abdusamad M, Ramirez I, Gholkar A, Zhang X, Gimeno TV, Torres JZ. Cul3 substrate adaptor SPOP targets Nup153 for degradation. Mol Biol Cell 2025; 36:ar24. [PMID: 39785820 PMCID: PMC11974958 DOI: 10.1091/mbc.e24-04-0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
SPOP is a Cul3 substrate adaptor responsible for the degradation of many proteins related to cell growth and proliferation. Because mutation or misregulation of SPOP drives cancer progression, understanding the suite of SPOP substrates is important to understanding the regulation of cell proliferation. Here, we identify Nup153, a component of the nuclear basket of the nuclear pore complex, as a novel substrate of SPOP. SPOP and Nup153 bind to each other and colocalize at the nuclear envelope and some nuclear foci in cells. The binding interaction between SPOP and Nup153 is complex and multivalent. Nup153 is ubiquitylated and degraded upon expression of SPOPWT but not its substrate binding-deficient mutant SPOPF102C. Depletion of SPOP via RNAi leads to Nup153 stabilization. Upon loss of SPOP activity, the nuclear envelope localization of spindle assembly checkpoint protein Mad1, which is tethered to the nuclear envelope by Nup153, is stronger. Altogether, our results demonstrate that SPOP regulates Nup153 levels and expands our understanding of the role of SPOP in protein and cellular homeostasis.
Collapse
Affiliation(s)
- Joseph Y. Ong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Mai Abdusamad
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Ivan Ramirez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Ankur Gholkar
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Xiaoxuan Zhang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Thomas V. Gimeno
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
| | - Jorge Z. Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
7
|
Ma C, He Y, Wang J, Zhang J, Hou X, Wang S, Chen L, Shu L. Expression levels of STAT3, and protein levels of IL‑6 and sPD‑L1 in different pathological characteristics of endometrial adenocarcinomas. Oncol Lett 2025; 29:156. [PMID: 39911150 PMCID: PMC11795251 DOI: 10.3892/ol.2025.14901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/13/2024] [Indexed: 02/07/2025] Open
Abstract
Endometrial cancer is a common type of cancer in women, with endometrial adenocarcinoma (EA) being the most common type. Monitoring the expression levels of signal transducer and activator of transcription 3 (STAT3), the protein levels of interleukin 6 (IL-6) and soluble programmed death ligand 1 (sPD-L1), and their differences in patients with various pathological characteristics is beneficial for accurately evaluating the disease stage and differentiation degree of patients in clinical practice. The aim of the present study was to assess the expression levels of STAT3, and the protein levels of IL-6 and sPD-L1 in EA. In the present retrospective study, data were retrieved from the medical records of 137 patients with EA who received surgical treatment at The First Affiliated Hospital of Hebei North University from January 2017 to December 2022. Of the 137 cases, 90 met the inclusion criteria. The patients with EA were matched with a cohort of 30 patients with atypical endometrial hyperplasia in a ratio of 3:1. Among the 90 patients with EA, 30 patients with well-differentiated EA were matched with 30 patients with moderately differentiated EA and 30 patients with poorly differentiated EA in a 1:1:1 ratio. Expression level of STAT3, and protein levels of IL-6 and sPD-L1 were recorded preoperatively and compared between patients with different pathological characteristics [such as differentiation degree, disease stage, depth of myometrial invasion and lymph node metastasis (LNM)] and prognosis. Levels of IL-6, STAT3 and sPD-L1 in the observation group were significantly higher compared with the control group (P<0.001). Additionally, there were significant differences in IL-6, STAT3 and sPD-L1 levels between patients with different differentiation degrees, disease stages, myometrial invasion and LNM (P<0.001). The increase in IL-6, STAT3 and sPD-L1 levels were significantly associated with the decrease in the differentiation degree and the increase in the disease stage, depth of myometrial invasion and LNM (P<0.001). IL-6, STAT3 and sPD-L1 levels in patients with a poor prognosis were significantly higher compared with patients with good prognoses (P<0.001). Overall, the expression levels of STAT3, and the protein levels of IL-6 and sPD-L1 were increased in patients with EA compared with in those without EA, and their increase is associated with the pathological characteristics of the disease. The levels of these indices may be detected in clinical practices to evaluate the disease and predict the prognosis.
Collapse
Affiliation(s)
- Chunxing Ma
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Ying He
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Jing Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Juan Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiaoxue Hou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Sisi Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Lihua Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Lisha Shu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
8
|
Cheng B, Li H, Peng X, Chen J, Shao C, Kong Z. Recent advances in developing targeted protein degraders. Eur J Med Chem 2025; 284:117212. [PMID: 39736199 DOI: 10.1016/j.ejmech.2024.117212] [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: 10/22/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Targeted protein degradation (TPD) represents a promising therapeutic approach, encompassing several innovative strategies, including but not limited to proteolysis targeting chimeras (PROTACs), molecular glues, hydrophobic tag tethering degraders (HyTTD), and lysosome-targeted chimeras (LYTACs). Central to TPD are small molecule ligands, which play a critical role in mediating the degradation of target proteins. This review summarizes the current landscape of small molecule ligands for TPD molecules. These small molecule ligands can utilize the proteasome, lysosome, autophagy, or hydrophobic-tagging system to achieve the degradation of target proteins. The article mainly focuses on introducing their design principles, application advantages, and potential limitations. A brief discussion on the development prospects and future directions of TPD technology was also provided.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, 435003, China; Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Hongqiao Li
- The Central Hospital of Huangshi, Huangshi, 435000, China
| | - Xiaopeng Peng
- College of Pharmacy, Gannan Medical University, Ganzhou, 314000, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chuxiao Shao
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| | - Zhihua Kong
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, FoShan, 528200, China.
| |
Collapse
|
9
|
Cui Y, Liu J, Wang X, Wu Y, Chang Y, Hu X, Zhao W. Baicalin attenuates the immune escape of oral squamous cell carcinoma by reducing lactate accumulation in tumor microenvironment. J Adv Res 2025:S2090-1232(25)00040-2. [PMID: 39814222 DOI: 10.1016/j.jare.2025.01.021] [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: 11/11/2024] [Revised: 01/01/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025] Open
Abstract
INTRODUCTION The acidic microenvironment caused by excessive lactate accumulation could inhibit immune lymphocytes antitumor activity and promote the immune escape of tumor cells. Baicalin is an active flavonoid isolated from Scutellaria baicalensis Georgi, a traditional Chinese medicinal herb with antioxidant and anti-inflammatory properties. OBJECTIVES The present study aims to investigate whether and how baicalin inhibits oral squamous cell carcinoma (OSCC) acidic microenvironment and attenuates immune escape. METHODS Baicalin was dose-dependently administrated to OSCC cells (0-50 μmol/L). Co-culture system was constructed by OSCC cells and activated PBMCs. The proliferation and migration of OSCC cells were tested by CCK-8, colony formation, EdU, transwell assays. The cytokines were tested by ELISA kits. Mechanistical exploration was verified by RNA immunoprecipitation (RIP), fluorescence in situ hybridization (FISH) and RNA stability assays. RESULTS Results indicated that baicalin dose-dependently repressed the proliferation and migration of OSCC cells, and strengthened the antitumor immune activity of activated PBMCs to OSCC cells. Moreover, baicalin repressed the lactate accumulation, acidification and m6A modification level of OSCC cells. Molecular docking and MeRIP-Seq revealed that baicalin targeted LDHA via m6A-IGF2BP3-dependent manner to reduce lactate accumulation and PD-L1 expression in co-culture microenvironment. CONCLUSION This study revealed the anti-tumor activity of baicalin for OSCC by reducing lactate accumulation and attenuating the immune escape in tumor microenvironment, which provided a novel insight to improve our understanding in the treatment of traditional Chinese medicine on human cancer.
Collapse
Affiliation(s)
- Yameng Cui
- Department of Integrative Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, No. 1 Huanhu West Road, Tianjin 300060, China
| | - Jingwen Liu
- Department of General/Emergency, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Xi Wang
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yulin Wu
- Intensive Care Unit, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, No. 1 Huanhu West Road, Tianjin 300060, China
| | - Yunhan Chang
- Department of General/Emergency, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Xin Hu
- Department of General/Emergency, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Wei Zhao
- Department of General/Emergency, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|
10
|
Awan AB, Osman MJA, Khan OM. Ubiquitination Enzymes in Cancer, Cancer Immune Evasion, and Potential Therapeutic Opportunities. Cells 2025; 14:69. [PMID: 39851497 PMCID: PMC11763706 DOI: 10.3390/cells14020069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
Ubiquitination is cells' second most abundant posttranslational protein modification after phosphorylation. The ubiquitin-proteasome system (UPS) is critical in maintaining essential life processes such as cell cycle control, DNA damage repair, and apoptosis. Mutations in ubiquitination pathway genes are strongly linked to the development and spread of multiple cancers since several of the UPS family members possess oncogenic or tumor suppressor activities. This comprehensive review delves into understanding the ubiquitin code, shedding light on its role in cancer cell biology and immune evasion. Furthermore, we highlighted recent advances in the field for targeting the UPS pathway members for effective therapeutic intervention against human cancers. We also discussed the recent update on small-molecule inhibitors and PROTACs and their progress in preclinical and clinical trials.
Collapse
Affiliation(s)
- Aiman B. Awan
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar; (A.B.A.); (M.J.A.O.)
| | - Maryiam Jama Ali Osman
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar; (A.B.A.); (M.J.A.O.)
- Research Branch, Sidra Medicine, Doha P.O. Box 34110, Qatar
| | - Omar M. Khan
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar; (A.B.A.); (M.J.A.O.)
| |
Collapse
|
11
|
Sun W, Cao K, Wang S, Lu M, Ma J, Wu C, Zhao Y. Pan-cancer analysis of IRF1 focusing on prognostic and immunological roles in non-small cell lung cancer. Heliyon 2024; 10:e39861. [PMID: 39605834 PMCID: PMC11600070 DOI: 10.1016/j.heliyon.2024.e39861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Interferon regulatory factor 1 (IRF1) significantly affects tumour occurrence and development. This study aimed to analyse its function as a pan-cancer prognostic indicator. We compared IRF1 expression and prognostic significance in normal and tumour samples from different databases. Accordingly, we performed in vitro experiments and immunohistochemistry (IHC) to investigate the role of IRF1 in non-small cell lung cancer (NSCLC). Our findings indicate that IRF1 expression is significantly correlated with prognosis, the tumour microenvironment, and immune cell infiltration. Furthermore, receiver operating characteristic (ROC) analysis revealed that IRF1 had high accuracy in distinguishing cancerous tissues from normal ones. Notably, IRF1 expression was linked to immune-related and immune checkpoint genes. Cell proliferation, invasion, and migration were significantly related to IRF1 expression. IHC indicated that IRF1 was downregulated in NSCLC tissues. Our study provides comprehensive bioinformatic analysis and experimental verification of IRF1, suggesting its potential as a prognostic biomarker in cancer.
Collapse
Affiliation(s)
- Weiling Sun
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
- Department of Endoscope, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Kui Cao
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Siran Wang
- Department of Preventive Dentistry, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, 510182, Guangzhou, China
| | - Mengdi Lu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Chunlong Wu
- Department of Endoscope, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yanbin Zhao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| |
Collapse
|
12
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Roozitalab G, Abedi B, Imani S, Farghadani R, Jabbarzadeh Kaboli P. Comprehensive assessment of TECENTRIQ® and OPDIVO®: analyzing immunotherapy indications withdrawn in triple-negative breast cancer and hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:889-918. [PMID: 38409546 DOI: 10.1007/s10555-024-10174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Atezolizumab (TECENTRIQ®) and nivolumab (OPDIVO®) are both immunotherapeutic indications targeting programmed cell death 1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1), respectively. These inhibitors hold promise as therapies for triple-negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) and have demonstrated encouraging results in reducing the progression and spread of tumors. However, due to their adverse effects and low response rates, the US Food and Drug Administration (FDA) has withdrawn the approval of atezolizumab in TNBC and nivolumab in HCC treatment. The withdrawals of atezolizumab and nivolumab have raised concerns regarding their effectiveness and the ability to predict treatment responses. Therefore, the current study aims to investigate the immunotherapy withdrawal of PD-1/PD-L1 inhibitors, specifically atezolizumab for TNBC and nivolumab for HCC. This study will examine both the structural and clinical aspects. This review provides detailed insights into the structure of the PD-1 receptor and its ligands, the interactions between PD-1 and PD-L1, and their interactions with the withdrawn antibodies (atezolizumab and nivolumab) as well as PD-1 and PD-L1 modifications. In addition, this review further assesses these antibodies in the context of TNBC and HCC. It seeks to elucidate the factors that contribute to diverse responses to PD-1/PD-L1 therapy in different types of cancer and propose approaches for predicting responses, mitigating the potential risks linked to therapy withdrawals, and optimizing patient outcomes. By better understanding the mechanisms underlying responses to PD-1/PD-L1 therapy and developing strategies to predict these responses, it is possible to create more efficient treatments for TNBC and HCC.
Collapse
Affiliation(s)
- Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
14
|
Hong Z, Liu F, Zhang Z. Ubiquitin modification in the regulation of tumor immunotherapy resistance mechanisms and potential therapeutic targets. Exp Hematol Oncol 2024; 13:91. [PMID: 39223632 PMCID: PMC11367865 DOI: 10.1186/s40164-024-00552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Although immune checkpoint-based cancer immunotherapy has shown significant efficacy in various cancers, resistance still limits its therapeutic effects. Ubiquitination modification is a mechanism that adds different types of ubiquitin chains to proteins, mediating protein degradation or altering their function, thereby affecting cellular signal transduction. Increasing evidence suggests that ubiquitination modification plays a crucial role in regulating the mechanisms of resistance to cancer immunotherapy. Drugs targeting ubiquitination modification pathways have been shown to inhibit tumor progression or enhance the efficacy of cancer immunotherapy. This review elaborates on the mechanisms by which tumor cells, immune cells, and the tumor microenvironment mediate resistance to cancer immunotherapy and the details of how ubiquitination modification regulates these mechanisms, providing a foundation for enhancing the efficacy of cancer immunotherapy by intervening in ubiquitination modification.
Collapse
Affiliation(s)
- Zihang Hong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
15
|
Lv Q, Su T, Liu W, Wang L, Hu J, Cheng Y, Ning C, Shan W, Luo X, Chen X. Low Serum Apolipoprotein A1 Levels Impair Antitumor Immunity of CD8+ T Cells via the HIF-1α-Glycolysis Pathway. Cancer Immunol Res 2024; 12:1058-1073. [PMID: 38752667 DOI: 10.1158/2326-6066.cir-23-0506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 08/02/2024]
Abstract
An immunosuppressive microenvironment promotes the occurrence and development of tumors. Low apolipoprotein A1 (ApoA1) is closely related to tumor development, but the underlying mechanisms are unclear. This study investigated the association between serum ApoA1 levels and the immune microenvironment in endometrial, ovarian, and lung cancers. The serum ApoA1 level was decreased significantly in patients with endometrial and ovarian cancers compared with healthy controls. In endometrial cancer (EC) tissues, the low serum ApoA1 level group showed increased CD163+ macrophage infiltration and decreased CD8+ T-cell infiltration compared with the normal serum ApoA1 group. Compromised tumor-infiltrating CD8+ T-cell functions and decreased CD8+ T-cell infiltration also were found in tumor-bearing Apo1-knockout mice. CD8+ T-cell depletion experiments confirmed that ApoA1 exerted its antitumor activity in a CD8+ T-cell-dependent manner. In vitro experiments showed that the ApoA1 mimetic peptide L-4F directly potentiated the antitumor activity of CD8+ T cells via a HIF-1α-mediated glycolysis pathway. Mechanistically, ApoA1 suppressed ubiquitin-mediated degradation of HIF-1α protein by downregulating HIF-1α subunit α inhibitor. This regulatory process maintained the stability of HIF-1α protein and activated the HIF-1α signaling pathway. Tumor-bearing Apoa1 transgenic mice showed an increased response to anti-PD-1 therapy, leading to reduced tumor growth along with increased infiltration of activated CD8+ T cells and enhanced tumor necrosis. The data reported herein demonstrate critical roles for ApoA1 in enhancing CD8+ T-cell immune functions via HIF-1α-mediated glycolysis and support clinical investigation of combining ApoA1 supplementation with anti-PD-1 therapy for treating cancer.
Collapse
Affiliation(s)
- Qiaoying Lv
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Tong Su
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Wei Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Lulu Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Jiali Hu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Yali Cheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Chengcheng Ning
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Weiwei Shan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Xuezhen Luo
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, PR China
| |
Collapse
|
16
|
Yuan J, Lin M, Yang S, Yin H, Ouyang S, Xie H, Tang H, Ou X, Zeng Z. The therapeutic effect and targets of herba Sarcandrae on breast cancer and the construction of a prognostic signature consisting of inflammation-related genes. Heliyon 2024; 10:e31137. [PMID: 38778969 PMCID: PMC11109893 DOI: 10.1016/j.heliyon.2024.e31137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Background The prevalence of breast cancer (BRCA), which is common among women, is on the rise. This study applied network pharmacology to explore the potential mechanism of action of herba sarcandrae in BRCA and construct a prognostic signature composed of inflammation-related genes. Methods The active ingredients of herba sarcandrae were screened using the SymMap, TCMID, and TCMSP platforms, and the molecular targets were determined in the UniProt database. The "drug-active compound-potential target" network was established with Cytoscape 3.7.2. The molecular targets were subjected to disease ontology, gene ontology (GO), and Kyoto Encyclopedia of Genes (KEGG) analyses. AutoDock software was used for molecular docking. Differentially expressed genes (DEGs) related to inflammation were obtained from the BRCA Cancer Genome Atlas (TCGA) database. In the training cohort, the univariate Cox regression model was applied to preliminarily screen prognostic genes. A multigene signature was built by the least absolute shrinkage and selection operator (LASSO) regression model, followed by validation through Kaplan‒Meier, Cox, and receiver operating characteristic (ROC) analyses. Results Forty-one active compounds were identified, and 265 therapeutic targets for herba sarcandrae were predicted. GO enrichment results revealed significant enrichment of biological processes, such as response to xenobiotic stimuli, response to nutrient levels, and response to lipopolysaccharide. KEGG analysis revealed significant enrichment of pathways such as AGE-RAGE and chemical carcinogenesis receptor activation signaling pathways. In addition, the herbs Marc-Andre and rutin were shown to mediate BRCA cell proliferation and apoptosis via the interferon regulatory factor 1 (IRF1)/signal transducer and activator of transcription 3 (STAT3)/programmed death-ligand 1 (PD-L1) pathway. Sixteen inflammatory signatures, including BST2, GPR132, IL12B, IL18, IL1R1, IL2RB, IRF1, and others, were constructed, and the risk score was found to be a strong independent prognostic factor for overall survival in BRCA patients. The 16-inflammation signature was associated with several clinical features (age, clinical stage, T, and N classifications) and could reflect immune cell infiltration in tumor microenvironments with different immune cells. Conclusions Herba sarcandrae and rutin were shown to mediate BRCA cell proliferation and apoptosis via the IRF1/STAT3/PD-L1 pathway, and the 16-member inflammatory signature might be a novel biomarker for predicting BRCA patient prognosis, providing more accurate guidance for clinical treatment prognosis evaluation and having important reference value for individualized treatment selection.
Collapse
Affiliation(s)
- Jie Yuan
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Minxia Lin
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Shaohua Yang
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hao Yin
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Shaoyong Ouyang
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hong Xie
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hongmei Tang
- Pharmaceutical Department, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowei Ou
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Zhiqiang Zeng
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| |
Collapse
|
17
|
Shi H, Chen L, Wang T, Zhang W, Liu J, Huang Y, Li J, Qi H, Wu Z, Wang Y, Chen H, Zhu Y, Li Q. Nur77-IRF1 axis inhibits esophageal squamous cell carcinoma growth and improves anti-PD-1 treatment efficacy. Cell Death Discov 2024; 10:254. [PMID: 38789431 PMCID: PMC11126585 DOI: 10.1038/s41420-024-02019-x] [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: 01/28/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The nuclear receptor Nur77 plays paradoxical roles in numerous cancers. However, whether Nur77 inhibits esophageal squamous cell carcinoma (ESCC) growth and affects immunological responses against ESCC has not been determined. The functional role of Nur77 in ESCC was investigated in this study using human ESCC cell lines, quantitative real-time polymerase chain reaction (PCR), cell proliferation and colony formation assays, flow cytometry analysis, western blotting and animal models. The target gene controlled by Nur77 was verified using dual-luciferase reporter assays, chromatin immunoprecipitation analysis and functional rescue experiments. To examine the clinical importance of Nur77, 72 human primary ESCC tissues were subjected to immunohistochemistry. Taken together, these findings showed that, both in vitro and in vivo, Nur77 dramatically reduced ESCC cell growth and triggered apoptosis. Nur77 directly interacts with the interferon regulatory factor 1 (IRF1) promoter to inhibit its activity in ESCC. Pharmacological induction of Nur77 using cytosporone B (CsnB) inhibited ESCC cell proliferation and promoted apoptosis both in vitro and in vivo. Furthermore, CsnB increased CD8+ T-cell infiltration and cytotoxicity to inhibit the formation of ESCC tumors in an immunocompetent mouse model. In ESCC tissues, Nur77 expression was downregulated, and IRF1 expression was increased; moreover, their expression levels were negatively related. IRF1 and Nur77 were strongly correlated with overall survival. These findings suggested that Nur77 targets and regulates the IRF1/PD-L1 axis to serve as a tumor suppressor in ESCC. Graphical abstract of the regulatory mechanism of Nur77 overexpression downregulates IRF1 in the inhibition of ESCC progression and enhance anti-PD-1 therapy efficacy.
Collapse
Affiliation(s)
- Huanying Shi
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Jiafeng Liu
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Yuxin Huang
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Jiyifan Li
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Zimei Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Yi Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China
| | - Haifei Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China.
| | - Yongjun Zhu
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, No.12 Urumqi Middle Road, Shanghai, 200040, China.
| |
Collapse
|
18
|
Li X, Wu M, Lu J, Yu J, Chen D. Interleukin-21 as an adjuvant in cancer immunotherapy: Current advances and future directions. Biochim Biophys Acta Rev Cancer 2024; 1879:189084. [PMID: 38354828 DOI: 10.1016/j.bbcan.2024.189084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/21/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Immunotherapy has revolutionized cancer treatment. However, it's well-recognized that a considerable proportion of patients fail to benefit from immunotherapy, and to improve immunotherapy response is clinically urgent. Insufficient immune infiltration and immunosuppressive tumor microenvironments (TME) are main contributors to immunotherapy resistance. Thus sustaining functional self-renewal capacity for immune cells and subverting immune-suppressive signals are potential strategies for boosting the efficacy of immunotherapy. Interleukin-21 (IL-21), a crucial cytokine, which could enhance cytotoxic function of immune cells and reduces immunosuppressive cells enrichment in TME, shows promising orientations as an immunoadjuvant in tumor immunotherapy. This review focuses on IL-21 in cancer treatment, including function and mechanisms of IL-21, preclinical and clinical studies, and future directions for IL-21-assisted therapies.
Collapse
Affiliation(s)
- Xinyang Li
- School of Clinical Medicine, Weifang Medical University, Weifang, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Lu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- School of Clinical Medicine, Weifang Medical University, Weifang, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Dawei Chen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
19
|
Deng Y, Ding W, Ma K, Zhan M, Sun L, Zhou Z, Lu L. SPOP point mutations regulate substrate preference and affect its function. Cell Death Dis 2024; 15:172. [PMID: 38409107 PMCID: PMC10897488 DOI: 10.1038/s41419-024-06565-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
The adaptor SPOP recruits substrates to CUL3 E3 ligase for ubiquitination and degradation. Structurally, SPOP harbors a MATH domain for substrate recognition, and a BTB domain responsible for binding CUL3. Reported point mutations always occur in SPOP's MATH domain and are through to disrupt affinities of SPOP to substrates, thereby leading to tumorigenesis. In this study, we identify the tumor suppressor IRF2BP2 as a novel substrate of SPOP. SPOP enables to attenuate IRF2BP2-inhibited cell proliferation and metastasis in HCC cells. However, overexpression of wild-type SPOP alone suppresses HCC cell proliferation and metastasis. In addition, a HCC-derived mutant, SPOP-M35L, shows an increased affinity to IRF2BP2 in comparison with wild-type SPOP. SPOP-M35L promotes HCC cell proliferation and metastasis, suggesting that M35L mutation possibly reprograms SPOP from a tumor suppressor to an oncoprotein. Taken together, this study uncovers mutations in SPOP's MATH lead to distinct functional consequences in context-dependent manners, rather than simply disrupting its interactions with substrates, raising a noteworthy concern that we should be prudent to select SPOP as therapeutic target for cancers.
Collapse
Affiliation(s)
- Yanran Deng
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 210009, Nanjing, China
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, 330022, Nanchang, China
| | - Wenhao Ding
- College of Life Sciences, Shandong Agricultural University, 271018, Tai'an, China
| | - Kaize Ma
- College of Life Sciences, Shandong Agricultural University, 271018, Tai'an, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), 519000, Zhuhai, Guangdong, China
| | - Li Sun
- Jiangsu Key laboratory of Drug Screening, China Pharmaceutical University, 210009, Nanjing, China.
| | - Zizhang Zhou
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, 330022, Nanchang, China.
- College of Life Sciences, Shandong Agricultural University, 271018, Tai'an, China.
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), 519000, Zhuhai, Guangdong, China.
| |
Collapse
|
20
|
Perevalova AM, Gulyaeva LF, Pustylnyak VO. Roles of Interferon Regulatory Factor 1 in Tumor Progression and Regression: Two Sides of a Coin. Int J Mol Sci 2024; 25:2153. [PMID: 38396830 PMCID: PMC10889282 DOI: 10.3390/ijms25042153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
IRF1 is a transcription factor well known for its role in IFN signaling. Although IRF1 was initially identified for its involvement in inflammatory processes, there is now evidence that it provides a function in carcinogenesis as well. IRF1 has been shown to affect several important antitumor mechanisms, such as induction of apoptosis, cell cycle arrest, remodeling of tumor immune microenvironment, suppression of telomerase activity, suppression of angiogenesis and others. Nevertheless, the opposite effects of IRF1 on tumor growth have also been demonstrated. In particular, the "immune checkpoint" molecule PD-L1, which is responsible for tumor immune evasion, has IRF1 as a major transcriptional regulator. These and several other properties of IRF1, including its proposed association with response and resistance to immunotherapy and several chemotherapeutic drugs, make it a promising object for further research. Numerous mechanisms of IRF1 regulation in cancer have been identified, including genetic, epigenetic, transcriptional, post-transcriptional, and post-translational mechanisms, although their significance for tumor progression remains to be explored. This review will focus on the established tumor-suppressive and tumor-promoting functions of IRF1, as well as the molecular mechanisms of IRF1 regulation identified in various cancers.
Collapse
Affiliation(s)
- Alina M. Perevalova
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| | - Lyudmila F. Gulyaeva
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| | - Vladimir O. Pustylnyak
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| |
Collapse
|
21
|
Yu Z, Wu X, Zhu J, Yan H, Li Y, Zhang H, Zhong Y, Lin M, Ye G, Li X, Jin J, Li K, Wang J, Zhuang H, Lin T, He J, Lu C, Xu Z, Zhang X, Li H, Jin X. BCLAF1 binds SPOP to stabilize PD-L1 and promotes the development and immune escape of hepatocellular carcinoma. Cell Mol Life Sci 2024; 81:82. [PMID: 38340178 PMCID: PMC10858942 DOI: 10.1007/s00018-024-05144-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/21/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Interaction between programmed death-1 (PD-1) ligand 1 (PD-L1) on tumor cells and PD-1 on T cells allows tumor cells to evade T cell-mediated immune surveillance. Strategies targeting PD-1/PD-L1 have shown clinical benefits in a variety of cancers. However, limited response rates in hepatocellular carcinoma (HCC) have prompted us to investigate the molecular regulation of PD-L1. Here, we identify B cell lymphoma-2-associated transcription factor 1 (BCLAF1) as a key PD-L1 regulator in HCC. Specifically, BCLAF1 interacts with SPOP, an E3 ligase that mediates the ubiquitination and degradation of PD-L1, thereby competitively inhibiting SPOP-PD-L1 interaction and subsequent ubiquitination and degradation of PD-L1. Furthermore, we determined an SPOP-binding consensus (SBC) motif mediating the BCLAF1-SPOP interaction on BCLAF1 protein and mutation of BCLAF1-SBC motif disrupts the regulation of the SPOP-PD-L1 axis. In addition, BCLAF1 expression was positively correlated with PD-L1 expression and negatively correlated with biomarkers of T cell activation, including CD3 and CD8, as well as with the level of immune cell infiltration in HCC tissues. Besides, BCLAF1 depletion leads to a significant reduction of PD-L1 expression in vitro, and this reduction of PD-L1 promoted T cell-mediated cytotoxicity. Notably, overexpression of BCLAF1 sensitized tumor cells to checkpoint therapy in an in vitro HCC cells-Jurkat cells co-culture model, whereas BCLAF1-SBC mutant decreased tumor cell sensitivity to checkpoint therapy, suggesting that BCLAF1 and its SBC motif serve as a novel therapeutic target for enhancing anti-tumor immunity in HCC.
Collapse
Affiliation(s)
- Zongdong Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Xiang Wu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
| | - Huan Yan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
| | - Yuxuan Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Hui Zhang
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Yeling Zhong
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Man Lin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Ganghui Ye
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Xinming Li
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Jiabei Jin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Kailang Li
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Hui Zhuang
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Ting Lin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Jian He
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China
| | - Changjiang Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
| | - Zeping Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
| | - Xie Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China
| | - Hong Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Li Huili Hospital, Ningbo University, Ningbo, 315040, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, China.
| |
Collapse
|
22
|
Cecchi D, Jackson N, Beckham W, Chithrani DB. Improving the Efficacy of Common Cancer Treatments via Targeted Therapeutics towards the Tumour and Its Microenvironment. Pharmaceutics 2024; 16:175. [PMID: 38399237 PMCID: PMC10891984 DOI: 10.3390/pharmaceutics16020175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer is defined as the uncontrolled proliferation of heterogeneous cell cultures in the body that develop abnormalities and mutations, leading to their resistance to many forms of treatment. Left untreated, these abnormal cell growths can lead to detrimental and even fatal complications for patients. Radiation therapy is involved in around 50% of cancer treatment workflows; however, it presents significant recurrence rates and normal tissue toxicity, given the inevitable deposition of the dose to the surrounding healthy tissue. Chemotherapy is another treatment modality with excessive normal tissue toxicity that significantly affects patients' quality of life. To improve the therapeutic efficacy of radiotherapy and chemotherapy, multiple conjunctive modalities have been proposed, which include the targeting of components of the tumour microenvironment inhibiting tumour spread and anti-therapeutic pathways, increasing the oxygen content within the tumour to revert the hypoxic nature of the malignancy, improving the local dose deposition with metal nanoparticles, and the restriction of the cell cycle within radiosensitive phases. The tumour microenvironment is largely responsible for inhibiting nanoparticle capture within the tumour itself and improving resistance to various forms of cancer therapy. In this review, we discuss the current literature surrounding the administration of molecular and nanoparticle therapeutics, their pharmacokinetics, and contrasting mechanisms of action. The review aims to demonstrate the advancements in the field of conjugated nanomaterials and radiotherapeutics targeting, inhibiting, or bypassing the tumour microenvironment to promote further research that can improve treatment outcomes and toxicity rates.
Collapse
Affiliation(s)
- Daniel Cecchi
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
| | - Nolan Jackson
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
| | - Wayne Beckham
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
- British Columbia Cancer-Victoria, Victoria, BC V8R 6V5, Canada
| | - Devika B. Chithrani
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
- Centre for Advanced Materials and Related Technologies, Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Department of Computer Science, Mathematics, Physics and Statistics, Okanagan Campus, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
23
|
Gan S, Qu F, Zhang X, Pan X, Xu D, Cui X, Hou J. LRP5 competes for SPOP binding to enhance tumorigenesis mediated by Daxx and PD-L1 in prostate cancer. Exp Cell Res 2024; 434:113857. [PMID: 38008278 DOI: 10.1016/j.yexcr.2023.113857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 11/28/2023]
Abstract
Genetic factors coordinate with environmental factors to drive the pathogenesis of prostate adenocarcinoma (PRAD). SPOP is one of the most mutated genes and LRP5 mediates lipid metabolism that is abnormally altered in PRAD. Here, we investigated the potential cross-talk between SPOP and LRP5 in PRAD. We find a negative correlation between SPOP and LRP5 proteins in PRAD. SPOP knockdown increased LRP5 protein while SPOP overexpression resulted in LRP5 reduction that was fully rescued by proteasome inhibitors. LRP5 intracellular tail has SPOP binding site and the direct interaction between LRP5 and SPOP was confirmed by Co-IP and GST-pulldown. Moreover, LRP5 competed with Daxx for SPOP-mediated degradation, establishing a dynamic balance among SPOP, LRP5 and Daxx. Overexpression of LRP5 tail could shift this balance to enhance Daxx-mediated transcriptional inhibition, and inhibit T cell activity in a co-culture system. Further, we generated human and mouse prostate cancer cell lines expressing SPOP variants (F133V, A227V, R368H). SPOP-F133V and SPOP-A227V have specific effects in up-regulating the protein levels of PD-1 and PD-L1. Consistently, SPOP-F133V and SPOP-A227V show robust inhibitory effects on T cells compared to WT SPOP in co-culture. This is further supported by the mouse syngeneic model showing that SPOP-F133V and SPOP-A227V enhance tumorigenesis of prostate cancer in in-vivo condition. Taken together, our study provides evidence that SPOP-LRP5 crosstalk plays an essential role, and the genetic variants of SPOP differentially modulate the expression and activity of immune checkpoints in prostate cancer.
Collapse
Affiliation(s)
- Sishun Gan
- Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Medical Center of Soochow University, PR China; Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, PR China
| | - Fajun Qu
- Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, PR China
| | - Xiangmin Zhang
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, PR China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, PR China
| | - Da Xu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, PR China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, PR China.
| | - Jianquan Hou
- Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Medical Center of Soochow University, PR China.
| |
Collapse
|
24
|
Gebrael G, Zengin Z, Swami U. Differential Impact of SPOP Mutation in Prostate and Endometrial Cancers. JCO Precis Oncol 2023; 7:e2300397. [PMID: 37972335 DOI: 10.1200/po.23.00397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023] Open
Abstract
Editorial on differential impact of mutSPOP on prostate & endometrial Ca
Collapse
Affiliation(s)
- Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
25
|
Cavalcante L, Deshmukh SK, Ribeiro JR, Carneiro BA, Dizon DS, Angara K, Mattox T, Wu S, Xiu J, Walker P, Oberley M, Nabhan C, Huang H, Antonarakis ES. Opposing Roles of SPOP Mutations in Human Prostate and Endometrial Cancers. JCO Precis Oncol 2023; 7:e2300088. [PMID: 37677121 DOI: 10.1200/po.23.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/09/2023] [Accepted: 07/12/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE Recurrent gene mutations in speckle-type POZ protein (SPOP), the substrate-binding component of E3 ubiquitin ligase, are associated with tumor progression in prostate and endometrial cancers. Here, we characterized SPOP mutations in these cancers and explored their association with molecular and immune signatures and patient outcomes. METHODS There were 7,398 prostate cancer and 19,188 endometrial cancer samples analyzed for clinical and molecular profiles at Caris Life Sciences. Overall survival (OS) was analyzed using Kaplan-Meier survival curves. Statistical significance was determined using chi-square and Mann-Whitney U tests, with P values adjusted for multiple comparisons. RESULTS SPOP mutations were identified in 9.2% of prostate and 4.3% of endometrial cancers. Mutations clustered in the SPOP meprin and TRAF-C homology domain, with no significant overlap between cancer types. SPOP mutation was associated with differential comutation profiles and opposing tumor immune microenvironment signatures for each cancer, with greater immune infiltration in SPOP-mutated endometrial cancer. SPOP-mutated prostate and endometrial cancers displayed altered epigenetic gene expression, including opposite regulation of BRD2 transcripts. In SPOP-mutant prostate cancer, higher expression of androgen receptor-regulated transcripts and improved OS after treatment with hormonal agents were observed. In endometrial cancer, hormone receptor expression was significantly lower in SPOP-mutated tumors and differences in OS were highly dependent on the particular hotspot mutation and histologic subtype. CONCLUSION These data indicate that SPOP mutations drive opposing molecular and immune landscapes in prostate and endometrial cancers-suggesting a loss-of-function mechanism in prostate cancer and gain-of-function mechanism in endometrial cancer-and provide a rationale for tailored therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Benedito A Carneiro
- Lifespan Cancer Institute, Legorreta Cancer Center at Brown University, Providence, RI
| | - Don S Dizon
- Lifespan Cancer Institute, Legorreta Cancer Center at Brown University, Providence, RI
| | | | | | | | | | | | | | | | - Haojie Huang
- Mayo Clinic College of Medicine and Science, Rochester, MN
| | | |
Collapse
|
26
|
Schwartz I, Vunjak M, Budroni V, Cantoran García A, Mastrovito M, Soderholm A, Hinterndorfer M, de Almeida M, Hacker K, Wang J, Froussios K, Jude J, Decker T, Zuber J, Versteeg GA. SPOP targets the immune transcription factor IRF1 for proteasomal degradation. eLife 2023; 12:e89951. [PMID: 37622993 PMCID: PMC10491434 DOI: 10.7554/elife.89951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Adaptation of the functional proteome is essential to counter pathogens during infection, yet precisely timed degradation of these response proteins after pathogen clearance is likewise key to preventing autoimmunity. Interferon regulatory factor 1 (IRF1) plays an essential role as a transcription factor in driving the expression of immune response genes during infection. The striking difference in functional output with other IRFs is that IRF1 also drives the expression of various cell cycle inhibiting factors, making it an important tumor suppressor. Thus, it is critical to regulate the abundance of IRF1 to achieve a 'Goldilocks' zone in which there is sufficient IRF1 to prevent tumorigenesis, yet not too much which could drive excessive immune activation. Using genetic screening, we identified the E3 ligase receptor speckle type BTB/POZ protein (SPOP) to mediate IRF1 proteasomal turnover in human and mouse cells. We identified S/T-rich degrons in IRF1 required for its SPOP MATH domain-dependent turnover. In the absence of SPOP, elevated IRF1 protein levels functionally increased IRF1-dependent cellular responses, underpinning the biological significance of SPOP in curtailing IRF1 protein abundance.
Collapse
Affiliation(s)
- Irene Schwartz
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Milica Vunjak
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Valentina Budroni
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Adriana Cantoran García
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Marialaura Mastrovito
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Adrian Soderholm
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Matthias Hinterndorfer
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Melanie de Almeida
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Kathrin Hacker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Jingkui Wang
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Kimon Froussios
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Julian Jude
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Thomas Decker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
- Medical University of Vienna, Vienna BioCenterViennaAustria
| | - Gijs A Versteeg
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| |
Collapse
|
27
|
Ong JY, Torres JZ. Cul3 substrate adaptor SPOP targets Nup153 for degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.13.540659. [PMID: 37293018 PMCID: PMC10245568 DOI: 10.1101/2023.05.13.540659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
SPOP is a Cul3 substrate adaptor responsible for degradation of many proteins related to cell growth and proliferation. Because mutation or misregulation of SPOP drives cancer progression, understanding the suite of SPOP substrates is important to understanding regulation of cell proliferation. Here, we identify Nup153, a component of the nuclear basket of the nuclear pore complex, as a novel substrate of SPOP. SPOP and Nup153 bind to each other and colocalize at the nuclear envelope and some nuclear foci in cells. The binding interaction between SPOP and Nup153 is complex and multivalent. Nup153 is ubiquitylated and degraded upon expression of SPOPWT but not its substrate binding-deficient mutant SPOPF102C. Depletion of SPOP via RNAi leads to Nup153 stabilization. Upon loss of SPOP, the nuclear envelope localization of spindle assembly checkpoint protein Mad1, which is tethered to the nuclear envelope by Nup153, is stronger. Altogether, our results demonstrate SPOP regulates Nup153 levels and expands our understanding of the role of SPOP in protein and cellular homeostasis.
Collapse
Affiliation(s)
- Joseph Y Ong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
28
|
Liu J, Zheng R, Zhang Y, Jia S, He Y, Liu J. The Cross Talk between Cellular Senescence and Melanoma: From Molecular Pathogenesis to Target Therapies. Cancers (Basel) 2023; 15:cancers15092640. [PMID: 37174106 PMCID: PMC10177054 DOI: 10.3390/cancers15092640] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Melanoma is a malignant skin tumor that originates from melanocytes. The pathogenesis of melanoma involves a complex interaction that occurs between environmental factors, ultraviolet (UV)-light damage, and genetic alterations. UV light is the primary driver of the skin aging process and development of melanoma, which can induce reactive oxygen species (ROS) production and the presence of DNA damage in the cells, and results in cell senescence. As cellular senescence plays an important role in the relationship that exists between the skin aging process and the development of melanoma, the present study provides insight into the literature concerning the topic at present and discusses the relationship between skin aging and melanoma, including the mechanisms of cellular senescence that drive melanoma progression, the microenvironment in relation to skin aging and melanoma factors, and the therapeutics concerning melanoma. This review focuses on defining the role of cellular senescence in the process of melanoma carcinogenesis and discusses the targeting of senescent cells through therapeutic approaches, highlighting the areas that require more extensive research in the field.
Collapse
Affiliation(s)
- Jiahua Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Runzi Zheng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Yanghuan Zhang
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Shuting Jia
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Yonghan He
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Jing Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
29
|
Hou B, Chen T, Zhang H, Li J, Wang P, Shang G. The E3 ubiquitin ligases regulate PD-1/PD-L1 protein levels in tumor microenvironment to improve immunotherapy. Front Immunol 2023; 14:1123244. [PMID: 36733484 PMCID: PMC9887025 DOI: 10.3389/fimmu.2023.1123244] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
The tumor microenvironment (TME) is the tumor surrounding environment, which is critical for tumor development and progression. TME is also involved in clinical intervention and treatment outcomes. Modulation of TME is useful for improving therapy strategies. PD-L1 protein on tumor cells interacts with PD-1 protein on T cells, contributing to T cell dysfunction and exhaustion, blockage of the immune response. Evidence has demonstrated that the expression of PD-1/PD-L1 is associated with clinical response to anti-PD-1/PD-L1 therapy in cancer patients. It is important to discuss the regulatory machinery how PD-1/PD-L1 protein is finely regulated in tumor cells. In recent years, studies have demonstrated that PD-1/PD-L1 expression was governed by various E3 ubiquitin ligases in TME, contributing to resistance of anti-PD-1/PD-L1 therapy in human cancers. In this review, we will discuss the role and molecular mechanisms of E3 ligases-mediated regulation of PD-1 and PD-L1 in TME. Moreover, we will describe how E3 ligases-involved PD-1/PD-L1 regulation alters anti-PD-1/PD-L1 efficacy. Altogether, targeting E3 ubiquitin ligases to control the PD-1/PD-L1 protein levels could be a potential strategy to potentiate immunotherapeutic effects in cancer patients.
Collapse
Affiliation(s)
- Bo Hou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiatong Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Guanning Shang,
| |
Collapse
|