1
|
Zhang L, Liu T, Chen M, Gao S, Staley CA, Yang L, Zhu L. Dual inhibition of oxidative phosphorylation and glycolysis using a hyaluronic acid nanoparticle NOX inhibitor enhanced response to radiotherapy in colorectal cancer. Biomaterials 2025; 323:123437. [PMID: 40449083 DOI: 10.1016/j.biomaterials.2025.123437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/04/2025] [Accepted: 05/25/2025] [Indexed: 06/02/2025]
Abstract
Metabolic reprogramming characterized by mitochondrial dysfunction and increased glycolysis is associated with aggressive tumor biology and poor therapeutic response. The interplays among NADPH oxidase (NOX)-mediated reactive oxygen species, regulation of glycolysis and oxidative phosphorylation (OXPHOS) in cancer cells suggest an opportunity to develop a new cancer therapy. We found that treatment with a hyaluronic acid nanoparticle encapsulated with GKT831 (HANP/GKT831), a NOX1/4 inhibitor, markedly inhibited the proliferation and invasion of cancer cells. Treated tumor cells had reduced levels of mitochondrial ROS, glycolysis, and OXPHOS. The combination of HANP/GKT831 with radiation reduced colony formation and invasion of tumor cells. The combination therapy markedly inhibited the levels of molecules in glycolysis, OXPHOS, and DNA repairing pathways in tumor cells. Systemic administrations of HANP/GKT831 combined with radiotherapy significantly inhibited tumor growth by 84.7 % in a mouse colorectal tumor model. Tumors treated with HANP/GKT831 and radiation had increased DNA damage and apoptotic cell death. Furthermore, the combined therapy increased intratumoral infiltration of activated cytotoxic T cells and M1 macrophages but reduced the levels of immunosuppressive fibroblasts and M2 macrophages. Our results support HANP/GKT831 as a cancer nanotherapeutic agent that induces redox and bioenergy stresses in cancer cells for enhanced therapeutic response to radiotherapy.
Collapse
Affiliation(s)
- Lumeng Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States
| | - Minglong Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Shi Gao
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Charles A Staley
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States.
| |
Collapse
|
2
|
Liao Y, Zhong L, Zhao Y, Wan P, Zhang Y, Deng Y, Zhang H, Wang M, Liu B. OTUB1 promotes the progression of acute myeloid leukemia by regulating glycolysis via deubiquitinating c-Myc. Cell Signal 2025; 131:111735. [PMID: 40081551 DOI: 10.1016/j.cellsig.2025.111735] [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: 11/19/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Acute myeloid leukemia (AML) is the most common type of adult leukemia and patients with AML often have poor prognosis, for which there remains an urgent need to identify novel selective targeted therapy. OTUB1, a deubiquitinating enzyme, is associated with the malignant progression of multiple cancers. However, the role of OTUB1 in AML is still unclear and warrants further investigations. Our study revealed that the expression of OTUB1 is significantly upregulated in AML. Next, we observed that knockdown of OTUB1 inhibits AML cell proliferation and promotes AML cell apoptosis and G0/G1 phase blockade using CCK-8 assay, western blotting, and flow cytometry. Mechanistically, OTUB1 drives the malignant development of AML through regulating cellular aerobic glycolysis by deubiquitinating c-Myc. Lastly, by investigating whether inhibition of OTUB1 enhances the sensitivity of chemotherapeutic agents commonly used in the clinical treatment of AML, we found that combining OTUB1 inhibition with daunorubicin treatment could achieve better therapeutic effects in AML. In brief, our results revealed a novel mechanism by which OTUB1 promotes glycolysis via deubiquitinating c-Myc in AML. Consequently, targeting OTUB1 may provide a promising strategy for enhancing the efficacy of AML treatment.
Collapse
Affiliation(s)
- Yang Liao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yi Zhao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Ying Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Ying Deng
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Hongyan Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Meng Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China; Clinical Laboratory of the Affiliated Rehabilitation Hospital, Chongqing Medical University, Chongqing 400050, China.
| |
Collapse
|
3
|
Huang C, Liao J, Cen X, Jiao C, Chen S, Liu D, Qu HS, Zhu J, He S. TJ0113-induced mitophagy in acute liver failure detected by Raman microspectroscopy. Redox Biol 2025; 83:103654. [PMID: 40318303 PMCID: PMC12098161 DOI: 10.1016/j.redox.2025.103654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
Impaired mitophagy underlies the pathophysiology of acute liver failure (ALF) and is closely associated with tissue damage and dysfunction. A novel mitophagy inducer, TJ0113, was used for treatment during ALF pathogenesis. In this study, we used a novel mitophagy inducer, TJ0113, to investigate the effects and mechanisms of TAA-induced ALF mice. The results showed that TJ0113 could enhance mitophagy through Parkin/PINK1 and ATG5 pathways, which in turn attenuated mitochondrial damage, hepatocyte apoptosis, nuclear factor (NF)-κB/NLRP3 signaling activation and inflammatory responses after TAA. Metabolomics results showed that TJ0113 mainly regulated lipid metabolism, amino acid metabolism and nucleotide metabolism in the livers of ALF mice. RNA sequencing (RNA-seq) analysis yielded that TJ0113 was involved in the development of ALF by regulating the P13K/AKT signaling pathway. The key highlight of this work is the use of an aberration-free line-scanning confocal Raman imager (AFLSCRI) to study the molecular changes in blood, liver tissue, gastrocnemius muscle, and mitochondrial extracts in ALF mice after TJ0113 treatment. Compared to the measurement with conventional assays, Raman microspectroscopy (micro-Raman) offers the benefits of being rapid, non-invasive, label-free and real-time. Our results found good agreement between Raman signals and histopathologic findings. The system has good performance with a spatial resolution of 2 μm, a spectral resolution of 4 cm-1 and a fast detection speed improved by 2 orders. Innovations in this test contribute to clinical diagnosis of disease, personalized treatment, effective intraoperative guidance and accurate prognosis. The data may help in the development of a non-invasive clinical device for mitochondrial damage using bedside micro-Raman.
Collapse
Affiliation(s)
- Chunlian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China; MedEnglnfo Collaborative Research Center, Zhejiang Engineering Research Center for Intelligent Medical Imaging, Sensing and Non-invasive Rapid Testing, Taizhou Hospital, Zhejiang University, Taizhou, China
| | - Jiaqi Liao
- Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310058, China; MedEnglnfo Collaborative Research Center, Zhejiang Engineering Research Center for Intelligent Medical Imaging, Sensing and Non-invasive Rapid Testing, Taizhou Hospital, Zhejiang University, Taizhou, China; National Engineering Research Center for Optical Instruments, Zhejiang University, Hangzhou, 310058, China
| | - Xufeng Cen
- Research Center of Clinical Pharmacy of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Changwei Jiao
- Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Sijia Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China
| | - Dong Liu
- Hangzhou PhecdaMed Co., Ltd. Third Floor, Building 2, No.2626. Yuhangtang Road, Yuhang District, Hangzhou City, Zhejiang Province, China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China
| | - Jiansheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China.
| | - Sailing He
- MedEnglnfo Collaborative Research Center, Zhejiang Engineering Research Center for Intelligent Medical Imaging, Sensing and Non-invasive Rapid Testing, Taizhou Hospital, Zhejiang University, Taizhou, China; National Engineering Research Center for Optical Instruments, Zhejiang University, Hangzhou, 310058, China; Department of Electromagnetic Engineering, School of Electrical Engineering, Royal Institute of Technology, 100 44, Stockholm, Sweden.
| |
Collapse
|
4
|
Zhang X, Zhao T, Wu C, Shen H, Yi J, Liu L. LncRNA CASC19 promotes the growth and glycolysis of colorectal cancer cells and tumor metastasis in mice. BMC Cancer 2025; 25:829. [PMID: 40329227 PMCID: PMC12053857 DOI: 10.1186/s12885-025-14170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
It has been reported that lncRNA CASC19 is abnormally highly expressed in colorectal cancer (CRC) progression, suggesting that it may regulate the occurrence and metastasis of CRC, but its specific mechanism is still unclear. To further explore the effect of CASC19 on CRC, we overexpressed or knocked down CASC19 in HR4838 cells. The results of Transwell invasion assay and cell clonogenic assay showed that CASC19 promoted cell invasion and proliferation. Flow cytometry results showed that CASC19 inhibited cell apoptosis. In addition, by detecting glucose uptake, lactate content and ATP production, it was found that CASC19 promoted glycolysis, while CASC19 silencing had the opposite effect. Interestingly, small nuclear ribonucleoprotein polypeptide A (SNRPA) is an RNA binding protein of CASC19. Overexpression of SNRPA promoted tumor cell invasion, proliferation, glycolysis, and inhibits apoptosis, while SNRPA silencing has the opposite effect. Moreover, SNRPA overexpression reversed the inhibitory effect of CASC19-sh on invasion, proliferation and glycolysis of HR4838 cells and the promoting effect on apoptosis, which was mediated by activating the Wnt/β-catenin pathway. In the subcutaneous transplantation tumor model of BALB/c nude mice, we observed that the tumor growth of CASC19 knockdown mice was slower, and the tumor weight and volume were smaller, which was related to the low expression of CASC19 and SNRPA. In conclusion, our results showed that CASC19 promoted the growth and glycolysis of CRC cells and tumor metastasis in mice by upregulating SNRPA, which may provide a new molecular marker for the diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Tingyu Zhao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Cheng Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hengyang Shen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jianing Yi
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
5
|
Wang S, Li Y, Wang Z, Geng C, Chen P, Li Z, Li C, Bai X. Constructing a mitochondrial-related genes model based on machine learning for predicting the prognosis and therapeutic effect in colorectal cancer. Discov Oncol 2025; 16:661. [PMID: 40317411 PMCID: PMC12049353 DOI: 10.1007/s12672-025-02462-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
The role of mitochondria in tumorigenesis and progression is has been increasingly demonstrated by numerous studies, but its prognostic value in colorectal cancer (CRC) remains unclear. To address this, we developed a mitochondrial-related gene prognostic model using 101 combinations of 10 machine learning algorithms. Patients in the high-risk group exhibited significantly shorter overall survival time. The high-risk group exhibited elevated tumor immune dysfunction and exclusion score, indicating diminished immunotherapy efficacy. To address the suboptimal treatment outcomes in these patients, we identified PYR-41 and pentostatin as potential therapeutic agents, which are anticipated to enhance therapeutic efficacy in the high-risk group. Additionally, four biomarkers (HSPA1A, CHDH, TRAP1, CDC25C) were validated by quantitative real-time PCR, with significant expression differences between normal intestinal epithelial cells and colon cancer cells. Our prognostic model provides accurate CRC outcome prediction and guides personalized therapeutic strategies.
Collapse
Affiliation(s)
- Shaoke Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Yien Li
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Zhihui Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Changhui Geng
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Peng Chen
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Zhengang Li
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Chenxu Li
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Xuefeng Bai
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
6
|
Xiao Z, Wang Y, Pan D, Liu X, Gan J, Huang L, Feng Y. USP3 promotes clear cell renal cell carcinoma progression by stabilizing MYC and enhancing glycolysis. Biochim Biophys Acta Gen Subj 2025; 1869:130801. [PMID: 40164288 DOI: 10.1016/j.bbagen.2025.130801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of renal malignancy, and the deubiquitinase USP3 has been implicated as a critical factor in tumor biology. However, the precise mechanisms by which USP3 contributes to ccRCC progression remain unclear. This study investigates the role of USP3 in ccRCC and elucidates its underlying molecular mechanisms. Data from TCGA and GTEx databases showed elevated USP3 expression in ccRCC tissues and cell lines compared to normal renal tissues. Further analysis using qPCR and Western blot confirmed this upregulation in ccRCC cell lines. Functional assays revealed that silencing USP3 significantly impaired cell proliferation, migration, and invasion, while promoting apoptosis. Additionally, co-immunoprecipitation assays demonstrated an interaction between USP3 and MYC, with subsequent ubiquitination assays showing that USP3 regulates MYC stability. USP3 depletion also led to alterations in glycolysis-related gene expression, which could be partially reversed by MYC overexpression. These findings suggest that USP3 modulates ccRCC progression by stabilizing MYC, highlighting its potential as a therapeutic target in ccRCC treatment.
Collapse
Affiliation(s)
- Zhiliang Xiao
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yuan Wang
- Guangzhou Medical University, Guangzhou 511436, China
| | - Dehua Pan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Xin Liu
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Jin Gan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Liang Huang
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yan Feng
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China.
| |
Collapse
|
7
|
Zhu K, Fan J, Cai H, Zhou C, Gong Z, Li Z, Yu J. The highly expressed GOLPH3 in colorectal cancer cells activates smoothened to drive glycolysis and promote cancer cell growth and radiotherapy resistance. J Gastrointest Oncol 2025; 16:415-434. [PMID: 40386617 PMCID: PMC12078822 DOI: 10.21037/jgo-2025-193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Background Colorectal cancer (CRC) is a frequently diagnosed cancer across the world and has increased in prevalence over the last decade. This study aimed to assess the biological roles, influences on radiosensitivity, and possible molecular mechanism of Golgi phosphoprotein 3 (GOLPH3) in CRC. Methods Western blotting, quantitative real-time polymerase chain reaction (qRT-PCR), and immunohistochemistry (IHC) were used to examine GOLPH3 expression. In vivo and in vitro assays were carried out to clarify the function of GOLPH3 in CRC. The differentially expressed genes (DEGs) in CRC cells with knockdown of GOLPH3 were identified through RNA sequencing (RNA-seq). Based on the DEGs associated with GOLPH3 knockdown and the data from The Cancer Genome Atlas (TCGA) database, the pathways that could be regulated by GOLPH3 were predicted via Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Results In CRC, GOLPH3 was upregulated, and GOLPH3 upregulation was predictive of a poor prognosis. GOLPH3 knockdown inhibited CRC cell proliferation, migration, and invasion but promoted apoptosis and reduced radiotherapy resistance. Conversely, in CRC cells with GOLPH3 overexpression, malignant biological behavior and radiotherapy resistance were enhanced. In vivo, GOLPH3 knockdown impeded tumor growth. Mechanistically, GOLPH3 promoted the localization of smoothened (SMO) on the cell membrane, thereby activating AMP-activated protein kinase (AMPK)-mediated glycolysis. Additionally, the final product of glycolysis, lactate, induced H3 lysine 18 lactylation (H3K18), which could be enriched on the promoter of GOLPH3 and stimulate the transcription of GOLPH3. Conclusions GOLPH3 promoted CRC progression and enhanced radiotherapy resistance via glycolysis mediated by the SMO-AMPK axis.
Collapse
Affiliation(s)
- Kunli Zhu
- Shandong University Cancer Center, Shandong University, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Jing Fan
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Hongchao Cai
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Changchun Zhou
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital &Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Zhe Gong
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Shandong University Cancer Center, Shandong University, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
8
|
Kang Y, Meng Y, Jin J, Dai Y, Li F, Chen N, Xie H, Cui Y. Mitochondrial metabolism-related features guiding precision subtyping and prognosis in breast cancer, revealing FADS2 as a novel therapeutic target. Transl Oncol 2025; 54:102330. [PMID: 39986190 PMCID: PMC11904520 DOI: 10.1016/j.tranon.2025.102330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Breast cancer is one of the most prevalent malignant tumors in women. Mitochondria, essential for cellular function, have altered metabolic activity in cancer cells, influencing tumor regulation and clinical outcomes. The connection between mitochondrial metabolism-related genes and breast cancer prognosis remains underexplored. This study aims to investigate the role of these genes in breast cancer by constructing risk models. METHODS Breast cancer transcriptome data were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and mitochondrial gene data were sourced from the MitoCarta3.01 database. Clustering analysis was conducted using the "ConsensusClusterPlus" package, followed by Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. A prognostic model was built using Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) algorithms. Immune cell infiltration levels were assessed via CIBERSORT and MCPcounter algorithms. Validation of key gene expression was performed on breast cancer tissue specimens and cell models to explore their biological functions in breast cancer cells. RESULTS The LASSO regression analysis of the TCGA BRCA dataset identified four prognosis-related mitochondrial metabolism genes: MYH11, LTF, FADS2, and PSPHP1. Validation using the GEO dataset confirmed that patients with high-risk scores (based on these four genes) had shorter overall survival compared to those with lower risk scores. Immunological analysis revealed that high-risk patients were less responsive to immunotherapy but more sensitive to conventional chemotherapies. This suggests that combining chemotherapy with immunotherapy might enhance T cell-based treatments. Univariate and multivariate Cox regression confirmed that the mitochondrial gene model was an independent predictor of overall survival, and a nomogram was developed to predict patient prognosis. Tissue validation showed consistent expression patterns with bioinformatic predictions. Functional assays confirmed that FADS2 was highly expressed in breast cancer cells, and its knockout significantly reduced cell invasion, migration, and colony formation. CONCLUSION This study reveals that mitochondrial metabolism-related genes are closely associated with breast cancer progression, clinical outcomes, and genetic alterations. The findings may offer new avenues for treatment strategies, early intervention, and prognosis prediction in breast cancer.
Collapse
Affiliation(s)
- Yakun Kang
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - You Meng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Jiangdong Jin
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yuhan Dai
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Fei Li
- Nanjing Medical University, Nanjing 211166, China
| | - Nuo Chen
- Nanjing Medical University, Nanjing 211166, China
| | - Hui Xie
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Yangyang Cui
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
9
|
Wang D, Wang S, Liu J, Shi X, Xiong T, Li R, Wei W, Ji L, Huang Q, Gong X, Ai K. Nanomedicine Penetrating Blood-Pancreas Barrier for Effective Treatment of Acute Pancreatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413925. [PMID: 39950925 PMCID: PMC11967758 DOI: 10.1002/advs.202413925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/01/2025] [Indexed: 04/05/2025]
Abstract
Acute pancreatitis (AP) is a primary contributor to hospitalization and in-hospital mortality worldwide. Targeted elimination of mitochondrial reactive oxygen species (mtROS) within pancreatic acinar cells (PACs) represents an ideal strategy for treating AP. However, existing drugs fail to overcome the physiological barriers of the pancreas to effectively reach PACs mitochondria due to the trade-off between conventional positively charged mitochondrial-targeting groups and their inability to penetrate the blood-pancreas barrier (BPB). Here, a tungsten-based heteropolyacid nano-antioxidant (mTWNDs) is introduced, co-modified with tannic acid (TA) and melanin, enabling site-specific clearance of mtROS in PACs, offering a highly effective treatment for AP. TA exhibits a strong affinity for proline-rich type III collagen and the mitochondrial outer membrane protein TOM20. This unique property allows mTWNDs to traverse the damaged BPB-exposing type III collagen to reach PACs and subsequently penetrate mitochondria for targeted mtROS elimination. In cerulein-induced AP mice, mTWNDs reversed AP at 1/50th the dose of N-acetylcysteine, suppressing PACs apoptosis and inflammation by blocking the stimulator of the interferon genes pathway activation in macrophage. This study establishes a mitochondrial-targeting antioxidant nanomedicine strategy for AP treatment.
Collapse
Affiliation(s)
- Dan Wang
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Shuya Wang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Jinjin Liu
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Tingli Xiong
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Ruishi Li
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Wei Wei
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Liandong Ji
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xuejun Gong
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of EducationXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
10
|
Liu X, Zhu Y, Huang W, Chen J, Lan J, Long X, Zhou J. MYG1 interacts with HSP90 to promote breast cancer progression through Wnt/β-catenin and Notch signaling pathways. Exp Cell Res 2025; 446:114448. [PMID: 39956425 DOI: 10.1016/j.yexcr.2025.114448] [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: 09/10/2024] [Revised: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND As an evolutionarily conserved gene involved in embryonic development, cell differentiation, and immune metabolism, MYG1 exhibits a dynamic expression pattern related to development in human and mouse embryonic tissues, especially upregulates in undifferentiated or pluripotent stem cells. However, MYG1 has been poorly studied in breast cancer and its functional mechanism still remains unclear. METHOD Immunohistochemistry and immunofluorescence were used to study MYG1 expression and localization in breast cancer. Lentivirus transfection combined with CCK8, colony formation, matrix gel experiment and breast fat pad tumor formation in nude mice were used for in vivo and in vitro functional assessment. GSEA enrichment analysis, immunofluorescence and Western blot were conducted to explore functional mechanism. RESULT MYG1 expression was upregulated in breast cancer and its higher expression correlated with a variety of clinicopathological characteristics indicating poor prognosis. In vitro and in vivo experiments showed that overexpression of MYG1 promoted breast cancer cells proliferation, migration, invasion and tumorigenesis, while downregulation of MYG1 had an opposite effect. Mechanistically, MYG1 interacted with HSP90 to significantly activate Wnt/β-catenin and Notch signaling pathways in breast cancer cells, thus promoting EMT, cell cycle process and breast cancer progression. CONCLUSION MYG1 is highly expressed in breast cancer and functions as an oncogene. Mechanistically, MYG1 interacts with HSP90 to accelerate EMT and cell cycle process by activating both Wnt/β-catenin and Notch signaling pathways.
Collapse
Affiliation(s)
- Xuming Liu
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Yurong Zhu
- Department of Pathology, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, 78 Wandao Road, Dongguan, 523039, China; Dongguan Clinical Pathology Diagnosis Center, 14 Yuhua Road, Dongguan, 523001, China; Dongguan Key Laboratory of Clinical Pathology, 14 Yuhua Road, Dongguan, 523001, China
| | - Wenqing Huang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jianxiong Chen
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jiawen Lan
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Xiaoli Long
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Jun Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China; Department of Pathology, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, 78 Wandao Road, Dongguan, 523039, China; Dongguan Clinical Pathology Diagnosis Center, 14 Yuhua Road, Dongguan, 523001, China; Dongguan Key Laboratory of Clinical Pathology, 14 Yuhua Road, Dongguan, 523001, China.
| |
Collapse
|
11
|
Shen G, Liu S, Cao Y, Chen Z, Wang G, Yu L, Sun L, Ran Y. HSP90 co-regulates the formation and nuclear distribution of the glycolytic output complex to promote resistance and poor prognosis in gastric cancer patients. J Transl Med 2025; 23:172. [PMID: 39930487 PMCID: PMC11812214 DOI: 10.1186/s12967-025-06196-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Resistance to treatment is a critical factor contributing to poor prognosis in gastric cancer patients. HSP90 has emerged as a promising therapeutic target; however, its role in regulating tumor metabolic pathways, particularly glycolysis, remains poorly understood, which limits its clinical application. METHODS We identified proteins that directly interact with HSP90 using immunoprecipitation (IP) followed by mass spectrometry. The relationship between HSP90 and glycolysis was further investigated through transcriptomic analyses and in vitro experiments. Mechanistic insights were obtained through mass spectrometry, co-immunoprecipitation (Co-IP) assays, drug sensitivity tests, and bioinformatics analyses. Additionally, we developed a scoring system based on transcriptomic data to evaluate its prognostic significance and association with treatment resistance in gastric cancer patients. RESULTS Our multi-omics and in vitro studies revealed that HSP90 regulates glycolysis and influences the stemness properties of gastric cancer cells. Mechanistically, HSP90 facilitates the assembly of a glycolytic multi-enzyme complex, termed the HGEO complex, which enhances glycolytic metabolism. Mechanistically, HSP90 facilitates the formation of a multienzyme complex comprising key enzymes including PGK1, PKM2, ENO1, and LDHA, thereby facilitating the production of the final glycolytic products. We refer to this as the "HSP90-Glycolytic Output Complex" (HGEO Complex). We quantified this phenomenon with a scoring system (HGScore), finding that patients with a high HGScore exhibited more malignant signatures, increased resistance to treatment, and poorer prognoses. Furthermore, we demonstrated that the HGEO complex is localized in the nucleus, regulated by the nuclear lamina protein LMNA, which further contributes to treatment resistance and adverse outcomes. In vitro experiments indicated that inhibiting the formation of this complex sensitizes gastric cancer cells to chemotherapy. CONCLUSION Our findings suggest that HSP90 and LMNA mediated the formation and nuclear localization of the HGEO complex, thereby enhancing the malignant traits and resistance mechanisms in gastric cancer. Targeting this pathway may offer a novel therapeutic strategy to improve treatment outcomes.
Collapse
Affiliation(s)
- Gaigai Shen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shiya Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanting Cao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zihao Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guanghui Wang
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, 810001, China
| | - Long Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lixin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yuliang Ran
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
12
|
Ma Q, Zhang W, Wu K, Shi L. The roles of KRAS in cancer metabolism, tumor microenvironment and clinical therapy. Mol Cancer 2025; 24:14. [PMID: 39806421 PMCID: PMC11727292 DOI: 10.1186/s12943-024-02218-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
KRAS is one of the most mutated genes, driving alternations in metabolic pathways that include enhanced nutrient uptaking, increased glycolysis, elevated glutaminolysis, and heightened synthesis of fatty acids and nucleotides. However, the beyond mechanisms of KRAS-modulated cancer metabolisms remain incompletely understood. In this review, we aim to summarize current knowledge on KRAS-related metabolic alterations in cancer cells and explore the prevalence and significance of KRAS mutation in shaping the tumor microenvironment and influencing epigenetic modification via various molecular activities. Given that cancer cells rely on these metabolic changes to sustain cell growth and survival, targeting these processes may represent a promising therapeutic strategy for KRAS-driven cancers.
Collapse
Affiliation(s)
- Qinglong Ma
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Wenyang Zhang
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Lei Shi
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| |
Collapse
|
13
|
Qin R, Fan X, Huang Y, Chen S, Ding R, Yao Y, Wu R, Duan Y, Li X, Khan HU, Hu J, Wang H. Role of glucose metabolic reprogramming in colorectal cancer progression and drug resistance. Transl Oncol 2024; 50:102156. [PMID: 39405607 PMCID: PMC11736406 DOI: 10.1016/j.tranon.2024.102156] [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: 08/13/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Colorectal cancer (CRC), with the incidence and mortality rising on a yearly basis, greatly threatens people's health. It is considered an important hallmark of tumorigenesis that aberrant glucose metabolism in cancer cells, particularly the Warburg effect. In CRC, the Warburg effect predominantly influences cancer development and progression via its involvement in the glycolytic pathway regarding cell metabolism. The critical mechanisms underlying this process include key glycolytic enzymes, transport proteins, regulatory molecules, and signaling pathways. Furthermore, targeting the reprogrammed glucose metabolism in cancer cells can be potentially used for CRC treatment. However, the mechanisms driving CRC onset and progression, especially in relation to glucose metabolism reprogramming, are not fully understood and represent an emerging field of research. The review aims at providing new insights into the role that glucose metabolism reprogramming plays in the progression of CRC progression together with its resistance to treatment. Ultimately, these insights strive to diminish the risks of CRC metastasis and recurrence.
Collapse
Affiliation(s)
- Rong Qin
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Xirui Fan
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Yun Huang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Sijing Chen
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Rui Ding
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Ying Yao
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Rui Wu
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Yiyao Duan
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Xiang Li
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hameed Ullah Khan
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| | - Jun Hu
- The First People's Hospital of Kunming, Yunnan 650034, China.
| | - Hui Wang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China; Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China.
| |
Collapse
|