1
|
Samsami Y, Akhlaghipour I, Taghehchian N, Palizkaran Yazdi M, Farrokhi S, Rahimi HR, Moghbeli M. MicroRNA-382 as a tumor suppressor during tumor progression. Bioorg Med Chem Lett 2024; 113:129967. [PMID: 39293533 DOI: 10.1016/j.bmcl.2024.129967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Despite the recent progresses in therapeutic and diagnostic methods, there is still a significantly high rate of mortality among cancer patients. One of the main reasons for the high mortality rate in cancer patients is late diagnosis, which leads to the failure of therapeutic strategies. Therefore, investigation of cancer biology can lead to the introduction of early diagnostic markers in these patients. MicroRNAs (miRNAs) play an important role in regulation of cellular processes associated with tumor progression. Due to the high stability of miRNAs in body fluids, these factors can be considered as the non-invasive tumor markers. Deregulation of miR-382 has been widely reported in different cancers. Therefore, in this review, we investigated the role of miR-382 during tumor development. It has shown that miR-382 has mainly a tumor suppressive, which inhibits the growth of tumor cells through the regulation of signaling pathways, RNA-binding proteins, and transcription factors. Therefore, miR-382 can be suggested as a diagnostic and therapeutic marker in cancer patients.
Collapse
Affiliation(s)
- Yalda Samsami
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Saba Farrokhi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Yan G, Zhu T, Zhou J, Li X, Wen Z, Miuhuitijiang B, Zhang Z, Du Y, Li C, Shi X, Tan W. GOLM1 promotes prostate cancer progression via interaction with PSMD1 and enhancing AR-driven transcriptional activation. J Cell Mol Med 2024; 28:e70186. [PMID: 39470578 PMCID: PMC11520440 DOI: 10.1111/jcmm.70186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Aberrant transcriptional activation of the androgen receptor (AR) is a predominant cause of prostate cancer (PCa), including both in the initial and androgen-independent stages. Our study highlights Golgi membrane protein 1 (GOLM1) as a key regulator of AR-driven transcriptional activity in PCa progression. Utilizing local clinical data and TCGA data, we have established a robust association between GOLM1 and AR target genes, and further demonstrated that GOLM1 can enhance the expression of AR target genes. We discovered that GOLM1 interacts with PSMD1, a component of the 19S regulatory complex in the 26S proteasome, using mass spectrometry and Co-IP analysis. It is well known that ubiquitin-proteasome plays a vital role in AR expression and transcriptional regulation. Our findings demonstrate that GOLM1 enhances ubiquitin proteasome activity by binding to PSMD1, thereby facilitating AR-driven transcriptional activity and PCa progression. These results indicate that GOLM1 and its associated proteins may become potential therapeutic targets for PCa characterized by dysregulated AR-driven transcriptional activation.
Collapse
Affiliation(s)
- Guang Yan
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Andrology, Shanghai Seventh People's HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tianhang Zhu
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiawei Zhou
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xia Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zonghua Wen
- Department of PathologyShenzhen University General HospitalShenzhenChina
| | | | - Zhiyong Zhang
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuejun Du
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Xiaojun Shi
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Wanlong Tan
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
3
|
Xie P, Wu M, Wang H, Zhang B, Zhang Z, Yan J, Yu M, Yu Q, Zhao Y, Huang D, Xu M, Xu W, Li H, Xu Y, Xiao Y, Guo L. GOLM1 dictates acquired Lenvatinib resistance by a GOLM1-CSN5 positive feedback loop upon EGFR signaling activation in hepatocellular carcinoma. Oncogene 2024; 43:3108-3120. [PMID: 39251847 DOI: 10.1038/s41388-024-03153-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/24/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
Lenvatinib is a multiple receptor tyrosine kinases inhibitor (TKI) authorized for first-line treatment of hepatocellular carcinoma (HCC). However, Lenvatinib resistance is common in HCC clinical treatment, highlighting the urgent need to understand mechanisms of resistance. Here, we identified Golgi membrane protein 1 (GOLM1), a type II transmembrane protein originally located in the Golgi apparatus, as a novel regulator of Lenvatinib resistance. We found GOLM1 was overexpressed in Lenvatinib resistant human HCC cell lines, blood and HCC samples. Additionally, GOLM1 overexpression contributes to Lenvatinib resistance and HCC progression in vitro and in vivo. Mechanistically, GOLM1 upregulates CSN5 expression through EGFR-STAT3 pathway. Reversely, CSN5 deubiquitinates and stabilizes GOLM1 protein by inhibiting ubiquitin-proteasome pathway of GOLM1. Furthermore, clinical specimens of HCC showed a positive correlation between the activation of the GOLM1-EGFR-STAT3-CSN5 axis. Finally, GOLM1 knockdown was found to act in synergy with Lenvatinib in subcutaneous and orthotopic mouse model. Overall, these findings identify a mechanism of resistance to Lenvatinib treatment for HCC, highlight an effective predictive biomarker of Lenvatinib response in HCC and show that targeting GOLM1 may improve the clinical benefit of Lenvatinib.
Collapse
Affiliation(s)
- Peiyi Xie
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Mengyuan Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Hui Wang
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, PR China
| | - Bo Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Zihao Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Jiuliang Yan
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Mincheng Yu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Qiang Yu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Yufei Zhao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, PR China
| | - Min Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Wenxin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Hui Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Translational Research Center, Shanghai, 200031, PR China.
| | - Yongfeng Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| | - Yongsheng Xiao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| | - Lei Guo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| |
Collapse
|
4
|
Cao P, Chen H, Zhang Y, Zhang Q, Shi M, Han H, Wang X, Jin L, Guo B, Hao R, Zhao X, Li Y, Gao C, Liu X, Wang Y, Yang A, Yang C, Si A, Li H, Song Q, He F, Zhou G. Genomic Amplification of TBC1D31 Promotes Hepatocellular Carcinoma Through Reducing the Rab22A-Mediated Endolysosomal Trafficking and Degradation of EGFR. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405459. [PMID: 39206796 PMCID: PMC11516053 DOI: 10.1002/advs.202405459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Hepatocellular carcinomas (HCCs) are characterized by a vast spectrum of somatic copy number alterations (CNAs); however, their functional relevance is largely unknown. By performing a genome-wide survey on prognosis-associated focal CNAs in 814 HCC patients by an integrative computational framework based on transcriptomic data, genomic amplification is identified at 8q24.13 as a promising candidate. Further evidence is provided that the 8q24.13 amplification-driven overexpression of Rab GTPase activating protein TBC1D31 exacerbates HCC growth and metastasis both in vitro and in vivo through activating Epidermal growth factor receptor (EGFR) signaling. Mechanistically, TBC1D31 acts as a Rab GTPase activating protein to catalyze GTP hydrolysis for Rab22A and then reduces the Rab22A-mediated endolysosomal trafficking and degradation of EGFR. Notably, overexpression of TBC1D31 markedly increases the resistance of HCC cells to lenvatinib, whereas inhibition of the TBC1D31-EGFR axis can reverse this resistance phenotype. This study highlights that TBC1D31 at 8q24.13 is a new critical oncogene, uncovers a novel mechanism of EGFR activation in HCC, and proposes the potential strategies for treating HCC patients with TBC1D31 amplification or overexpression.
Collapse
Affiliation(s)
- Pengbo Cao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Hongxia Chen
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Ying Zhang
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Qi Zhang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
- University of South ChinaHengyang421001China
| | | | - Huihui Han
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Xiaowen Wang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of LifeomicsBeijing102206China
| | - Liang Jin
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Bingqian Guo
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | | | - Xi Zhao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Yuanfeng Li
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Chengming Gao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Xinyi Liu
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Yahui Wang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Aiqing Yang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Chenning Yang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Anfeng Si
- Jinling HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210002China
| | - Hua Li
- Department of OncologyChengdu Military General HospitalChengdu610083China
| | - Qingfeng Song
- Affiliated Cancer Hospital of Guangxi Medical UniversityNanning530021China
| | - Fuchu He
- School of Life SciencesTsinghua UniversityBeijing100084China
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of LifeomicsBeijing102206China
| | - Gangqiao Zhou
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
- School of Life SciencesTsinghua UniversityBeijing100084China
- University of South ChinaHengyang421001China
- Guangxi Medical UniversityNanning530021China
- Hebei UniversityBaoding071000China
| |
Collapse
|
5
|
Lee ZY, Lee WH, Lim JS, Ali AAA, Loo JSE, Wibowo A, Mohammat MF, Foo JB. Golgi apparatus targeted therapy in cancer: Are we there yet? Life Sci 2024; 352:122868. [PMID: 38936604 DOI: 10.1016/j.lfs.2024.122868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Membrane trafficking within the Golgi apparatus plays a pivotal role in the intracellular transportation of lipids and proteins. Dysregulation of this process can give rise to various pathological manifestations, including cancer. Exploiting Golgi defects, cancer cells capitalise on aberrant membrane trafficking to facilitate signal transduction, proliferation, invasion, immune modulation, angiogenesis, and metastasis. Despite the identification of several molecular signalling pathways associated with Golgi abnormalities, there remains a lack of approved drugs specifically targeting cancer cells through the manipulation of the Golgi apparatus. In the initial section of this comprehensive review, the focus is directed towards delineating the abnormal Golgi genes and proteins implicated in carcinogenesis. Subsequently, a thorough examination is conducted on the impact of these variations on Golgi function, encompassing aspects such as vesicular trafficking, glycosylation, autophagy, oxidative mechanisms, and pH alterations. Lastly, the review provides a current update on promising Golgi apparatus-targeted inhibitors undergoing preclinical and/or clinical trials, offering insights into their potential as therapeutic interventions. Significantly more effort is required to advance these potential inhibitors to benefit patients in clinical settings.
Collapse
Affiliation(s)
- Zheng Yang Lee
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Wen Hwei Lee
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Jing Sheng Lim
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Afiqah Ali Ajmel Ali
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Jason Siau Ee Loo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancements Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Agustono Wibowo
- Faculty of Applied Science, Universiti Teknologi MARA (UiTM) Pahang, Jengka Campus, 26400 Bandar Tun Abdul Razak Jengka, Pahang, Malaysia
| | - Mohd Fazli Mohammat
- Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancements Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| |
Collapse
|
6
|
Bai W, Li B, Wu P, Li X, Huang X, Shi N, Yang C, Hu F, Xie X. The first structure of human Golm1 coiled coil domain reveals an unexpected tetramer and highlights its structural diversity. Int J Biol Macromol 2024; 275:133624. [PMID: 38964685 DOI: 10.1016/j.ijbiomac.2024.133624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Golgi membrane protein 1 (Golm1), a transmembrane protein with diverse subcellular localizations, has garnered significant attention in recent years due to its strong association with the development and progression of liver diseases and numerous cancers. Interestingly, although Golm1 is a membrane protein, the C-terminal of Golm1, which contains a coiled coil domain and a flexible acid region, can also be detected in the plasma of patients with various liver diseases. Notably, the coiled coil domain of serum Golm1 is postulated to play a pivotal role in physiological and pathological functions. However, little is currently known about the structure of this coiled coil domain and the full-length protein, which may limit our understanding of Golm1. Therefore, this study aims to address this gap in knowledge and reports the first crystal structure of the coiled coil domain of Golm1 at a resolution of 2.28 Å. Meanwhile, we have also confirmed that the Golm1 coiled coil domain in solution can form tetramer. Our results reveal that Golm1 can form a novel tetrameric structure that differs from the previous reported dimeric structure Golm1 could assemble, which may provide novel insights into the diversity of physiological functions and pathological roles.
Collapse
Affiliation(s)
- Wenfeng Bai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Bowen Li
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Pei Wu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Xinzhu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaochen Huang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Ning Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Congcong Yang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| | - Fen Hu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Department of Etiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350002, China.
| | - Xi Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
| |
Collapse
|
7
|
Chida K, Kanazawa H, Kinoshita H, Roy AM, Hakamada K, Takabe K. The role of lidocaine in cancer progression and patient survival. Pharmacol Ther 2024; 259:108654. [PMID: 38701900 PMCID: PMC11162934 DOI: 10.1016/j.pharmthera.2024.108654] [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: 01/08/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
Since its development in 1943, lidocaine has been one of the most commonly used local anesthesia agents for surgical procedures. Lidocaine alters neuronal signal transmission by prolonging the inactivation of fast voltage-gated sodium channels in the cell membrane of neurons, which are responsible for action potential propagation. Recently, it has attracted attention due to emerging evidence suggesting its potential antitumor properties, particularly in the in vitro setting. Further, local administration of lidocaine around the tumor immediately prior to surgical removal has been shown to improve overall survival in breast cancer patients. However, the exact mechanisms driving these antitumor effects remain largely unclear. In this article, we will review the existing literature on the mechanism of lidocaine as a local anesthetic, its effects on the cancer cells and the tumor microenvironment, involved pathways, and cancer progression. Additionally, we will explore recent reports highlighting its impact on clinical outcomes in cancer patients. Taken together, there remains significant ambiguity surrounding lidocaine's functions and roles in cancer biology, particularly in perioperative setting.
Collapse
Affiliation(s)
- Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | - Hirofumi Kanazawa
- The University of Texas Health Science Center at Tyler School of Medicine, TX, USA.
| | - Hirotaka Kinoshita
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan.
| | - Arya Mariam Roy
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan; Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14263, USA; Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
8
|
Lin Y, Wei D, He X, Huo L, Wang J, Zhang X, Wu Y, Zhang R, Gao Y, Kang T. RAB22A sorts epithelial growth factor receptor (EGFR) from early endosomes to recycling endosomes for microvesicles release. J Extracell Vesicles 2024; 13:e12494. [PMID: 39051763 PMCID: PMC11270584 DOI: 10.1002/jev2.12494] [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: 01/21/2024] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Microvesicles (MVs) containing proteins, nucleic acid or organelles are shed from the plasma membrane. Although the mechanisms of MV budding are well elucidated, the connection between endosomal trafficking and MV formation remains poorly understood. In this report, RAB22A is revealed to be crucial for EGFR-containing MVs formation by the RAB GTPase family screening. RAB22A recruits TBC1D2B, a GTPase-activating protein (GAP) of RAB7A, to inactivate RAB7A, thus preventing EGFR from being transported to late endosomes and lysosomes. RAB22A also engages SH3BP5L, a guanine-nucleotide exchange factor (GEF) of RAB11A, to activate RAB11A on early endosomes. Consequently, EGFR is recycled to the cell surface and packaged into MVs. Furthermore, EGFR can phosphorylate RAB22A at Tyr136, which in turn promotes EGFR-containing MVs formation. Our findings illustrate that RAB22A acts as a sorter on early endosomes to sort EGFR to recycling endosomes for MV shedding by both activating RAB11A and inactivating RAB7A.
Collapse
Affiliation(s)
- Yujie Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Denghui Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Xiaobo He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Lanqing Huo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Jingxuan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Xia Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Ying Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
| |
Collapse
|
9
|
Ye J, Gao X, Huang X, Huang S, Zeng D, Luo W, Zeng C, Lu C, Lu L, Huang H, Mo K, Huang J, Li S, Tang M, Wu T, Mai R, Luo M, Xie M, Wang S, Li Y, Lin Y, Liang R. Integrating Single-Cell and Spatial Transcriptomics to Uncover and Elucidate GP73-Mediated Pro-Angiogenic Regulatory Networks in Hepatocellular Carcinoma. RESEARCH (WASHINGTON, D.C.) 2024; 7:0387. [PMID: 38939041 PMCID: PMC11208919 DOI: 10.34133/research.0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/21/2024] [Indexed: 06/29/2024]
Abstract
Hepatocellular carcinoma (HCC) was characterized as being hypervascular. In the present study, we generated a single-cell spatial transcriptomic landscape of the vasculogenic etiology of HCC and illustrated overexpressed Golgi phosphoprotein 73 (GP73) HCC cells exerting cellular communication with vascular endothelial cells with high pro-angiogenesis potential via multiple receptor-ligand interactions in the process of tumor vascular development. Specifically, we uncovered an interactive GP73-mediated regulatory network coordinated with c-Myc, lactate, Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway, and endoplasmic reticulum stress (ERS) signals in HCC cells and elucidated its pro-angiogenic roles in vitro and in vivo. Mechanistically, we found that GP73, the pivotal hub gene, was activated by histone lactylation and c-Myc, which stimulated the phosphorylation of downstream STAT3 by directly binding STAT3 and simultaneously enhancing glucose-regulated protein 78 (GRP78)-induced ERS. STAT3 potentiates GP73-mediated pro-angiogenic functions. Clinically, serum GP73 levels were positively correlated with HCC response to anti-angiogenic regimens and were essential for a prognostic nomogram showing good predictive performance for determining 6-month and 1-year survival in patients with HCC treated with anti-angiogenic therapy. Taken together, the aforementioned data characterized the pro-angiogenic roles and mechanisms of a GP73-mediated network and proved that GP73 is a crucial tumor angiogenesis niche gene with favorable anti-angiogenic potential in the treatment of HCC.
Collapse
Affiliation(s)
- Jiazhou Ye
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
| | - Xing Gao
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Xi Huang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Shilin Huang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Dandan Zeng
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Wenfeng Luo
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Can Zeng
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Cheng Lu
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Lu Lu
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Hongyang Huang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Kaixiang Mo
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Julu Huang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Shizhou Li
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Minchao Tang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Tianzhun Wu
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Rongyun Mai
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Min Luo
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Mingzhi Xie
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Shan Wang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yongqiang Li
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yan Lin
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Rong Liang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| |
Collapse
|
10
|
Weng J, Wang Z, Hu Z, Xu W, Sun JL, Wang F, Zhou Q, Liu S, Xu M, Xu M, Gao D, Shen YH, Yi Y, Shi Y, Dong Q, Zhou C, Ren N. Repolarization of Immunosuppressive Macrophages by Targeting SLAMF7-Regulated CCL2 Signaling Sensitizes Hepatocellular Carcinoma to Immunotherapy. Cancer Res 2024; 84:1817-1833. [PMID: 38484085 DOI: 10.1158/0008-5472.can-23-3106] [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: 10/08/2023] [Revised: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 06/05/2024]
Abstract
Immune checkpoint inhibitors have limited efficacy in hepatocellular carcinoma (HCC). Macrophages are the most abundant immune cells in HCC, suggesting that a better understanding of the intrinsic processes by which tumor cells regulate macrophages could help identify strategies to improve response to immunotherapy. As signaling lymphocytic activation molecule (SLAM) family members regulate various immune functions, we investigated the role of specific SLAM receptors in the immunobiology of HCC. Comparison of the transcriptomic landscapes of immunotherapy-responsive and nonresponsive patients with advanced HCC identified SLAMF7 upregulation in immunotherapy-responsive HCC, and patients with HCC who responded to immunotherapy also displayed higher serum levels of SLAMF7. Loss of Slamf7 in liver-specific knockout mice led to increased hepatocarcinogenesis and metastasis, elevated immunosuppressive macrophage infiltration, and upregulated PD-1 expression in CD8+ T cells. HCC cell-intrinsic SLAMF7 suppressed MAPK/ATF2-mediated CCL2 expression to regulate macrophage migration and polarization in vitro. Mechanistically, SLAMF7 associated with SH2 domain-containing adaptor protein B (SHB) through its cytoplasmic 304 tyrosine site to facilitate the recruitment of SHIP1 to SLAMF7 and inhibit the ubiquitination of TRAF6, thereby attenuating MAPK pathway activation and CCL2 transcription. Pharmacological antagonism of the CCL2/CCR2 axis potentiated the therapeutic effect of anti-PD-1 antibody in orthotopic HCC mouse models with low SLAMF7 expression. In conclusion, this study highlights SLAMF7 as a regulator of macrophage function and a potential predictive biomarker of immunotherapy response in HCC. Strategies targeting CCL2 signaling to induce macrophage repolarization in HCC with low SLAMF7 might enhance the efficacy of immunotherapy. SIGNIFICANCE CCL2 upregulation caused by SLAMF7 deficiency in hepatocellular carcinoma cells induces immunosuppressive macrophage polarization and confers resistance to immune checkpoint blockade, providing potential biomarkers and targets to improve immunotherapy response in patients.
Collapse
Affiliation(s)
- Jialei Weng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Zhiqiu Hu
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, P.R. China
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Wenxin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Jia-Lei Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Fu Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qiang Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Shaoqing Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Min Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Minghao Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Dongmei Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Yi Shi
- Biomedical Research Centre, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Chenhao Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
| | - Ning Ren
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, P.R. China
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
11
|
Zhang Z, Xu S, Song M, Huang W, Yan M, Li X. Association between blood lipid levels and the risk of liver cancer: a systematic review and meta-analysis. Cancer Causes Control 2024; 35:943-953. [PMID: 38376693 PMCID: PMC11129988 DOI: 10.1007/s10552-024-01853-9] [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: 08/08/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE The association between blood lipid levels and the risk of developing liver cancer remains a subject of ongoing debate. To elucidate this association, we conducted a meta-analysis by systematically incorporating data from all relevant prospective cohort studies. METHODS We conducted a systematic search of the PubMed, Embase, Web of Science, and Cochrane Library databases covering studies published from database inception through July 2023. This study included prospective cohort studies related to lipid profiles (e.g., total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) levels) that reported hazard ratios (HRs) or relative risks (RRs) with corresponding 95% confidence intervals (95% CIs) to investigate their association with the risk of liver cancer. During the analysis process, we used fixed-effects or random-effects models based on the level of heterogeneity among the studies and obtained pooled risk ratios using these models. To ensure the robustness and reliability of the study findings, we also conducted sensitivity analyses and publication bias analyses. RESULTS After conducting a systematic search, 12 studies were identified from a total of 11,904 articles and were included in the meta-analysis. These studies included a combined population of 10,765,221 participants, among whom 31,055 cases of liver cancer were reported. The analysis revealed that the pooled HR for the serum TC concentration (highest versus lowest) was 0.45 (95% CI = 0.35-0.58, I2 = 78%). For TGs, the HR was 0.67 (95% CI = 0.46-0.96, I2 = 86%), while for HDL-C, the HR was 0.72 (95% CI = 0.58-0.90, I2 = 65%). The HR for LDL-C was 0.51 (95% CI = 0.23-1.13, I2 = 93%). CONCLUSION The findings of this study indicate that serum TC, TG, and HDL-C levels are negatively associated with liver cancer risk, suggesting that higher concentrations of these lipids are associated with a reduced risk of liver cancer. However, no significant association has been found between LDL-C levels and liver cancer risk.
Collapse
Affiliation(s)
- Zhihui Zhang
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shicong Xu
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Meixuan Song
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
| | - Weirong Huang
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Manlin Yan
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xianrong Li
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
12
|
Hu W, Liu Y, Lian C, Lu H. Genetic insight into putative causes of xanthelasma palpebrarum: a Mendelian randomization study. Front Immunol 2024; 15:1347112. [PMID: 38601164 PMCID: PMC11004296 DOI: 10.3389/fimmu.2024.1347112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Xanthelasma palpebrarum (XP) is the most common form of cutaneous xanthoma, with a prevalence of 1.1%~4.4% in the population. However, the cause of XP remains largely unknown. In the present study, we used Mendelian randomization to assess the genetic association between plasma lipids, metabolic traits, and circulating protein with XP, leveraging summary statistics from large genome-wide association studies (GWAS). Genetically predicted plasma cholesterol and LDL-C, but not HDL-C or triglyceride, were significantly associated with XP. Metabolic traits, including BMI, fasting glucose, type 2 diabetes, systolic and diastolic blood pressure, were not significantly associated with XP. Furthermore, we found genetically predicted 12 circulating proteins were associated with XP, including FN1, NTM, FCN2, GOLM1, ICAM5, PDE5A, C5, CLEC11A, CXCL1, CCL2, CCL11, CCL13. In conclusion, this study identified plasma cholesterol, LDL-C, and 12 circulating proteins to be putative causal factors for XP, highlighting the role of plasma cholesterol and inflammatory response in XP development.
Collapse
Affiliation(s)
- Wenting Hu
- Department of Dermatology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Yaozhong Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI, United States
| | - Cuihong Lian
- Department of Dermatology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Haocheng Lu
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Huang F, Guo J, Zhao N, Hou M, Gai X, Yang S, Cai P, Wang Y, Ma Q, Zhao Q, Li L, Yang H, Jing Y, Jin D, Hu Z, Zha X, Wang H, Mao Y, Liu F, Zhang H. PTEN deficiency potentiates HBV-associated liver cancer development through augmented GP73/GOLM1. J Transl Med 2024; 22:254. [PMID: 38459588 PMCID: PMC10924424 DOI: 10.1186/s12967-024-04976-4] [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: 09/24/2023] [Accepted: 02/10/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Although hepatitis B virus (HBV) infection is a major risk factor for hepatic cancer, the majority of HBV carriers do not develop this lethal disease. Additional molecular alterations are thus implicated in the process of liver tumorigenesis. Since phosphatase and tensin homolog (PTEN) is decreased in approximately half of liver cancers, we investigated the significance of PTEN deficiency in HBV-related hepatocarcinogenesis. METHODS HBV-positive human liver cancer tissues were checked for PTEN expression. Transgenic HBV, Alb-Cre and Ptenfl/fl mice were inter-crossed to generate WT, HBV, Pten-/- and HBV; Pten-/- mice. Immunoblotting, histological analysis and qRT-PCR were used to study these livers. Gp73-/- mice were then mated with HBV; Pten-/- mice to illustrate the role of hepatic tumor biomarker golgi membrane protein 73 (GP73)/ golgi membrane protein 1 (GOLM1) in hepatic oncogenesis. RESULTS Pten deletion and HBV transgene synergistically aggravated liver injury, inflammation, fibrosis and development of mixed hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). GP73 was augmented in HBV; Pten-/- livers. Knockout of GP73 blunted the synergistic effect of deficient Pten and transgenic HBV on liver injury, inflammation, fibrosis and cancer development. CONCLUSIONS This mixed HCC-ICC mouse model mimics liver cancer patients harboring HBV infection and PTEN/AKT signaling pathway alteration. Targeting GP73 is a promising therapeutic strategy for cancer patients with HBV infection and PTEN alteration.
Collapse
Affiliation(s)
- Fuqiang Huang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Jing Guo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Na Zhao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
- Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mengjie Hou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Xiaochen Gai
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Shuhui Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Pei Cai
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Yanan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Qian Ma
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qi Zhao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Li Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Jing
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Di Jin
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Hongyang Wang
- International Co-Operation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fangming Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China.
| | - Hongbing Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China.
| |
Collapse
|
14
|
Zhang Z, Leng XK, Zhai YY, Zhang X, Sun ZW, Xiao JY, Lu JF, Liu K, Xia B, Gao Q, Jia M, Xu CQ, Jiang YN, Zhang XG, Tao KS, Wu JW. Deficiency of ASGR1 promotes liver injury by increasing GP73-mediated hepatic endoplasmic reticulum stress. Nat Commun 2024; 15:1908. [PMID: 38459023 PMCID: PMC10924105 DOI: 10.1038/s41467-024-46135-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Liver injury is a core pathological process in the majority of liver diseases, yet the genetic factors predisposing individuals to its initiation and progression remain poorly understood. Here we show that asialoglycoprotein receptor 1 (ASGR1), a lectin specifically expressed in the liver, is downregulated in patients with liver fibrosis or cirrhosis and male mice with liver injury. ASGR1 deficiency exacerbates while its overexpression mitigates acetaminophen-induced acute and CCl4-induced chronic liver injuries in male mice. Mechanistically, ASGR1 binds to an endoplasmic reticulum stress mediator GP73 and facilitates its lysosomal degradation. ASGR1 depletion increases circulating GP73 levels and promotes the interaction between GP73 and BIP to activate endoplasmic reticulum stress, leading to liver injury. Neutralization of GP73 not only attenuates ASGR1 deficiency-induced liver injuries but also improves survival in mice received a lethal dose of acetaminophen. Collectively, these findings identify ASGR1 as a potential genetic determinant of susceptibility to liver injury and propose it as a therapeutic target for the treatment of liver injury.
Collapse
Affiliation(s)
- Zhe Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiang Kai Leng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yuan Yuan Zhai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiao Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhi Wei Sun
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Jun Ying Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Kun Liu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Air Force Medical University, Xi'an, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qi Gao
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Miao Jia
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Cheng Qi Xu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Na Jiang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Gang Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Kai Shan Tao
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Air Force Medical University, Xi'an, China.
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
15
|
Hou K, Xu X, Ge X, Jiang J, Ouyang F. Blockade of PD-1 and CTLA-4: A potent immunotherapeutic approach for hepatocellular carcinoma. Biofactors 2024; 50:250-265. [PMID: 37921427 DOI: 10.1002/biof.2012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 09/07/2023] [Indexed: 11/04/2023]
Abstract
Immune checkpoints (ICPs) can promote tumor growth and prevent immunity-induced cancer cell apoptosis. Fortunately, targeting ICPs, such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4), has achieved great success in the past few years and has gradually become an effective treatment for cancers, including hepatocellular carcinoma (HCC). However, many patients do not respond to ICP therapy due to acquired resistance and recurrence. Therefore, clarifying the specific mechanisms of ICP in the development of HCC is very important for enhancing the efficacy of anti-PD-1 and anti-CTLA-4 therapy. In particular, antigen presentation and interferon-γ (IFN-γ) signaling were reported to be involved in the development of resistance. In this review, we have explained the role and regulatory mechanisms of ICP therapy in HCC pathology. Moreover, we have also elaborated on combinations of ICP inhibitors and other treatments to enhance the antitumor effect. Collectively, recent advances in the pharmacological targeting of ICPs provide insights for the development of a novel alternative treatment for HCC.
Collapse
Affiliation(s)
- Kai Hou
- Clinical Research Center of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Xiaohui Xu
- Department of Medicine of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Xin Ge
- Clinical Research Center of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Jiacen Jiang
- Department of Medicine of the Second Affiliated Hospital, University of South China, Hengyang, Hunan, PR China
| | - Fan Ouyang
- Department of Cardiology, Zhuzhou Hospital, the Affiliated Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, PR China
| |
Collapse
|
16
|
Wang Y, Hu D, Wan L, Yang S, Liu S, Wang Z, Li J, Li J, Zheng Z, Cheng C, Wang Y, Wang H, Tian X, Chen W, Li S, Zhang J, Zha X, Chen J, Zhang H, Xu KF. GOLM1 Promotes Pulmonary Fibrosis through Upregulation of NEAT1. Am J Respir Cell Mol Biol 2024; 70:178-192. [PMID: 38029327 DOI: 10.1165/rcmb.2023-0151oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/29/2023] [Indexed: 12/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal progressive disease with elusive molecular mechanisms and limited therapeutic options. Aberrant activation of fibroblasts is a central hallmark of lung fibrosis. Here, we report that Golgi membrane protein 1 (GOLM1, also known as GP73 or GOLPH2) was increased in the lungs of patients with pulmonary fibrosis and mice with bleomycin (BLM)-induced pulmonary fibrosis. Loss of GOLM1 inhibited proliferation, differentiation, and extracellular matrix deposition of fibroblasts, whereas overexpression of GOLM1 exerted the opposite effects. Similarly, worsening pulmonary fibrosis after BLM treatment was observed in GOLM1-knock-in mice, whereas BLM-treated Golm1-knockout mice exhibited alleviated pulmonary fibrosis and collagen deposition. Furthermore, we identified long noncoding RNA NEAT1 downstream of GOLM1 as a potential mediator of pulmonary fibrosis through increased GOLM1 expression. Depletion of NEAT1 inhibited fibroblast proliferation and extracellular matrix production and reversed the profibrotic effects of GOLM1 overexpression. Additionally, we identified KLF4 as a downstream mediator of GOLM1 signaling to NEAT1. Our findings suggest that GOLM1 plays a pivotal role in promoting pulmonary fibrosis through the GOLM1-KLF4-NEAT1 signaling axis. Targeting GOLM1 and its downstream pathways may represent a novel therapeutic strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Yani Wang
- Department of Pulmonary and Critical Care Medicine and
| | - Danjing Hu
- Department of Pulmonary and Critical Care Medicine and
| | - Linyan Wan
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhui Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song Liu
- Medical Science Center, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixi Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Jie Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Li
- Department of Pulmonary and Critical Care Medicine and
| | - Zhoude Zheng
- Department of Pulmonary and Critical Care Medicine and
| | | | - Yanan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanghang Wang
- Department of Pulmonary and Critical Care Medicine and
| | - Xinlun Tian
- Department of Pulmonary and Critical Care Medicine and
| | - Wenhui Chen
- Department of Lung Transplantation, Centre for Lung Transplantation, Centre for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and
| | - Ji Zhang
- Lung Transplantation Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Jingyu Chen
- Lung Transplantation Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Hongbing Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai-Feng Xu
- Department of Pulmonary and Critical Care Medicine and
| |
Collapse
|
17
|
Sun C, Zhang Y, Wang Z, Chen J, Zhang J, Gu Y. TMED2 promotes glioma tumorigenesis by being involved in EGFR recycling transport. Int J Biol Macromol 2024; 262:130055. [PMID: 38354922 DOI: 10.1016/j.ijbiomac.2024.130055] [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: 11/02/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Aberrant epidermal growth factor receptor (EGFR) signaling is the core signaling commonly activated in glioma. The transmembrane emp24 protein transport domain protein 2 (TMED2) interacts with cargo proteins involved in protein sorting and transport between endoplasmic reticulum (ER) and Golgi apparatus. In this study, we found the correlation between TMED2 with glioma progression and EGFR signaling through database analysis. Moreover, we demonstrated that TMED2 is essential for glioma cell proliferation, migration, and invasion at the cellular levels, as well as tumor formation in mouse models, underscoring its significance in the pathobiology of gliomas. Mechanistically, TMED2 was found to enhance EGFR-AKT signaling by facilitating EGFR recycling, thereby providing the initial evidence of TMED2's involvement in the membrane protein recycling process. In summary, our findings shed light on the roles and underlying mechanisms of TMED2 in the regulation of glioma tumorigenesis and EGFR signaling, suggesting that targeting TMED2 could emerge as a promising therapeutic strategy for gliomas and other tumors associated with aberrant EGFR signaling.
Collapse
Affiliation(s)
- Changning Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Yihan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Zhuangzhi Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Jin Chen
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Junhua Zhang
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China.
| |
Collapse
|
18
|
Geng ZH, Du JX, Chen YD, Fu PY, Zhou PH, Qin WZ, Luo YH. YY1: a key regulator inhibits gastric cancer ferroptosis and mediating apatinib-resistance. Cancer Cell Int 2024; 24:71. [PMID: 38347631 PMCID: PMC10863212 DOI: 10.1186/s12935-024-03262-z] [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: 09/12/2023] [Accepted: 02/05/2024] [Indexed: 02/15/2024] Open
Abstract
OBJECTIVE Gastric cancer (GC) stands as a prevalent and deadly global malignancy. Despite its role as a preoperative neoadjuvant therapy, Apatinib's effectiveness is curtailed among GC patients exhibiting elevated YY1 expression. YY1's connection to adverse prognosis, drug resistance, and GC metastasis is established, yet the precise underlying mechanisms remain elusive. This study aims to unravel potential pathogenic pathways attributed to YY1. DESIGN Utilizing bioinformatics analysis, we conducted differentially expressed genes, functional annotation, and pathway enrichment analyses, and further validation through cellular and animal experiments. RESULTS Higher YY1 expression correlated with diminished postoperative progression-free survival (PFS) and disease-specific survival (DSS) rates in TCGA analysis, identifying YY1 as an independent DSS indicator in gastric cancer (GC) patients. Notably, YY1 exhibited significantly elevated expression in tumor tissues compared to adjacent normal tissues. Bioinformatics analysis revealed noteworthy differentially expressed genes (DEGs), transcriptional targets, factors, and co-expressed genes associated with YY1. LASSO Cox analysis unveiled Transferrin as a prospective pivotal protein regulated by YY1, with heightened expression linked to adverse DSS and PFS outcomes. YY1's role in governing the p53 signaling pathway and ferroptosis in GC cells was further elucidated. Moreover, YY1 overexpression dampened immune cell infiltration within GC tumors. Additionally, YY1 overexpression hindered GC cell ferroptosis and mediated Apatinib resistance via the p53 pathway. Remarkably, IFN-a demonstrated efficacy in reversing Apatinib resistance and immune suppression in GC tissues. CONCLUSIONS Our findings underscore the pivotal role of YY1 in driving GC progression and influencing prognosis, thus pinpointing it as a promising therapeutic target to enhance patient outcomes.
Collapse
Affiliation(s)
- Zi-Han Geng
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Shanghai Collaborative Innovation Center of Endoscopy, 200032, Shanghai, China
| | - Jun-Xian Du
- Department of General Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Yue-Da Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), 361004, Xiamen, Fujian, China
| | - Pei-Yao Fu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
- Shanghai Collaborative Innovation Center of Endoscopy, 200032, Shanghai, China
| | - Ping-Hong Zhou
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Shanghai Collaborative Innovation Center of Endoscopy, 200032, Shanghai, China.
| | - Wen-Zheng Qin
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Shanghai Collaborative Innovation Center of Endoscopy, 200032, Shanghai, China.
| | - Yi-Hong Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
19
|
Bapat J, Yamamoto TM, Woodruff ER, Qamar L, Mikeska RG, Aird KM, Watson ZL, Brubaker LW, Bitler BG. CASC4/GOLM2 drives high grade serous carcinoma anoikis resistance through the recycling of EGFR. Cancer Gene Ther 2024; 31:300-310. [PMID: 38030811 PMCID: PMC10874890 DOI: 10.1038/s41417-023-00703-1] [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: 05/31/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
Ovarian cancer is the deadliest gynecological malignancy, and accounts for over 150,000 deaths per year worldwide. The high grade serous ovarian carcinoma (HGSC) subtype accounts for almost 70% of ovarian cancers and is the deadliest. HGSC originates in the fimbria of the fallopian tube and disseminates through the peritoneal cavity. HGSC survival in peritoneal fluid requires cells to resist anoikis (anchorage-independent apoptosis). Most anoikis resistant mechanisms are dependent on microenvironment interactions with cell surface-associated proteins, such as integrins and receptor tyrosine kinases (RTKs). We previously identified the gene CASC4 as a driver of anoikis resistance. CASC4 is predicted to be a Golgi-associated protein that may regulate protein trafficking to the plasma membrane, but CASC4 is largely uncharacterized in literature; thus, we sought to determine how CASC4 confers anoikis resistance to HGSC cells. Mining of publicly available ovarian cancer datasets (TCGA) showed that CASC4 is associated with worse overall survival and increased resistance to platinum-based chemotherapies. For experiments, we cultured three human HGSC cell lines (PEO1, CaOV3, OVCAR3), and a murine HGSC cell line, (ID8) with shRNA-mediated CASC4 knockdowns (CASC4 KD) in suspension, to recapitulate the peritoneal fluid environment in vitro. CASC4 KD significantly inhibited cell proliferation and colony formation ability, and increased apoptosis. A Reverse Phase Protein Assay (RPPA) showed that CASC4 KD resulted in a broad re-programming of membrane-associated proteins. Specifically, CASC4 KD led to decreased protein levels of the RTK Epidermal Growth Factor Receptor (EGFR), an initiator of several oncogenic signaling pathways, leading us to hypothesize that CASC4 drives HGSC survival through mediating recycling and trafficking of EGFR. Indeed, loss of CASC4 led to a decrease in both EGFR membrane localization, reduced turnover of EGFR, and increased EGFR ubiquitination. Moreover, a syngeneic ID8 murine model of ovarian cancer showed that knocking down CASC4 leads to decreased tumor burden and dissemination.
Collapse
Affiliation(s)
- Jaidev Bapat
- Cancer Biology Graduate Program, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tomomi M Yamamoto
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth R Woodruff
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lubna Qamar
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Railey G Mikeska
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine M Aird
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zachary L Watson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lindsay W Brubaker
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin G Bitler
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Xing Y, Gao Z, Bai Y, Wang W, Chen C, Zheng Y, Meng Y. Golgi Protein 73 Promotes LPS-Induced Cardiac Dysfunction via Mediating Myocardial Apoptosis and Autophagy. J Cardiovasc Pharmacol 2024; 83:116-125. [PMID: 37755435 DOI: 10.1097/fjc.0000000000001487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/02/2023] [Indexed: 09/28/2023]
Abstract
ABSTRACT Sepsis-induced cardiac dysfunction represents a major cause of high mortality in intensive care units with limited therapeutic options. Golgi protein 73 (GP73) has been implicated in various diseases. However, the role of GP73 in lipopolysaccharide (LPS)-induced cardiac dysfunction is unclear. In this study, we established a sepsis-induced cardiac dysfunction model by LPS administration in wild-type and GP73 knockout ( GP73-/- ) mice. We found that GP73 was increased in LPS-treated mouse hearts and LPS-cultured neonatal rat cardiomyocytes (NRCMs). Knockout of GP73 alleviated myocardial injury and improved cardiac dysfunction. Moreover, depletion of GP73 in NRCMs relieved LPS-induced cardiomyocyte apoptosis and activated myocardial autophagy. Therefore, GP73 is a negative regulator in LPS-induced cardiac dysfunction by promoting cardiomyocyte apoptosis and inhibiting cardiomyocyte autophagy.
Collapse
Affiliation(s)
- Yaqi Xing
- Department of Pathology, Capital Medical University, Beijing, China
| | - Zhenqiang Gao
- Department of Pathology, Capital Medical University, Beijing, China
| | - Yunfei Bai
- Department of Pathology, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Pathology, Capital Medical University, Beijing, China
- National Demonstration Center for Experimental Basic Medical Education, Experimental Teaching Center of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen Chen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China; and
| | - Yuanyuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing, China
| | - Yan Meng
- Department of Pathology, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Tian B, Pang Y, Gao Y, Meng Q, Xin L, Sun C, Tang X, Wang Y, Li Z, Lin H, Wang L. A pan-cancer analysis of the oncogenic role of Golgi transport 1B in human tumors. J Transl Int Med 2023; 11:433-448. [PMID: 38130634 PMCID: PMC10732491 DOI: 10.2478/jtim-2023-0002] [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] [Indexed: 12/23/2023] Open
Abstract
Background Owing to the aggressiveness and treatment-refractory nature of cancer, ideal candidates for early diagnosis and treatment are needed. Golgi transport 1B (GOLT1B) has been associated with cellular malignant behaviors and immune responses in colorectal and lung cancer, but a systematic pan-cancer analysis on GOLT1B has not been conducted. Methods The expression status and clinical association of GOLT1B in The Cancer Genome Atlas (TCGA) were analyzed. Genetic and methylation alterations in GOLT1B were explored. The relationship between GOLT1B and immune cell infiltration was also investigated. Genes related to GOLT1B expression were selected and analyzed. Results GOLT1B was highly expressed in most tumors, and there was a positive correlation between GOLT1B expression and clinical pathological parameters. High expression levels of GOLT1B have been associated with poor prognosis of most cancers. Copy number amplification was the primary type of GOLT1B genetic alterations, which was related to the prognosis of pan-cancer cases. There were different levels of GOLT1B promoter methylation across cancer types. The methylation level of the probe cg07371838 and cg25816357 was closely associated with prognosis in diverse cancers. There was also a positive correlation between GOLT1B genetic alterations and CD4+ T lymphocytes, especially the Th2 subset, as well as between GOLT1B expression and the estimated infiltration value of cancer-associated fibroblasts. Serine/threonine kinase receptor-associated protein (STRAP), integrator complex subunit 13 (INTS13), and ethanolamine kinase 1 (ETNK1) were the most relevant genes for GOLT1B expression, and their interactions with GOLT1B were involved in regulating the transforming growth factor (TGF)-β receptor signaling pathway and epithelial-mesenchymal transition (EMT). Conclusions This pan-cancer analysis provided a comprehensive understanding of the oncogenic role of GOLT1B, highlighting a potential mechanism whereby GOLT1B influences the tumor microenvironment, as well as cancer immunotherapy.
Collapse
Affiliation(s)
- Bo Tian
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Yanan Pang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai200433, China
| | - Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Qianqian Meng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Xin Tang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Yilin Wang
- Georgetown Preparatory School, North Bethesda20852, MD, USA
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| | - Luowei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai200433, China
| |
Collapse
|
22
|
Sweed D, Gammal SSE, Kilany S, Abdelsattar S, Elhamed SMA. The expression of VEGF and cyclin D1/EGFR in common primary liver carcinomas in Egypt: an immunohistochemical study. Ecancermedicalscience 2023; 17:1641. [PMID: 38414954 PMCID: PMC10898887 DOI: 10.3332/ecancer.2023.1641] [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: 08/28/2023] [Indexed: 02/29/2024] Open
Abstract
Background The most common types of primary malignant liver tumours are hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Treatment options for patients who are inoperable/advanced, or recurring are challenging. Cyclin D1, epidermal growth factor (EGFR) and vascular endothelial growth factor (VEGR) are common carcinogenic proteins that have potential therapeutic targets in various cancers. They have been implicated in the development of HCC and CCA. In this study, we aimed to evaluate the oncogenic function expression of cyclin D1, EGFR and VEGF in HCC and CCA of Egyptian patients. This could help to validate their therapeutic potential. Material and methods Tumour cases were selected from 82 cases of primary liver carcinomas, with 58 cases being from HCC and 24 cases from CCA compared to 51 non-tumour adjacent liver cases and 18 from normal liver tissue. The immunohistochemical study of cyclin D1, EGFR and VEGR was conducted. Results Cyclin D1, EGFR and VEGF are overexpressed in HCC and CCA as compared to the control group (p < 0.001). Cyclin D1 was related to well-differentiated grade and early pathologic stage in HCC (p = 0.016 and p = 0.042, respectively). The well-differentiated grade showed significantly higher VEGF levels (p = 0.04). In the CCA group, however, EGFR was strongly related to high tumour size (p = 0.047). EGFR and VEGF were overexpressed in HCC raised in the non-cirrhotic liver compared to those developed in post-hepatitic liver cirrhosis (p = 0.003 and p = 0.014). Conclusion Cyclin D1, EGFR and VEGF shared significant overexpression in HCC and CCA. EGFR and VEGF may play an oncogenic function in the development of HCC in non-cirrhotic liver. Furthermore, cyclin D1 and VEGF may play a good prognostic function in HCC, but EGFR may play a bad prognostic role in CCA.
Collapse
Affiliation(s)
- Dina Sweed
- Pathology Department, National Liver Institute, Shebin Elkom, Menofia University, Shebin Elkom 32511, Menoufia, Egypt
- https://orcid.org/0000-0001-6483-5056
| | - Shaymaa Sabry El Gammal
- Pathology Department, National Liver Institute, Shebin Elkom, Menofia University, Shebin Elkom 32511, Menoufia, Egypt
| | - Shimaa Kilany
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebin Elkom 32511, Menoufia, Egypt
| | - Shimaa Abdelsattar
- Clinical Biochemistry and Molecular Diagnostics Department, National Liver Institute, Menoufia University, Shebin Elkom 32511, Menoufia, Egypt
| | - Sara Mohamed Abd Elhamed
- Pathology Department, National Liver Institute, Shebin Elkom, Menofia University, Shebin Elkom 32511, Menoufia, Egypt
- https://orcid.org/0000-0003-0526-2627
| |
Collapse
|
23
|
Yu WM, Li GW, Lou MG, Wu ZY. A meta-analysis of the prognostic impact of tissue golgi protein 73 (tGP73) in hepatocellular carcinoma. BMC Gastroenterol 2023; 23:401. [PMID: 37978447 PMCID: PMC10656938 DOI: 10.1186/s12876-023-03050-5] [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: 04/13/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
INTRODUCTION To date, an increasing number of studies have revealed that GP73 may have prognostic value in liver cancer. However, most of the studies evaluated serum GP73, and the results regarding the prognostic value of tGP73 in liver cancer are still controversial. Therefore, in this meta-analysis, we aimed to determine whether tGP73 has any prognostic value in patients with HCC. MATERIALS AND METHODS Relevant publications were searched for in PubMed, EMBASE, OVID, the Cochrane Library, and the Web of Science databases up to March 2023. The hazard ratio (HR) or odds ratio (OR) with corresponding 95% confidence intervals (95% CIs) of eligible studies were assessed by fixed-effects or random-effects models. In addition, subgroup analyses were conducted to investigate the possible causes of heterogeneity, and publication bias analysis was also performed to assess the reliability of the meta-analysis results. RESULTS A total of 10 studies were included. These studies included 1569 HCC patients, and a meta-analysis was performed. The results of our meta-analysis showed that higher GP73 expression levels were significantly associated with poorer OS (HR = 1.87, 95% CI: 1.41-2.48, P < 0.0001, I2 = 58%). However, there was no significant correlation between high GP73 expression and disease-free survival (DFS) (HR: 1.43, 95% CI: 0.93-2.33, P = 0.100). In addition, abnormal GP73 expression was also related to higher tumour tissue differentiation grade (OR = 3.03, 95% CI = 2.01-4.57, P < 0.0001, I2 = 89%), later tumour stage (OR = 5.89, 95% CI = 2.31-14.99, P < 0.0001, I2 = 0%), vascular invasion (OR = 1.72, 95% CI = 1.12-2.64, P = 0.010, I2 = 0%), multiple tumours (OR = 2.44, 95% CI = 1.37-3.68, P = 0.001, I2 = 44%) and early postoperative tumour recurrence (OR = 1.92, 95% CI = 1.10-3.28, P = 0.020, I2 = 62%). CONCLUSIONS The meta-analysis showed that the overexpression of GP73 may be related to a poor prognosis of HCC, and it may also have a predictive effect on the invasion and metastasis of HCC.
Collapse
Affiliation(s)
- Wei-Ming Yu
- Department of Hepatobiliary and pancreatic surgery, The First People's Hospital of Fuyang District, Fuyang First Hospital Affiliated to Binjiang College of Zhejiang Chinese Medicine University, Hangzhou, China.
| | - Guo-Wei Li
- Department of Hepatobiliary and pancreatic surgery, The First People's Hospital of Fuyang District, Fuyang First Hospital Affiliated to Binjiang College of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Ming-Geng Lou
- Department of Hepatobiliary and pancreatic surgery, The First People's Hospital of Fuyang District, Fuyang First Hospital Affiliated to Binjiang College of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Zheng-Yu Wu
- Department of Hepatobiliary and pancreatic surgery, The First People's Hospital of Fuyang District, Fuyang First Hospital Affiliated to Binjiang College of Zhejiang Chinese Medicine University, Hangzhou, China
| |
Collapse
|
24
|
Fan Y, Wang L, Han X, Ma H, Zhang N, She L. LncRNA ASB16-AS1 accelerates cellular process and chemoresistance of ovarian cancer cells by regulating GOLM1 expression via targeting miR-3918. Biochem Biophys Res Commun 2023; 675:1-9. [PMID: 37429067 DOI: 10.1016/j.bbrc.2023.06.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND Reportedly, ovarian cancer (OC) is a major threat to women's health. Long non-coding RNA (lncRNA) ASB16-AS1 has been uncovered to participate in cancer progression. Nevertheless, the role of ASB16-AS1 in OC remains to be revealed. PURPOSE This study aimed to unveil the biological function of ASB16-AS1 and its underlying mechanisms in OC cells. METHODS QRT-PCR was done to test ASB16-AS1 expression in OC cells. Functional assays were performed to evaluate the malignant behaviors and cisplatin resistance of OC cells. Mechanistic analyses were done to investigate the regulatory molecular mechanism in OC cells. RESULTS ASB16-AS1 was found to be highly expressed in OC cells. ASB16-AS1 knockdown repressed proliferation, migration, and invasion of OC cells, while facilitating cell apoptosis. ASB16-AS1 was further validated to up-regulate GOLM1 through competitively binding with miR-3918. Moreover, miR-3918 overexpression was corroborated to suppress OC cell growth. Rescue assays further uncovered that ASB16-AS1 modulated the malignant processes of OC cells via targeting miR-3918/GOLM1 axis. CONCLUSION ASB16-AS1 facilitates the malignant processes and chemoresistance of OC cells via serving as miR-3918 sponge and positively modulating GOLM1 expression.
Collapse
Affiliation(s)
- Yang Fan
- Department of Gynaecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China.
| | - Long Wang
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xuechuang Han
- Department of Gynaecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Hongyun Ma
- Department of Gynaecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Na Zhang
- Department of Gynaecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Lina She
- Department of Gynaecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| |
Collapse
|
25
|
Asghariazar V, Kadkhodayi M, Sarailoo M, Jolfayi AG, Baradaran B. MicroRNA-143 as a potential tumor suppressor in cancer: An insight into molecular targets and signaling pathways. Pathol Res Pract 2023; 250:154792. [PMID: 37689002 DOI: 10.1016/j.prp.2023.154792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/25/2023] [Accepted: 09/02/2023] [Indexed: 09/11/2023]
Abstract
MicroRNAs (MiRNAs), which are highly conserved and small noncoding RNAs, negatively regulate gene expression and influence signaling pathways involved in essential biological activities, including cell proliferation, differentiation, apoptosis, and cell invasion. MiRNAs have received much attention in the past decade due to their significant roles in cancer development. In particular, microRNA-143 (miR-143) is recognized as a tumor suppressor and is downregulated in most cancers. However, it seems that miR-143 is upregulated in rare cases, such as prostate cancer stem cells, and acts as an oncogene. The present review will outline the current studies illustrating the impact of miR-143 expression levels on cancer progression and discuss its target genes and their relevant signaling pathways to discover a potential therapeutic way for cancer.
Collapse
Affiliation(s)
- Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Deputy of Research and Technology, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mahtab Kadkhodayi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Animal Biology, Faculty of Natural Sciences, The University of Tabriz, Tabriz, Iran
| | - Mehdi Sarailoo
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amir Ghaffari Jolfayi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Frans MT, Kuipers EM, Bianchi F, van den Bogaart G. Unveiling the impact of GOLM1/GP73 on cytokine production in cancer and infectious disease. Immunol Cell Biol 2023; 101:727-734. [PMID: 37332154 DOI: 10.1111/imcb.12664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
The Golgi membrane protein GOLM1/GP73/GOLPH2 has been found to impact cytokine production in both infectious disease and cancer. In viral infections, GOLM1 levels are increased, and this lowers the production of type I interferons and other inflammatory cytokines. However, elevated GOLM1 expression levels due to mutations are linked to a higher production of interleukin (IL)-6 during Candida infections, potentially explaining an increased susceptibility to candidemia in individuals carrying these mutations. In cancer, the protease Furin produces a soluble form of GOLM1 that has oncogenic properties by promoting the production of the chemokine CCL2 and suppressing the production of inflammatory cytokines such as IL-12 and interferon gamma. This review will focus on the role of GOLM1 in cytokine production, highlighting how it can both promote and inhibit cytokine production. It is crucial to understand this in order to effectively target GOLM1 for therapeutic purposes in diseases associated with abnormal cytokine production, including cancer and infectious disease.
Collapse
Affiliation(s)
- Myrthe T Frans
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Ella M Kuipers
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Frans Bianchi
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
27
|
Hu X, Yuan S, Zhou S, Sun T, Wang C, Ying S, Zhu H, Luo J, Jin H, Liu Y. Golgi-protein 73 facilitates vimentin polymerization in hepatocellular carcinoma. Int J Biol Sci 2023; 19:3694-3708. [PMID: 37564210 PMCID: PMC10411459 DOI: 10.7150/ijbs.85431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/08/2023] [Indexed: 08/12/2023] Open
Abstract
Golgi-protein 73 (GP73) is highly expressed in hepatocellular carcinoma (HCC) and, as a secretory protein, it has been proposed as a serum biomarker indicating progression of HCC. The underlying mechanism by which GP73 may promote HCC metastasis is still poorly understood. In this study, we discovered that GP73 interacted with vimentin to facilitate Serine/Threonine-protein phosphatase PP1-alpha (PP1A)-mediated dephosphorylation of vimentin at S56 and facilitated vimentin polymerization, which blocked vimentin degradation via TRIM56-mediated ubiquitin/proteasome-dependent pathway. Strikingly, Clomipramine, a 5-hydroxytryptamine receptor (5-HTR) agonist approved for the treatment of depression, impaired GP73-mediated vimentin polymerization to effectively inhibit metastasis of HCC with high GP73 expression, which provided a new strategy against HCC metastasis. Lastly, it was found that serum GP73 (sGP73) correlated positively with vimentin in primary tissues of HCC, suggesting that sGP73 might serve as a potential serum biomarker for companion diagnosis of HCC with highly expressed vimentin. In summary, this study reveals the process of GP73-mediated vimentin polymerization and proves that Clomipramine serves as a potential drug targeting vimentin for metastatic HCC patients with high sGP73 level.
Collapse
Affiliation(s)
- Xinyang Hu
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Shijin Yuan
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Sining Zhou
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ting Sun
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Chaoqun Wang
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang 322100, China
| | - Shilong Ying
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Heping Zhu
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Jingfeng Luo
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Yiming Liu
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
28
|
Zheng Y, Zhu L, Qin ZY, Guo Y, Wang S, Xue M, Shen KY, Hu BY, Wang XF, Wang CQ, Qin LX, Dong QZ. Modulation of cellular metabolism by protein crotonylation regulates pancreatic cancer progression. Cell Rep 2023; 42:112666. [PMID: 37347667 DOI: 10.1016/j.celrep.2023.112666] [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: 07/06/2022] [Revised: 02/10/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Protein lysine crotonylation has been recently identified as a vital posttranslational modification in cellular processes, particularly through the modification of histones. We show that lysine crotonylation is an important modification of the cytoplastic and mitochondria proteins. Enzymes in glycolysis, the tricarboxylic acid (TCA) cycle, fatty acid metabolism, glutamine metabolism, glutathione metabolism, the urea cycle, one-carbon metabolism, and mitochondrial fusion/fission dynamics are found to be extensively crotonylated in pancreatic cancer cells. This modulation is mainly controlled by a pair of crotonylation writers and erasers including CBP/p300, HDAC1, and HDAC3. The dynamic crotonylation of metabolic enzymes is involved in metabolism regulation, which is linked with tumor progression. Interestingly, the activation of MTHFD1 by decrotonylation at Lys354 and Lys553 promotes the development of pancreatic cancer by increasing resistance to ferroptosis. Our study suggests that crotonylation represents a metabolic regulatory mechanism in pancreatic cancer progression.
Collapse
Affiliation(s)
- Yan Zheng
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhao-Yu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Yu Guo
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Min Xue
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Ke-Yu Shen
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Xu-Feng Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Chao-Qun Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China; Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
29
|
Cho WC, Wong CF, Li KP, Fong AH, Fung KY, Au JS. miR-145 as a Potential Biomarker and Therapeutic Target in Patients with Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:10022. [PMID: 37373169 DOI: 10.3390/ijms241210022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Our previous study found that miR-145 was downregulated in non-small cell lung cancer (NSCLC) tissues and that it could inhibit the cell proliferation in transfected NSCLC cells. In this study, we found that miR-145 was downregulated in NSCLC plasma samples compared to healthy controls. A receiver operating characteristic curve analysis indicated that plasma miR-145 expression was correlated with NSCLC in patient samples. We further revealed that the transfection of miR-145 inhibited the proliferation, migration, and invasion of NSCLC cells. Most importantly, miR-145 significantly delayed the tumor growth in a mouse model of NSCLC. We further identified GOLM1 and RTKN as the direct targets of miR-145. A cohort of paired tumors and adjacent non-malignant lung tissues from NSCLC patients was used to confirm the downregulated expression and diagnostic value of miR-145. The results were highly consistent between our plasma and tissue cohorts, confirming the clinical value of miR-145 in different sample groups. In addition, we also validated the expressions of miR-145, GOLM1, and RTKN using the TCGA database. Our findings suggested that miR-145 is a regulator of NSCLC and it plays an important role in NSCLC progression. This microRNA and its gene targets may serve as potential biomarkers and novel molecular therapeutic targets in NSCLC patients.
Collapse
Affiliation(s)
- William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Chi F Wong
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Kwan P Li
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Alvin H Fong
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - King Y Fung
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Joseph S Au
- Oncology Center, Hong Kong Adventist Hospital, Hong Kong SAR, China
| |
Collapse
|
30
|
Liu B, Cao J, Wu B, Hao K, Wang X, Chen X, Shen Z. METTL3 and STAT3 form a positive feedback loop to promote cell metastasis in hepatocellular carcinoma. Cell Commun Signal 2023; 21:121. [PMID: 37231451 DOI: 10.1186/s12964-023-01148-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND It is well-established that most Hepatocellular carcinoma (HCC) patients die of metastasis, yet the potential mechanisms orchestrating metastasis remain poorly understood. Current evidence suggests that the dysregulation of METTL3-mediated m6A methylation modification is closely associated with cancer progression. STAT3 is an oncogenic transcription factor that reportedly plays a central role in the occurrence and development of HCC. However, the relationship between METTL3 and STAT3 in HCC metastasis remains unclear. METHODS The relationship between METTL3 expression and the survival of HCC patients was assessed by online tools GEPIA and Kaplan-Meier Plotter. Western blotting, Tissue microarray (TMA), and immunohistochemistry (IHC) staining were used to evaluate the expression levels of METTL3 and STAT3 in HCC cell lines and metastatic and non-metastatic tissues. Methylated RNA immunoprecipitation (MeRIP), MeRIP sequencing (MeRIP-seq), qRT-PCR, RNA immunoprecipitation (RIP), Western blotting and luciferase reporter gene assay were utilized to clarify the mechanism of METTL3 regulating STAT3 expression. Immunofluorescence staining, Western blotting, qRT-PCR, Co-immunoprecipitation (Co-IP), IHC staining, TMA and Chromatin immunoprecipitation (ChIP) assay were performed to explore the mechanism of STAT3 modulating METTL3 localization. Cell viability, wound healing and transwell assay, and orthotopic xenograft model were used to evaluate the role of METTL3-STAT3 feedback loop in the promotion of HCC metastasis in vitro and in vivo. RESULTS METTL3 and STAT3 are both abundantly expressed in high-metastatic HCC cells and tissues. Moreover, a positive correlation was found between the expression of STAT3 and METTL3 in HCC tissues. Mechanistically, METTL3 could induce the m6A modification of STAT3 mRNA, and then promote the translation of m6A-contained STAT3 mRNA by interacting with the translation initiation machinery. In contrast, STAT3 promoted nuclear localization of METTL3 via transcriptionally upregulating WTAP, a vital member of the methyltransferase complex, and facilitated the methyltransferase function of METTL3. METTL3 and STAT3 form a positive feedback loop to accelerate HCC metastasis in vitro and in vivo. CONCLUSIONS Our findings reveal a novel mechanism of HCC metastasis and uncover the METTL3-STAT3 feedback signaling as a potential target for the anti-metastatic treatment of HCC. Video Abstract.
Collapse
Affiliation(s)
- Bowen Liu
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China.
| | - Jinling Cao
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Biting Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Kaixuan Hao
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiangyun Wang
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Xin Chen
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Zhifa Shen
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China.
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
31
|
Sun L, Liu Z, Wu Z, Ning K, Hu J, Chen Z, Wu Z, Yin X. Molecular subtype identification and signature construction based on Golgi apparatus-related genes for better prediction prognosis and immunotherapy response in hepatocellular carcinoma. Front Immunol 2023; 14:1113455. [PMID: 37051238 PMCID: PMC10083374 DOI: 10.3389/fimmu.2023.1113455] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/15/2023] [Indexed: 03/28/2023] Open
Abstract
IntroductionThe Golgi apparatus (GA) is the center of protein and lipid synthesis and modification in normal cells and is involved in regulating various cellular process as a signaling hub, the dysfunction of which can lead to the development of various pathological conditions, including tumors. Mutations in Golgi apparatus-related genes (GARGs) are prevalent in most tumors, and their mutations can make them pro-tumor metastatic. The aim of this study was to analyze the predictive role of GARGs in the prognosis and immunotherapeutic outcome of hepatocellular carcinoma.MethodsWe used TCGA, GEO and ICGC databases to classify hepatocellular carcinoma samples into two molecular subtypes based on the expression of GARGs. Signature construction was then performed using GARGs, and signature genes were selected for expression validation and tumor phenotype experiments to determine the role of GARGs in the prognosis of hepatocellular carcinoma.ResultsUsing the TCGA, GEO and ICGC databases, two major subtypes of molecular heterogeneity among hepatocellular carcinoma tumors were identified based on the expression of GARGs, C1 as a high-risk subtype (low survival) and C2 as a low-risk subtype (high survival). The high-risk subtype had lower StromalScore, ImmuneScore, ESTIMATEScore and higher TumorPurity, indicating poorer treatment outcome for ICI. Meanwhile, we constructed a new risk assessment profile for hepatocellular carcinoma based on GARGs, and we found that the high-risk group had a worse prognosis, a higher risk of immune escape, and a higher TP53 mutation rate. Meanwhile, TME analysis showed higher tumor purity TumorPurity and lower ESTIMATEScore, ImmuneScore and StromalScore in the high-risk group. We also found that the high-risk group responded more strongly to a variety of anticancer drugs, which is useful for guiding clinical drug use. Meanwhile, the expression of BSG was experimentally found to be associated with poor prognosis of HCC. After interfering with the expression of BSG in HCC cells SMMC-7721, the proliferation and migration ability of HCC cells were significantly restricted.DiscussionThe signature we constructed using GARGs can well predict the prognosis and immunotherapy effect of hepatocellular carcinoma, providing new ideas and strategies for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Liang Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zitao Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhengyi Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ke Ning
- Department of Emergency, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junwen Hu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhendong Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhipeng Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiangbao Yin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xiangbao Yin,
| |
Collapse
|
32
|
Yu J, Feng H, Sang Q, Li F, Chen M, Yu B, Xu Z, Pan T, Wu X, Hou J, Zhu Z, Yan C, Su L, Li J, Liu B. VPS35 promotes cell proliferation via EGFR recycling and enhances EGFR inhibitors response in gastric cancer. EBioMedicine 2023; 89:104451. [PMID: 36738481 PMCID: PMC9931929 DOI: 10.1016/j.ebiom.2023.104451] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Vacuolar protein sorting-associated protein 35 (VPS35) is a core component of the retromer complex which mediates intracellular protein transport. It is well known that dysfunctional VPS35 functions in the accumulation of pathogenic proteins. In our previous study, VPS35 was found to be a potential gene related to poor prognosis in gastric cancer. However, the biological functions of VPS35 in gastric cancer remain unclear. METHODS Cell viability assays were performed to examine whether VPS35 affected cell proliferation. Immunoprecipitation and biotin assays showed that VPS35 bound to epidermal growth factor receptor (EGFR) in the cytoplasm and recycled it to the cell surface. Patient-derived xenografts and organoids were used to evaluate the effect of VPS35 on the response of gastric cancer to EGFR inhibitors. FINDINGS VPS35 expression levels were upregulated in tumour tissues and correlated with local tumour invasion and poor survival in patients with gastric cancer. VPS35 promoted cell proliferation and increased tumour growth. Mechanistically, VPS35 selectively bound to endocytosed EGFR in early endosomes and recycled it back to the cell surface, leading to the downstream activation of the ERK1/2 pathway. We also found that high VPS35 expression levels increased the sensitivity of the xenograft and organoid models to EGFR inhibitors. INTERPRETATION VPS35 promotes cell proliferation by recycling EGFR to the cell surface, amplifying the network of receptor trafficking. VPS35 expression levels are positively correlated with gastric cancer sensitivity to EGFR inhibitors, which offers a potential method to stratify patients for EGFR inhibitor utilisation. FUNDING National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Junxian Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Qingqing Sang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Fangyuan Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Mengdi Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Beiqin Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Zhuoqing Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Tao Pan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Xiongyan Wu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Junyi Hou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Chao Yan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Liping Su
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Jianfang Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China.
| | - Bingya Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China.
| |
Collapse
|
33
|
PELI1 and EGFR cooperate to promote breast cancer metastasis. Oncogenesis 2023; 12:9. [PMID: 36841821 PMCID: PMC9968314 DOI: 10.1038/s41389-023-00457-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/27/2023] Open
Abstract
Pellino-1 (PELI1) is an E3 ubiquitin ligase acting as a key regulator for the inflammation and autoimmunity via the ubiquitination of the substrate proteins. There is increasing evidence to support that PELI1 functions as an oncoprotein in tumorigenesis and metastasis. However, the molecular mechanism underlying the high expression and oncogenic roles of PELI1 in cancers remains limited. Herein, we revealed a novel regulation mechanism by which PELI1 and EGFR cooperate to promote breast cancer metastasis. EGFR is positively correlated with PELI1 expression in breast cancers, and its activation led to the phosphorylation of PELI1 at Tyr154 and Thr264, which subsequently activated its E3 ubiquitin ligase. Simultaneously, PELI1 physically interacted with and enhanced the stability of EGFR via the K63-linked polyubiquitination in reverse. The co-inhibition of the PELI1-EGFR showed synergetic effect to repress breast cancer metastasis. Furthermore, we identified a compound S62 as a small molecule disruptor of PELI1/EGFR that effectively repressed breast cancer metastasis. Our study not only uncovered the emerging roles of PELI1/EGFR interaction in the progression of breast cancer, but also provided an effective strategy for the inhibition of metastasis in breast cancer.
Collapse
|
34
|
Liu X, Hu Y, Xue Z, Zhang X, Liu X, Liu G, Wen M, Chen A, Huang B, Li X, Yang N, Wang J. Valtrate, an iridoid compound in Valeriana, elicits anti-glioblastoma activity through inhibition of the PDGFRA/MEK/ERK signaling pathway. J Transl Med 2023; 21:147. [PMID: 36829235 PMCID: PMC9960449 DOI: 10.1186/s12967-023-03984-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Valtrate, a natural compound isolated from the root of Valeriana, exhibits antitumor activity in many cancers through different mechanisms. However, its efficacy for the treatment of glioblastoma (GBM), a tumor type with a poor prognosis, has not yet been rigorously investigated. METHODS GBM cell lines were treated with valtrate and CCK-8, colony formation and EdU assays, flow cytometry, and transwell, 3D tumor spheroid invasion and GBM-brain organoid co-culture invasion assays were performed to assess properties of proliferation, viability, apoptosis and invasion/migration. RNA sequencing analysis on valtrate-treated cells was performed to identify putative target genes underlying the antitumor activity of the drug in GBM cells. Western blot analysis, immunofluorescence and immunohistochemistry were performed to evaluate protein levels in valtrate-treated cell lines and in samples obtained from orthotopic xenografts. A specific activator of extracellular signal-regulated kinase (ERK) was used to identify the pathways mediating the effect. RESULTS Valtrate significantly inhibited the proliferation of GBM cells in vitro by inducing mitochondrial apoptosis and suppressed invasion and migration of GBM cells by inhibiting levels of proteins associated with epithelial mesenchymal transition (EMT). RNA sequencing analysis of valtrate-treated GBM cells revealed platelet-derived growth factor receptor A (PDGFRA) as a potential target downregulated by the drug. Analysis of PDGFRA protein and downstream mediators demonstrated that valtrate inhibited PDGFRA/MEK/ERK signaling. Finally, treatment of tumor-bearing nude mice with valtrate led to decreased tumor volume (fivefold difference at day 28) and enhanced survival (day 27 vs day 36, control vs valtrate-treated) relative to controls. CONCLUSIONS Taken together, our study demonstrated that the natural product valtrate elicits antitumor activity in GBM cells through targeting PDGFRA and thus provides a candidate therapeutic compound for the treatment of GBM.
Collapse
Affiliation(s)
- Xuemeng Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Yaotian Hu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Zhiyi Xue
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xun Zhang
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xiaofei Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Guowei Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Muzi Wen
- grid.284723.80000 0000 8877 7471School of Public Health, Southern Medical University, Foushan, 528000 China
| | - Anjing Chen
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Bin Huang
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xingang Li
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Ning Yang
- Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012, China. .,Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117, China. .,Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, 250012, China.
| | - Jian Wang
- Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012, China. .,Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117, China. .,Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway.
| |
Collapse
|
35
|
Shahini E, Pasculli G, Solimando AG, Tiribelli C, Cozzolongo R, Giannelli G. Updating the Clinical Application of Blood Biomarkers and Their Algorithms in the Diagnosis and Surveillance of Hepatocellular Carcinoma: A Critical Review. Int J Mol Sci 2023; 24:ijms24054286. [PMID: 36901717 PMCID: PMC10001986 DOI: 10.3390/ijms24054286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
The most common primary liver cancer is hepatocellular carcinoma (HCC), and its mortality rate is increasing globally. The overall 5-year survival of patients with liver cancer is currently 10-20%. Moreover, because early diagnosis can significantly improve prognosis, which is highly correlated with tumor stage, early detection of HCC is critical. International guidelines advise using α-FP biomarker with/without ultrasonography for HCC surveillance in patients with advanced liver disease. However, traditional biomarkers are sub-optimal for risk stratification of HCC development in high-risk populations, early diagnosis, prognostication, and treatment response prediction. Since about 20% of HCCs do not produce α-FP due to its biological diversity, combining α-FP with novel biomarkers can enhance HCC detection sensitivity. There is a chance to offer promising cancer management methods in high-risk populations by utilizing HCC screening strategies derived from new tumor biomarkers and prognostic scores created by combining biomarkers with distinct clinical parameters. Despite numerous efforts to identify molecules as potential biomarkers, there is no single ideal marker in HCC. When combined with other clinical parameters, the detection of some biomarkers has higher sensitivity and specificity in comparison with a single biomarker. Therefore, newer biomarkers and models, such as the Lens culinaris agglutinin-reactive fraction of Alpha-fetoprotein (α-FP), α-FP-L3, Des-γ-carboxy-prothrombin (DCP or PIVKA-II), and the GALAD score, are being used more frequently in the diagnosis and prognosis of HCC. Notably, the GALAD algorithm was effective in HCC prevention, particularly for cirrhotic patients, regardless of the cause of their liver disease. Although the role of these biomarkers in surveillance is still being researched, they may provide a more practical alternative to traditional imaging-based surveillance. Finally, looking for new diagnostic/surveillance tools may help improve patients' survival. This review discusses the current roles of the most used biomarkers and prognostic scores that may aid in the clinical management of HCC patients.
Collapse
Affiliation(s)
- Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy
- Correspondence: ; Tel.: +39-0804994249
| | - Giuseppe Pasculli
- National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy
| | - Antonio Giovanni Solimando
- Guido Baccelli Unit of Internal Medicine, Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), University of Bari “A. Moro”, 70121 Bari, Italy
| | | | - Raffaele Cozzolongo
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy
| | - Gianluigi Giannelli
- Scientific Director, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy
| |
Collapse
|
36
|
Yang X, Fan X, Feng J, Fan T, Li J, Huang L, Wan L, Yang H, Li H, Gong J, Zhang Y, Gao Q, Zheng F, Xu L, Lin H, Zhang D, Song H, Wang Y, Ma X, Sun Z, Cao C, Yang X, Zhong H, Fang Y, Wei C. GP73 blockade alleviates abnormal glucose homeostasis in diabetic mice. J Mol Endocrinol 2023; 70:JME-22-0103. [PMID: 36394986 DOI: 10.1530/jme-22-0103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Golgi protein 73 (GP73), also called Golgi membrane protein 1 (GOLM1), is a resident Golgi type II transmembrane protein and is considered as a serum marker for the detection of a variety of cancers. A recent work revealed the role of the secreted GP73 in stimulating liver glucose production and systemic glucose homeostasis. Since exaggerated hepatic glucose production plays a key role in the pathogenesis of type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), GP73 may thus represent a potential therapeutic target for treating diabetic patients with pathologically elevated levels. Here, in this study, we found that the circulating GP73 levels were significantly elevated in T2DM and positively correlated with hemoglobin A1c. Notably, the aberrantly upregulated GP73 levels were indispensable for the enhanced protein kinase A signaling pathway associated with diabetes. In diet-induced obese mouse model, GP73 siRNA primarily targeting liver tissue was potently effective in alleviating abnormal glucose metabolism. Ablation of GP73 from whole animals also exerted a profound glucose-lowering effect. Importantly, neutralizing circulating GP73 improved glucose metabolism in streptozotocin (STZ) and high-fat diet/STZ-induced diabetic mice. We thus concluded that GP73 was a feasible therapeutic target for the treatment of diabetes.
Collapse
Affiliation(s)
- Xiaopan Yang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Xiaojing Fan
- Department of Endocrinology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiangyue Feng
- Beijing Sungen Biomedical Technology Co. Ltd., Beijing, China
| | - Tinghui Fan
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Huan Yang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Huilong Li
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Jing Gong
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Yanhong Zhang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Qi Gao
- Beijing Sungen Biomedical Technology Co. Ltd., Beijing, China
| | - Fei Zheng
- Beijing Sungen Biomedical Technology Co. Ltd., Beijing, China
| | - Lei Xu
- Beijing Sungen Biomedical Technology Co. Ltd., Beijing, China
| | - Haotian Lin
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Dandan Zhang
- Department of Laboratory, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Laboratory, General Hospital of Armed Police Forces, Anhui Medical University, Hefei, China
| | - Hongbin Song
- Department of Laboratory, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Laboratory, General Hospital of Armed Police Forces, Anhui Medical University, Hefei, China
| | - Yufei Wang
- Department of Laboratory, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Laboratory, General Hospital of Armed Police Forces, Anhui Medical University, Hefei, China
| | - Xueping Ma
- Department of Laboratory, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Laboratory, General Hospital of Armed Police Forces, Anhui Medical University, Hefei, China
| | - Zhiwei Sun
- Beijing Sungen Biomedical Technology Co. Ltd., Beijing, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Xiaoli Yang
- Department of Laboratory, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Laboratory, General Hospital of Armed Police Forces, Anhui Medical University, Hefei, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Yi Fang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- Department of Endocrinology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Guo Y, Liu B, Huang T, Qi X, Li S. HOTAIR modulates hepatocellular carcinoma progression by activating FUT8/core-fucosylated Hsp90/MUC1/STAT3 feedback loop via JAK1/STAT3 cascade. Dig Liver Dis 2023; 55:113-122. [PMID: 35504805 DOI: 10.1016/j.dld.2022.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/22/2022] [Accepted: 04/13/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Glycosylation exhibits crucial effect on hepatocellular carcinoma (HCC) progression. Long non-coding RNAs (lncRNAs) are involved in multilevel regulation of gene transcription during tumor development. The purpose of this study is to clarify the potential mechanism that HOTAIR modulates hepatocellular carcinoma progression by activating FUT8/core-fucosylated Hsp90/MUC1/STAT3 feedback loop via JAK1/STAT3 cascade. METHODS qRT-PCR was used to show the differential expression of genes. Functional experiments were used to measure the malignancy of HCC cells. ChIP and co-IP assays showed the directly interaction of the key molecules. Xenografts was conducted to show the in vivo effects. RESULTS Upregulation of FUT8 showed closely correlation with HCC progression. Core-fucosylation of Hsp90 stabilized MUC1 binding to the downstream p-STAT3, which involved in the activation of JAK1/STAT3 cascade. STAT3 was identified as the regulator of FUT8 and MUC1 transcription, while FUT8 and MUC1 impacted STAT3 level both in nuclear and cytoplasm. HOTAIR recruited P300 to efficiently bind with STAT3. The transcript complex co-modulated the transcrption of FUT8 and MUC1. Moreover, highly HOTAIR expression also exhibited closely correlation with HCC progression. CONCLUSIONS FUT8 triggered core-fucosylated-Hsp90/MUC1/P300-HOTAIR-STAT3 cascade via JAK1/STAT3 pathway, which exhibited as positive feedback loop during HCC progression.
Collapse
Affiliation(s)
- Yanru Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China
| | - Bing Liu
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Tong Huang
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Xia Qi
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Shijun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China; College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
38
|
Zhang Z, Wu L, Li J, Chen J, Yu Q, Yao H, Xu Y, Liu L. Identification of ZBTB9 as a potential therapeutic target against dysregulation of tumor cells proliferation and a novel biomarker in Liver Hepatocellular Carcinoma. J Transl Med 2022; 20:602. [PMID: 36522647 PMCID: PMC9756481 DOI: 10.1186/s12967-022-03790-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Zinc finger and bric-a-brac/tramtrack/broad (ZBTB) domain-containing proteins have been reported to be associated with many tumors' development. However, in tumor initiation and progression, the role of ZBTB9, one of the protein family, and its prognostic value were yet to be elucidated in Liver Hepatocellular Carcinoma (LIHC). METHODS We used R software and online bioinformatics analysis tools such as GEPIA2, cBioPortal, TIMER2, Metascape, UALCAN, STRING, TISIDB, and COSMIC to investigate ZBTB9's characteristics and function in LIHC, including abnormal expression, carcinogenic role, related signaling pathways and prognostic value. Furthermore, cell experiments (such as formation, wound healing, and transwell assays) and analyses based on clinical samples (such as immunohistochemistry (IHC) and promoter methylation analysis) were conducted to verify pivotal conclusions. RESULTS ZBTB9 was overexpressed in LIHC samples compared to adjacent normal tissues. Through the analysis of genomic alteration and promoter hypomethylation, the clinical value and etiology of abnormal expression of ZBTB9 were preliminarily exlpored. Subsequent evidence showed that it could result in tumor progression and poor prognosis via activating cell cycle, DNA repair, MYC, and KRAS-associated signaling pathways as well as rendering immune dysregulation. After the knockdown of ZBTB9, evidently inhibited capacities of tumor cells proliferation and migration were observed. These results together indicated that ZBTB9 could be a promising prognostic biomarker and had the potential value to offer novel therapeutic targets for LIHC treatment. CONCLUSIONS ZBTB9 was identified as a novel biomarker to predict the prognosis and tumor progression in LIHC, and a promising therapeutic target to invert tumor development.
Collapse
Affiliation(s)
- Zhenshan Zhang
- grid.412532.3Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China ,grid.452404.30000 0004 1808 0942Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 People’s Republic of China ,grid.452404.30000 0004 1808 0942Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Leilei Wu
- grid.412532.3Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juan Li
- grid.452404.30000 0004 1808 0942Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 People’s Republic of China
| | - Jiayan Chen
- grid.452404.30000 0004 1808 0942Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 People’s Republic of China
| | - Qi Yu
- grid.412532.3Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China ,Shanghai Concord Cancer Center, Shanghai, 200240 China
| | - Hui Yao
- grid.490481.0Department of Radiation Oncology, Shanghai International Medical Center, Shanghai, China
| | - Yaping Xu
- grid.412532.3Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Liu
- grid.412532.3Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China ,grid.452404.30000 0004 1808 0942Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 People’s Republic of China ,grid.490481.0Department of Radiation Oncology, Shanghai International Medical Center, Shanghai, China
| |
Collapse
|
39
|
Bongolo CC, Thokerunga E, Yan Q, Yacouba MBM, Wang C. Exosomes Derived from microRNA-27a-3p Overexpressing Mesenchymal Stem Cells Inhibit the Progression of Liver Cancer through Suppression of Golgi Membrane Protein 1. Stem Cells Int 2022; 2022:9748714. [PMID: 36530488 PMCID: PMC9750777 DOI: 10.1155/2022/9748714] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/28/2022] [Accepted: 11/12/2022] [Indexed: 08/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a significant health burden to date. Its early diagnosis and treatment are complicated by the lack of early diagnosis markers and multidrug resistance. microRNA regulation of HCC oncogenes are among the new diagnostic and therapeutic strategies being explored, although the mode of delivery of a therapeutic dose of the miRNA remains a challenge. In this study, we explored the use of exosomes from umbilical mesenchymal stem cells transfected with miR-27a-3p to interact with the oncogene GOLM1 in HCC and inhibit HCC progression both in vitro and in vivo. We first determined and compared the expression levels of miR-27a-3p in blood, various cell lines and tissues of HCC and their corresponding normal controls. We then employed bioinformatics analysis to determine the gene target for miR-27a-3p in HCC and later transfected upregulated miR-27a-3p in mesenchymal stem cells, and treated HCC cells with exosomes extracted from the transfected stem cells. We then created mouse models of HCC using balbc/nude mice and equally treated them with exosomes from miR-27a-3p transfected stem cells. The results showed that miR-27a-3p is downregulated in blood, cell lines, and tissues of HCC patients compared to normal controls. Exosomes from the miR-27a-3p transfected mesenchymal stem cells prevented HCC cell proliferation, invasion, and metastasis both in vitro and in vivo. Upregulation of miR-27a-3p prevented HCC through interacting with and downregulating GOLM1 as its target oncogene. In conclusion, miR-27a-3p is a potential therapeutic target for HCC acting through GOLM1.
Collapse
Affiliation(s)
- Christian Cedric Bongolo
- Wuhan Sheba Precision Medical Technology Co. Ltd., Wuhan, 430022 Hubei, China
- Department & Program of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 43007, China
| | - Erick Thokerunga
- Department & Program of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 43007, China
- Department of Medical Laboratory Science, Faculty of Medicine, Mbarara University of Science and Technology, 1410 Mbarara, Uganda
| | - Qian Yan
- Wuhan Sheba Precision Medical Technology Co. Ltd., Wuhan, 430022 Hubei, China
| | | | - Chao Wang
- Department of General Surgery, Clinical Research Center of Geriatric Diseases in Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
40
|
Elevation of spermine remodels immunosuppressive microenvironment through driving the modification of PD-L1 in hepatocellular carcinoma. Cell Commun Signal 2022; 20:175. [DOI: 10.1186/s12964-022-00981-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Spermine is frequently elevated in tumor tissues and body fluids of cancer patients and is critical for cancer cell proliferation, migration and invasion. However, the immune functions of spermine in hepatocellular carcinoma progression remains unknown. In the present study, we aimed to elucidate immunosuppressive role of spermine in hepatocellular carcinoma and to explore the underlying mechanism.
Methods
Whole-blood spermine concentration was measured using HPLC. Human primary HCC tissues were collected to examine the expression of CaSR, p-Akt, β-catenin, STT3A, PD-L1, and CD8. Mouse model of tumorigenesis and lung metastasis were established to evaluate the effects of spermine on hepatocellular carcinoma. Western blotting, immunofluorescence, real time PCR, digital Ca2+ imaging, and chromatin immunoprecipitation assay were used to investigate the underlying mechanisms by which spermine regulates PD-L1 expression and glycosylation in hepatocellular carcinoma cells.
Results
Blood spermine concentration in the HCC patient group was significantly higher than that in the normal population group. Spermine could facilitate tumor progression through inducing PD-L1 expression and decreasing the CD8+ T cell infiltration in HCC. Mechanistically, spermine activates calcium-sensing receptor (CaSR) to trigger Ca2+ entry and thereby promote Akt-dependent β-catenin stabilization and nuclear translocation. Nuclear β-catenin induced by spermine then activates transcriptional expression of PD-L1 and N-glycosyltransferase STT3A, while STT3A in turn increases the stability of PD-L1 through inducing PD-L1 protein N-glycosylation in HCC cells.
Conclusions
This study reveals the crucial function of spermine in establishing immune privilege by increasing the expression and N-glycosylation of PD-L1, providing a potential strategy for the treatment of hepatocellular carcinoma.
Collapse
|
41
|
Research progress on the role of cholesterol in hepatocellular carcinoma. Eur J Pharmacol 2022; 938:175410. [DOI: 10.1016/j.ejphar.2022.175410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
|
42
|
Integrative Analysis Reveals the Potential Role and Prognostic Value of GOLM1 in Hepatocellular Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8284500. [PMID: 36211823 PMCID: PMC9535134 DOI: 10.1155/2022/8284500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022]
Abstract
Overexpression of Golgi membrane protein 1 (GOLM1) is closely associated with hepatocellular carcinoma (HCC) vascular invasion. How GOLM1 may be involved in angiogenesis in HCC remains unclear. We explored how GOLM1 promotes angiogenesis in HCC and potential prognostic value. Expression levels of GOLM1 in HCC patients and healthy controls were obtained from The Cancer Genome Atlas (TCGA). Differentially expressed genes (DEGs) between HCC patients and controls were compared. GOLM1 was knocked out in the HCC cell line, and RNA sequencing and DEG expression analysis were performed compared with control cells. Based on TCGA data and cell line RNA sequencing data, DEGs affected by a high expression of GOLM1 were identified. Subsequently, enrichment analysis was performed to explore the functions and pathways of the DEGs affected by a high expression of GOLM1. A relevant network analysis was built. Cox regression, genomic variance analysis scores, minimum absolute shrinkage and selection operator regression, and random forest regression models were applied to determine the best prognostic model and validated using the GSE54236 dataset from the Gene Expression Omnibus (GEO). We determined the effect of GOLM1 expression on immune cell infiltration in liver cancer. GOLM1 was overexpressed in HCC tissues compared with controls, and its level correlated with tumor purity and prognosis. 400 DEGs affected by highly expressed GOLM1 were identified in TCGA and cell line RNA sequencing data. Enrichment analysis revealed that these DEGs may be related to biological processes of oxidative stress and angiogenesis and involved in the VEGF signaling pathway and protein processing in endoplasmic reticulum. We predicted a comprehensive regulatory network in which GOLM1 activated VEGF signaling to promote HCC angiogenesis. GOLM1 may interact with E2F1 and IGF2BP3 to promote angiogenesis. GOLM1 overexpression was associated with greater immune cell infiltration. A random forest regression model was the best prognostic model. Our study reveals a potential molecular mechanism of GOLM1 in promoting HCC. We developed two prognostic models based on DEG associated with GOLM1 overexpression to help stratify HCC prognosis and improve individualized treatment.
Collapse
|
43
|
Loosen SH, Halpaap J, Labuhn S, Bednarsch J, Alizai PH, Roeth AA, Lang SA, Vucur M, Kather JN, Knoefel WT, Ulmer TF, Neumann UP, Roderburg C, Luedde T. Golgi Protein 73 (GP73) Serum Levels Predict Outcome after Resection of Biliary Tract Cancer. Cancers (Basel) 2022; 14:cancers14184428. [PMID: 36139589 PMCID: PMC9497317 DOI: 10.3390/cancers14184428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Biliary tract cancer (BTC) represents a rare liver malignancy with unfavorable outcome. It is often challenging to identify the ideal surgical candidates and present stratification algorithms are rarely based on the individual tumor biology. In the present manuscript, we evaluated a role of serum Golgi protein 73 (GP73) in patients with resectable BTC. We could show that elevated levels of GP73 before surgery identified a subgroup of BTC patients with a significantly reduced overall survival after tumor resection. Therefore, measurement of GP73 serum levels might become a novel tool in the challenging preoperative stratification process of patients with resectable BTC. Abstract Background: Tumor resection represents the only potentially curative therapy for patients with biliary tract cancer. Nevertheless, disease recurrence is observed in about 50% of patients, leading to a 5-years survival rate of less than 50%. The Golgi protein 73 (GP73), a type II Golgi transmembrane protein, exerts important functions of intracellular protein processing and transportation. Circulating GP73 has recently been suggested as a prognostic marker following resection of hepatocellular carcinoma (HCC) but its role in the context of BTC has remained unknown. In this study, we evaluate a potential role of circulating GP73 as a novel biomarker in patients with resectable BTC. Methods: GP73 serum levels were measured by immunoassay in n = 97 BTC and n = 40 HCC patients as well as n = 31 healthy controls. Results were correlated with clinical data. Results: Serum GP73 levels were significantly elevated in BTC patients compared to healthy controls but lower compared to HCC patients. The combination of GP73/CA19-9 showed a sensitivity and specificity of 83.5% and 90.3% regarding the differentiation of BTC patients and healthy controls. BTC patients with baseline GP73 levels above the ideal cut-off value (42.47 ng/mL) showed a significantly reduced median overall survival (193 days) compared to patients with preoperative GP73 levels below this cut-off (882 days). These results were confirmed in uni- and multivariate Cox-regression analysis including several clinicopathological parameters such as age, ECOG performance status, tumor stage as well as established tumor markers and parameters of liver and kidney function. Conclusions: GP73 represents a previously unrecognized biomarker in the patients with resectable BTC that identifies patients with an impaired postoperative outcome. If larger clinical trials confirmed these findings, measurement of GP73 serum levels might become a novel tool in the challenging preoperative stratification process of patients with resectable BTC.
Collapse
Affiliation(s)
- Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: (S.H.L.); (T.L.); Tel.: +49-211-81-16630 (S.H.L. & T.L.); Fax: +49-211-81-04489 (S.H.L. & T.L.)
| | - Justus Halpaap
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Simon Labuhn
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jan Bednarsch
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Patrick H. Alizai
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Anjali A. Roeth
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Sven A. Lang
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Mihael Vucur
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jakob N. Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, 01069 Dresden, Germany
| | - Wolfram T. Knoefel
- Department of General, Visceral and Pediatric Surgery, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tom F. Ulmer
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Ulf P. Neumann
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: (S.H.L.); (T.L.); Tel.: +49-211-81-16630 (S.H.L. & T.L.); Fax: +49-211-81-04489 (S.H.L. & T.L.)
| |
Collapse
|
44
|
GOLM1 depletion modifies cellular sphingolipid metabolism and adversely affects cell growth. J Lipid Res 2022; 63:100259. [PMID: 35948172 PMCID: PMC9475319 DOI: 10.1016/j.jlr.2022.100259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022] Open
Abstract
Golgi membrane protein 1 (GOLM1) is a Golgi-resident type 2 transmembrane protein known to be overexpressed in several cancers, including hepatocellular carcinoma (HCC), as well as in viral infections. However, the role of GOLM1 in lipid metabolism remains enigmatic. In this study, we employed siRNA-mediated GOLM1 depletion in Huh-7 HCC cells to study the role of GOLM1 in lipid metabolism. Mass spectrometric lipidomic analysis in GOLM1 knockdown cells showed an aberrant accumulation of sphingolipids, such as ceramides, hexosylceramides, dihexosylceramides, sphinganine, sphingosine, and ceramide phosphate, along with cholesteryl esters. Furthermore, we observed a reduction in phosphatidylethanolamines and lysophosphatidylethanolamines. In addition, Seahorse extracellular flux analysis indicated a reduction in mitochondrial oxygen consumption rate upon GOLM1 depletion. Finally, alterations in Golgi structure and distribution were observed both by electron microscopy imaging and immunofluorescence microscopy analysis. Importantly, we found that GOLM1 depletion also affected cell proliferation and cell cycle progression in Huh-7 HCC cells. The Golgi structural defects induced by GOLM1 reduction might potentially affect the trafficking of proteins and lipids leading to distorted intracellular lipid homeostasis, which may result in organelle dysfunction and altered cell growth. In conclusion, we demonstrate that GOLM1 depletion affects sphingolipid metabolism, mitochondrial function, Golgi structure, and proliferation of HCC cells.
Collapse
|
45
|
Ma XL, Zhu KY, Chen YD, Tang WG, Xie SH, Zheng H, Tong Y, Wang YC, Ren N, Guo L, Lu RQ. Identification of a novel Calpain-2-SRC feed-back loop as necessity for β-Catenin accumulation and signaling activation in hepatocellular carcinoma. Oncogene 2022; 41:3554-3569. [PMID: 35697802 DOI: 10.1038/s41388-022-02367-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 11/09/2022]
Abstract
Rapid progression is the major cause of the poor prognosis of hepatocellular carcinoma (HCC); however, the underlying mechanism remained unclear. Here, we found Calpain-2 (CAPN2), a well-established protease that accelerates tumor progression in several malignancies, is overexpressed in HCC and acts as an independent predictor for poor outcomes. Furthermore, CAPN2 promoted the proliferation and invasion of HCC, and showed a positive correlation with the levels of invasion-related markers. Mechanistically, a novel CAPN2-SRC positive regulatory loop was identified upstream of β-catenin to prevent its ubiquitination and degradation, and subsequently promoted HCC progression: CAPN2 could proteolyze PTP1B to form a truncation of approximately 42 kDa with increased phosphatase activity, resulting in reduced SRC Y530 phosphorylation and increased SRC kinase activity; meanwhile, CAPN2 itself was a bone fide substrate of SRC that was primarily phosphorylated at Y625 by SRC and exhibited increased proteolysis activity upon phosphorylation. Interestingly, the CAPN2-SRC loop could not only restrain most of cytoplasmic β-catenin degradation by inhibiting GSK3β pathway, but also prevented TRIM33-induced nuclear β-catenin degradation even in β-catenin-mutant cells. Present study identified a CAPN2-SRC positive loop responsible for intracellular β-catenin accumulation and signaling activation, and targeting CAPN2 protease activity might be a promising approach for preventing HCC progression.
Collapse
Affiliation(s)
- Xiao-Lu Ma
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ke-Yu Zhu
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yue-Da Chen
- Department of general surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei-Guo Tang
- Liver cancer institute, Zhongshan hospital, Fudan university, Shanghai, 200032, China.,Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Su-Hong Xie
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Hui Zheng
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ying Tong
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yan-Chun Wang
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ning Ren
- Liver cancer institute, Zhongshan hospital, Fudan university, Shanghai, 200032, China. .,Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China.
| | - Lin Guo
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| | - Ren-Quan Lu
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Tong J, Yao M, Mu X, Wang L, Wen X, Zhai X, Xu X, Wang Y, Chen J, Zhai X, Guan C, Lu F, Hu J. Relationship between the Level of Serum Golgi Protein 73 and the Risk of Short-term Death in Patients with ALD-ACLF. J Clin Transl Hepatol 2022; 10:449-457. [PMID: 35836755 PMCID: PMC9240251 DOI: 10.14218/jcth.2020.00133] [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: 11/28/2020] [Revised: 06/08/2021] [Accepted: 09/21/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS As a hepatocellular carcinoma biomarker, serum Golgi protein 73 (GP73) is reportedly related to inflammation. Acute-on-chronic liver failure (ACLF) is characterized by severe systemic inflammation. In this study, we aimed to explore the association between the GP73 level and short-term mortality in patients with alcohol-associated liver disease-related ACLF (ALD-ACLF). METHODS This retrospective cohort study involved 126 Chinese adults with ALD-ACLF. Baseline serum GP73 level was measured using enzyme-linked immunosorbent assay. Patients were followed-up for 90 d and outcomes were assessed. Data were analyzed using multivariate Cox regression and piecewise linear regression analyses. The predictive value of GP73 and classic models for the short-term prognosis of participants were evaluated and compared using receiver operating characteristic curves. RESULTS The serum GP73 level was independently associated with an increased mortality risk in patients with ALD-ACLF. Compared with the lowest tertile, the highest serum GP73 level predisposed patients with ALD-ACLF to a higher mortality risk in the fully adjusted model [at 28 days: hazard ratio (HR): 4.29 (0.99-18.54), p=0.0511; at 90 days: HR: 3.52 (1.15-10.79), p=0.0276]. Further analysis revealed a positive linear association. GP73 significantly improved the accuracy of the Child-Turcotte-Pugh score, model for end-stage liver disease score, and model for end-stage liver disease-sodium score in predicting short-time prognosis of patients with ALD-ACLF. CONCLUSIONS The serum GP73 level is a significant predictor of the subsequent risk of death in patients with ALD-ACLF. GP73 improved the predictive value of classic prognostic scores.
Collapse
Affiliation(s)
- Jingjing Tong
- Chinese PLA Medical School, Beijing, China
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Mingjie Yao
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiuying Mu
- Peking University 302 Clinical Medical School, Beijing, China
| | - Leijie Wang
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiajie Wen
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xingran Zhai
- Peking University 302 Clinical Medical School, Beijing, China
| | - Xiang Xu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yu Wang
- Chinese PLA Medical School, Beijing, China
| | - Jing Chen
- Chinese PLA Medical School, Beijing, China
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xiangwei Zhai
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chongdan Guan
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Fengmin Lu
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Correspondence to: Jinhua Hu, Chinese PLA Medical School, No. 28, Fuxing Road, Haidian District, Beijing 100853, China; Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, 100 Xisihuan Road, Fengtai District, Beijing 100039, China. ORCID: https://orcid.org/0000-0002-0647-9898. Tel: +86-10-66933405, Fax: +86-10-66933434, E-mail: ; Fengmin Lu, Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China. ORCID: https://orcid.org/0000-0002-1832-3209. Tel: +86-10-82805136, Fax: +86-10-82805136, E-mail:
| | - Jinhua Hu
- Chinese PLA Medical School, Beijing, China
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Peking University 302 Clinical Medical School, Beijing, China
- Correspondence to: Jinhua Hu, Chinese PLA Medical School, No. 28, Fuxing Road, Haidian District, Beijing 100853, China; Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, 100 Xisihuan Road, Fengtai District, Beijing 100039, China. ORCID: https://orcid.org/0000-0002-0647-9898. Tel: +86-10-66933405, Fax: +86-10-66933434, E-mail: ; Fengmin Lu, Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China. ORCID: https://orcid.org/0000-0002-1832-3209. Tel: +86-10-82805136, Fax: +86-10-82805136, E-mail:
| |
Collapse
|
47
|
Spano D, Colanzi A. Golgi Complex: A Signaling Hub in Cancer. Cells 2022; 11:1990. [PMID: 35805075 PMCID: PMC9265605 DOI: 10.3390/cells11131990] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 02/01/2023] Open
Abstract
The Golgi Complex is the central hub in the endomembrane system and serves not only as a biosynthetic and processing center but also as a trafficking and sorting station for glycoproteins and lipids. In addition, it is an active signaling hub involved in the regulation of multiple cellular processes, including cell polarity, motility, growth, autophagy, apoptosis, inflammation, DNA repair and stress responses. As such, the dysregulation of the Golgi Complex-centered signaling cascades contributes to the onset of several pathological conditions, including cancer. This review summarizes the current knowledge on the signaling pathways regulated by the Golgi Complex and implicated in promoting cancer hallmarks and tumor progression.
Collapse
Affiliation(s)
- Daniela Spano
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Antonino Colanzi
- Institute for Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy;
| |
Collapse
|
48
|
Ding D, Zhang Y, Zhang X, Shi K, Shang W, Ying J, Wang L, Chen Z, Hong H. MiR-30a-3p Suppresses the Growth and Development of Lung Adenocarcinoma Cells Through Modulating GOLM1/JAK-STAT Signaling. Mol Biotechnol 2022; 64:1143-1151. [PMID: 35438415 DOI: 10.1007/s12033-022-00497-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Abstract
A considerable amount of people succumbs to lung adenocarcinoma (LUAD) due to its high incidence and mortality. This study attempted to reveal the impacts of GOLM1 on LUAD. This work analyzed GOLM1 expression in LUAD and normal tissue and studied its prognostic value utilizing data from The Cancer Genome Atlas. RNA and protein levels were, respectively, determined utilizing qRT-PCR and western blot. Cell-aggressive behaviors were assessed employing Cell Counting Kit-8, scratch healing, and Transwell assays. The targetting relationship between GOLM1 and miR-30a-3p was assayed by dual-luciferase method. GOLM1 up-regulation in LUAD was found in TCGA and it was also a negative factor for survival in patients. GOLM1 overexpression promoted cell progression in LUAD. Down-regulated miR-30a-3p in LUAD was an upstream regulatory miRNA of GOLM1 in terms of molecular mechanism. Further, rescue assays illustrated that miR-30a-3p overexpression attenuated the GOLM1 facilitating impacts on LUAD progression. Finally, we proved that miR-30a-3p/GOLM1 regulated progression of LUAD cells via JAK-STAT pathway. Collectively, the inhibitory impacts of miR-30a-3p on LUAD growth may be mediated by GOLM1/JAK-STAT, which may contribute to the diagnosis of LUAD therapy and the development of therapeutic tools.
Collapse
Affiliation(s)
- Dongxiao Ding
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Yunqiang Zhang
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Xuede Zhang
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Ke Shi
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Wenjun Shang
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Junjie Ying
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Li Wang
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Zhongjie Chen
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China
| | - Haihua Hong
- Department of Thoracic Surgery, The People's Hospital of Beilun District (Beilun Branch of the First Affiliated Hospital of Medical College of Zhejiang University), No.1288, East Lushan Road, Xinqi Sub-District, Beilun District, Zhejiang Province, Ningbo City, 315800, China.
| |
Collapse
|
49
|
Shao WQ, Zhu WW, Luo MJ, Fan MH, Li Q, Wang SH, Lin ZF, Zhao J, Zheng Y, Dong QZ, Lu L, Jia HL, Zhang JB, Lu M, Chen JH, Qin LX. Cholesterol suppresses GOLM1-dependent selective autophagy of RTKs in hepatocellular carcinoma. Cell Rep 2022; 39:110712. [PMID: 35443161 DOI: 10.1016/j.celrep.2022.110712] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/21/2022] [Accepted: 03/29/2022] [Indexed: 01/03/2023] Open
Abstract
Aberrant activation of receptor tyrosine kinases (RTKs) and the subsequent metabolic reprogramming play critical roles in cancer progression. Our previous study has shown that Golgi membrane protein 1 (GOLM1) promotes hepatocellular carcinoma (HCC) metastasis by enhancing the recycling of RTKs. However, how this RTK recycling process is regulated and coupled with RTK degradation remains poorly defined. Here, we demonstrate that cholesterol suppresses the autophagic degradation of RTKs in a GOLM1-dependent manner. Further mechanistic studies reveal that GOLM1 mediates the selective autophagy of RTKs by interacting with LC3 through an LC3-interacting region (LIR), which is regulated by a cholesterol-mTORC1 axis. Lowering cholesterol by statins improves the efficacy of multiple tyrosine kinase inhibitors (TKIs) in vivo. Our findings indicate that cholesterol serves as a signal to switch GOLM1-RTK degradation to GOLM1-RTK recycling and suggest that lowering cholesterol by statin may be a promising combination strategy to improve the TKI efficiency in HCC.
Collapse
Affiliation(s)
- Wei-Qing Shao
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Wen-Wei Zhu
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Meng-Jun Luo
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ming-Hao Fan
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Qin Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sheng-Hao Wang
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Zhi-Fei Lin
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Jing Zhao
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Yan Zheng
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Qiong-Zhu Dong
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Lu Lu
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Hu-Liang Jia
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Ju-Bo Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ming Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Jin-Hong Chen
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
| | - Lun-Xiu Qin
- General Surgery Department of Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
50
|
EGFR/MET promotes hepatocellular carcinoma metastasis by stabilizing tumor cells and resisting to RTKs inhibitors in circulating tumor microemboli. Cell Death Dis 2022; 13:351. [PMID: 35428350 PMCID: PMC9012802 DOI: 10.1038/s41419-022-04796-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022]
Abstract
The receptor tyrosine kinases (RTKs) family is well-recognized as vital targets for the treatment of hepatocarcinoma cancer (HCC) clinically, whereas the survival benefit of target therapy sorafenib is not satisfactory for liver cancer patients due to metastasis. EGFR and MET are two molecules of the RTK family that were related to the survival time of liver cancer patients and resistance to targeted therapy in clinical reports. However, the mechanism and clinical therapeutic value of EGFR/MET in HCC metastasis are still not completely clarified. The study confirmed that EGFR/MET was highly expressed in HCC cells and tissues and the phosphorylation was stable after metastasis. The expression of EGFR/MET was up-regulated in circulating tumor microemboli (CTM) to accelerate IL-8 production and resistance to the lethal effect of leukocytes. Meanwhile, highly expressed EGFR/MET effectively regulated the Ras/MAPK pathway and stabilized suspended HCC cells by facilitating proliferation and inhibiting apoptosis. Moreover, EGFR/MET promoted phosphorylation of hetero-RTKs, which was dependent on high-energy phosphoric acid compounds rather than their direct interactions. In conclusion, highly expressed EGFR/MET could be used in CTM identification and suitable for preventing metastasis of HCC in clinical practice.
Collapse
|