1
|
Song X, Tong X, Yang K, Qi Y, Liu W, Sun Y, Wang C, Xun F, Wang Z, Jiang M, Zhang Y, Ren T, Chen D, Hou S, Song A, Gao H, Zhao Q. Discovery and evaluation of novel Benzohydroxamic acid-indole derivatives as dual inhibitors of ADAM17 and HDAC2 with antitumor activity. Bioorg Chem 2025; 157:108308. [PMID: 40049049 DOI: 10.1016/j.bioorg.2025.108308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/18/2025]
Abstract
Hepatocellular carcinoma (HCC) has garnered significant attention from researchers due to its high recurrence rate and invasive characteristics. The design of drugs with dual-target combined effects represents a promising strategy in cancer treatment. Our observations suggest that ADAM17 and HDAC may inhibit the unfavorable prognostic signaling pathway Notch1 in HCC through distinct mechanisms, thereby suppressing tumor cell proliferation and metastasis. Consequently, this study utilized the ADAM17 inhibitor ZLDI-8 as a lead compound and developed a series of dual ADAM17/HDAC2 inhibitors by integrating strategies such as backbone leaping and pharmacophore fusion. We assessed the anti-hepatocellular carcinoma activity of these compounds, focusing on their anti-proliferative, pro-apoptotic, and anti-metastatic properties. Notably, ZSNI-21 effectively inhibited the proliferation of Bel-7402 cells and demonstrated significant anti-metastatic capabilities against HCC-LM3 cells, with its targeting confirmed. Additionally, its in vivo safety was validated. To date, there have been no reports on dual ADAM17/HDAC2 inhibitors, marking this as a novel endeavor.
Collapse
Affiliation(s)
- Xiaoxuan Song
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, Shenyang, Liaoning 110016, PR China
| | - Xin Tong
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Kaisi Yang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Yiming Qi
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Wenwu Liu
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, PR China
| | - Yuzhu Sun
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China. Shenyang, Liaoning 110016, PR China
| | - Chengkang Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Fanghua Xun
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Ziyi Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Muxuan Jiang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Yingshi Zhang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Tianshu Ren
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Di Chen
- Department of Pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences;, Beijing 100730, PR China; General Hospital of Northern Theater Command, China Medical University, PR China
| | - Shanbo Hou
- Luoxin Pharmaceuticals Group Stock Co., Ltd., Linyi, PR China
| | - Aigang Song
- Luoxin Pharmaceuticals Group Stock Co., Ltd., Linyi, PR China
| | - Huiyuan Gao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, Shenyang, Liaoning 110016, PR China; Key Laboratory of Pharmacodynamic Substances Research & Translational Medicine of Immune Diseases of Shenyang, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, Shenyang, Liaoning 110016, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenhe District, Shenyang 110016, PR China..
| |
Collapse
|
2
|
Zhang Y, Lu Y, Wang N, Hao F, Chen Y, Fei X, Wang J. Ascending E2F7a/b ratio facilitates KLF13 transcription in hepatocellular carcinoma and correlates with the abundance of binuclear hepatocytes (ABH) modulation for short-term recurrence. FASEB J 2025; 39:e70485. [PMID: 40116212 PMCID: PMC11926945 DOI: 10.1096/fj.202402520r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Short-term recurrence after surgery severely threatens patients' lives and leads to dismal outcomes in hepatocellular carcinoma (HCC). Our previous research proposed the abundance of binuclear hepatocytes (ABH) as an independent indicator related to the cytokinesis regulator Anillin and significantly associated with HCC recurrence. The exact mechanism of ABH modulation has not been clearly illustrated. In this study, we intensively investigated the probable regulation mechanism and noticed a contradiction between E2F7 upregulation and ABH attenuation. As we discovered, E2F7 has two isoforms, E2F7a and E2F7b, and we innovatively define a value of the E2F7a/b ratio using a cutoff value of 6.5. E2F7 upregulation in the paracancerous tissues was predominantly presented by the E2F7a isoform, leading to an increase in the E2F7a/b ratio, instead of E2F7b as a main component in non-cancerous tissues, and is associated with short-term recurrence. We further found that KLF13 transcriptionally promotes Anillin expression in HCC and was suppressively impacted by E2F7b, but not by the highly expressed E2F7a. Hence, the ascending E2F7a/b ratio induced significant upregulation of KLF13 and participated in the attenuation of ABH in the paracancerous liver tissues. In conclusion, E2F7 presents a particular expression status in HCC by predominantly upregulating E2F7a rather than E2F7b. The ascending E2F7a/b ratio weakens the suppressive effect on KLF13 transcription and sequentially participates in ABH attenuation, associated with short-term HCC recurrence post-operation.
Collapse
Affiliation(s)
- Yian Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yiquan Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Nan Wang
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
3
|
Zhang C, Hu S, Yin C, Wang G, Liu P. STAT3 orchestrates immune dynamics in hepatocellular carcinoma: A pivotal nexus in tumor progression. Crit Rev Oncol Hematol 2025; 207:104620. [PMID: 39818308 DOI: 10.1016/j.critrevonc.2025.104620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) presents a formidable challenge in oncology, attributed to its association with chronic liver diseases and global prevalence. The immune microenvironment profoundly influences HCC progression, balancing immune suppression and antitumor responses. The Signal Transducer and Activator of Transcription 3 (STAT3) is central to this equilibrium, orchestrating immune dynamics and intertwining tumor progression with immune evasion mechanisms. Dysregulated STAT3 signaling, activated by various stimuli, including cytokines and growth factors, promotes an immunosuppressive milieu within HCC tumors, fostering tumor survival and proliferation while hindering immune surveillance. Non-coding RNAs and other molecules regulate this process, modulating STAT3 activity and influencing immune cell function. Moreover, therapeutic interventions targeting the STAT3 pathway, alongside advancements in radiotherapy, cancer vaccines, and diabetes-related drugs, offer promising strategies in HCC management. Integrating natural compounds with immunotherapy emerges as a novel approach, leveraging their ability to enhance antitumor immunity and counter immune evasion strategies. Understanding the multifaceted role of STAT3 and its interactions within the immune landscape of HCC is paramount for devising effective therapeutic interventions and improving patient outcomes.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Songbai Hu
- Department of Cancer Center, Yuexi County Hospital, Anqing, Anhui Province 246600, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Hubei, China.
| |
Collapse
|
4
|
Ji C, Zhong Q, Su H, Xue X, Yang R, Li N. DDX21 Is a Potential Biomarker for Predicting Recurrence and Prognosis in Hepatocellular Carcinoma. Anal Cell Pathol (Amst) 2025; 2025:1018820. [PMID: 39866844 PMCID: PMC11760617 DOI: 10.1155/ancp/1018820] [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: 09/21/2023] [Revised: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 01/28/2025] Open
Abstract
DEAD-box helicase 21 (DDX21) is a conserved Asp-Glu-Ala-Asp (DEAD) box RNA helicase with multiple functions that is involved in various cellular processes and diseases. However, the role of DDX21 in the recurrence and prognosis of hepatocellular carcinoma (HCC) patients remains unknown. In the current study, we examined the protein expression of DDX21 in HCC tissues through immunohistochemical staining and analyzed the correlation between DDX21 protein expression and clinical outcome via Kaplan-Meier survival analysis. The Cox proportional hazards regression model was used to assess the interrelationships between the outcome and variable over time. Our results showed that increased expression of DDX21 protein was observed in HCC tissues compared with paracancerous tissues and was associated with advanced BCLC stage. Recurrent HCC patients had higher levels of DDX21 protein than nonrecurrent cases. Notably, DDX21 was an independent risk factor for predicting worse overall survival and recurrence-free survival in HCC patients. Furthermore, lack of DDX21 abated the growth and mobility of Hep3B cells. Taken together, our data highlight the clinical significance of DDX21 in the recurrence and prognosis of HCC patients and indicate that targeting DDX21 may represent an effective therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Chengjie Ji
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Laboratory Medicine, The People's Hospital of Jianyang City, Chengdu, Sichuan, China
| | - Qing Zhong
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Anesthesiology, The People's Hospital of Jianyang, Chengdu, Sichuan, China
| | - Huilan Su
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoli Xue
- Department of Laboratory Medicine, Yingshan County People's Hospital, Nanchong, Sichuan, China
| | - Renxiang Yang
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Na Li
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Jia J, Zhou X, Chu Q. Mechanisms and therapeutic prospect of the JAK-STAT signaling pathway in liver cancer. Mol Cell Biochem 2025; 480:1-17. [PMID: 38519710 DOI: 10.1007/s11010-024-04983-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 03/25/2024]
Abstract
Liver cancer (LC) poses a significant global health challenge due to its high incidence and poor prognosis. Current systemic treatment options, such as surgery, chemotherapy, radiofrequency ablation, and immunotherapy, have shown limited effectiveness for advanced LC patients. Moreover, owing to the heterogeneous nature of LC, it is crucial to uncover more in-depth pathogenic mechanisms and develop effective treatments to address the limitations of the existing therapeutic modalities. Increasing evidence has revealed the crucial role of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in the pathogenesis of LC. The specific mechanisms driving the JAK-STAT pathway activation in LC, participate in a variety of malignant biological processes, including cell differentiation, evasion, anti-apoptosis, immune escape, and treatment resistance. Both preclinical and clinical investigations on the JAK-STAT pathway inhibitors have exhibited potential in LC treatment, thereby opening up avenues for the development of more targeted therapeutic strategies for LC. In this study, we provide an overview of the JAK-STAT pathway, delving into the composition, activation, and dynamic interplay within the pathway. Additionally, we focus on the molecular mechanisms driving the aberrant activation of the JAK-STAT pathway in LC. Furthermore, we summarize the latest advancements in targeting the JAK-STAT pathway for LC treatment. The insights presented in this review aim to underscore the necessity of research into the JAK-STAT signaling pathway as a promising avenue for LC therapy.
Collapse
Affiliation(s)
- JunJun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China.
| | - Xuelian Zhou
- Division of Endocrinology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
6
|
Su L, Zhao D, Zhou C, Zhang B. NEK6 Accelerates Hepatocellular Carcinoma Progression and Glycolysis through Ubiquitination of TCP10L. Crit Rev Eukaryot Gene Expr 2025; 35:1-13. [PMID: 40228222 DOI: 10.1615/critreveukaryotgeneexpr.2025057446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Never in mitosis a related kinases 6 (NEK6) is a serine/threonine kinase, and dysregulation of NEK6 is associated with malignant progression of human cancers. Nonetheless, the biological function and molecular mechanism of NEK6 in hepatocellular carcinoma (HCC) are unknown. Our study found that NEK6 was obviously raised in HCC patient tissues and cells, and patients with high NEK6 expression had a worse prognosis. Silencing of NEK6 reduced the growth, metastasis, cell cycle, and glycolysis of HCC cells while facilitating apoptosis. In vivo experiments also showed that NEK6 knockdown dramatically hampered tumor growth, suggesting that NEK6 enhanced HCC progression in vivo and in vitro. Next, we proved that TCP10L was a target gene of NEK6, and NEK6 negatively regulated TCP10L expression. Mechanistically, we confirmed that NEK6 was bound to TCP10L, and NEK6 degraded TCP10L protein expression through ubiquitination. Rescue experiments also declared that TCP10L reversed the effect of NEK6 on HCC cells. Our results disclosed that NEK6 heightened HCC progression and glycolysis through ubiquitination of TCP10L. Our study may provide a new perspective for the treatment of HCC.
Collapse
Affiliation(s)
- Ling Su
- Department of Pathology, The People's Hospital of Suzhou New District, Suzhou 215011, Jiangsu, China
| | - Dehong Zhao
- Department of General Surgery, Yancheng No. 1 People's Hospital, Yancheng 224000, Jiangsu, China
| | - Cheng Zhou
- Department of General Surgery, Yancheng No. 1 People's Hospital, Yancheng 224000, Jiangsu, China
| | | |
Collapse
|
7
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [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: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Zhu CX, Yan K, Chen L, Huang RR, Bian ZH, Wei HR, Gu XM, Zhao YY, Liu MC, Suo CX, Li ZK, Yang ZY, Lu MQ, Hua XF, Li L, Zhao ZB, Sun LC, Zhang HF, Gao P, Lian ZX. Targeting OXCT1-mediated ketone metabolism reprograms macrophages to promote antitumor immunity via CD8 + T cells in hepatocellular carcinoma. J Hepatol 2024; 81:690-703. [PMID: 38759889 DOI: 10.1016/j.jhep.2024.05.007] [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: 09/04/2023] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND & AIMS The liver is the main organ of ketogenesis, while ketones are mainly metabolized in peripheral tissues via the critical enzyme 3-oxoacid CoA-transferase 1 (OXCT1). We previously found that ketolysis is reactivated in hepatocellular carcinoma (HCC) cells through OXCT1 expression to promote tumor progression; however, whether OXCT1 regulates antitumor immunity remains unclear. METHODS To investigate the expression pattern of OXCT1 in HCC in vivo, we conducted multiplex immunohistochemistry experiments on human HCC specimens. To explore the role of OXCT1 in mouse HCC tumor-associated macrophages (TAMs), we generated LysMcreOXCT1f/f (OXCT1 conditional knockout in macrophages) mice. RESULTS Here, we found that inhibiting OXCT1 expression in tumor-associated macrophages reduced CD8+ T-cell exhaustion through the succinate-H3K4me3-Arg1 axis. Initially, we found that OXCT1 was highly expressed in liver macrophages under steady state and that OXCT expression was further increased in TAMs. OXCT1 deficiency in macrophages suppressed tumor growth by reprogramming TAMs toward an antitumor phenotype, reducing CD8+ T-cell exhaustion and increasing CD8+ T-cell cytotoxicity. Mechanistically, high OXCT1 expression induced the accumulation of succinate, a byproduct of ketolysis, in TAMs, which promoted Arg1 transcription by increasing the H3K4me3 level in the Arg1 promoter. In addition, pimozide, an inhibitor of OXCT1, suppressed Arg1 expression as well as TAM polarization toward the protumor phenotype, leading to decreased CD8+ T-cell exhaustion and slower tumor growth. Finally, high expression of OXCT1 in macrophages was positively associated with poor survival in patients with HCC. CONCLUSIONS In conclusion, our results demonstrate that OXCT1 epigenetically suppresses antitumor immunity, suggesting that suppressing OXCT1 activity in TAMs could be an effective approach for treating liver cancer. IMPACT AND IMPLICATIONS The intricate metabolism of liver macrophages plays a critical role in shaping hepatocellular carcinoma progression and immune modulation. Targeting macrophage metabolism to counteract immune suppression presents a promising avenue for hepatocellular carcinoma treatment. Herein, we found that the ketogenesis gene OXCT1 was highly expressed in tumor-associated macrophages (TAMs) and promoted tumor growth by reprogramming TAMs toward a protumor phenotype. Pharmacological targeting or genetic downregulation of OXCT1 in TAMs enhances antitumor immunity and slows tumor growth. Our results suggest that suppressing OXCT1 activity in TAMs could be an effective approach for treating liver cancer.
Collapse
Affiliation(s)
- Chu-Xu Zhu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Kai Yan
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liang Chen
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Rong-Rong Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhen-Hua Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Hao-Ran Wei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xue-Mei Gu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yang-Yang Zhao
- School of Medicine, South China University of Technology, Guangzhou, China; Biomedical Engineering Cockrell School of Engineering, University of Texas at Austin, Austin, United States
| | - Meng-Chu Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Cai-Xia Suo
- Department of Colorectal Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhi-Kun Li
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhi-Yi Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Min-Qiang Lu
- Department of Hepatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, China
| | - Xue-Feng Hua
- Department of Hepatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, China
| | - Liang Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhi-Bin Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lin-Chong Sun
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hua-Feng Zhang
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Ping Gao
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Zhe-Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Botwe G, Fang X, Mukhtar YM, Zhou Y, Tang H, Wang M, Zhang J, Fu M, Jiang P, Gu J, Zhang X. Circular RNAs as biomarkers and therapeutic targets for gastrointestinal cancers. J Gastroenterol Hepatol 2024; 39:1230-1246. [PMID: 38504413 DOI: 10.1111/jgh.16536] [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: 09/06/2023] [Revised: 02/03/2024] [Accepted: 02/24/2024] [Indexed: 03/21/2024]
Abstract
Circular RNAs are a class of noncoding RNAs with covalently linked 5' and 3' ends that arise from backsplicing events. The absence of a 5' cap and a 3' poly(A) tail makes circular RNAs relatively more stable than their linear counterparts. They are evolutionary conserved and tissue-specific, and some show disease-specific expression patterns. Although their biological functions remain largely unknown, circular RNAs have been shown to play regulatory roles by acting as microRNA sponges, regulators of RNA-binding proteins, alternative splicing, and parental gene expression, and they could even encode proteins. Over the past few decades, circular RNAs have attracted wide attention in oncology owing to their implications in various tumors. Many circular RNAs have been characterized as key players in gastrointestinal cancers and influence cancer growth, progression, metastasis, and therapeutic resistance. Accumulating evidence reveals that their unique characteristics, coupled with their critical roles in tumorigenesis, make circular RNAs promising non-invasive clinical biomarkers for gastrointestinal cancers. In the present review, we summarized the biological roles of the emerging circular RNAs and their potential as biomarkers and therapeutic targets, which may help better understand their clinical significance in the management of gastrointestinal cancers.
Collapse
Affiliation(s)
- Godwin Botwe
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Institute of Digestive Diseases, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinjian Fang
- Department of Oncology, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, Jiangsu, China
| | - Yusif Mohammed Mukhtar
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yue Zhou
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Haozhou Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Min Fu
- Institute of Digestive Diseases, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Pengcheng Jiang
- Institute of Digestive Diseases, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jianmei Gu
- Department of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
10
|
Pan T, Ding P, Huang A, Tang B, Song K, Sun G, Wu Y, Yang S, Chen X, Wang D, Zhu X. Reconstitution of double-negative T cells after cord blood transplantation and its predictive value for acute graft-versus-host disease. Chin Med J (Engl) 2024; 137:1207-1217. [PMID: 37620289 PMCID: PMC11101234 DOI: 10.1097/cm9.0000000000002807] [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: 01/27/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND With an increasing number of patients with hematological malignancies being treated with umbilical cord blood transplantation (UCBT), the correlation between immune reconstitution (IR) after UCBT and graft-versus-host disease (GVHD) has been reported successively, but reports on double-negative T (DNT) cell reconstitution and its association with acute GVHD (aGVHD) after UCBT are lacking. METHODS A population-based observational study was conducted among 131 patients with hematological malignancies who underwent single-unit UCBT as their first transplant at the Department of Hematology, the First Affiliated Hospital of USTC, between August 2018 and June 2021. IR differences were compared between the patients with and without aGVHD. RESULTS The absolute number of DNT cells in the healthy Chinese population was 109 (70-157)/μL, accounting for 5.82 (3.98-8.19)% of lymphocytes. DNT cells showed delayed recovery and could not reach their normal levels even one year after transplantation. Importantly, the absolute number and percentage of DNT cells were significantly higher in UCBT patients without aGVHD than in those with aGVHD within one year ( F = 4.684, P = 0.039 and F = 5.583, P = 0.026, respectively). In addition, the number of DNT cells in the first month after transplantation decreased significantly with the degree of aGVHD increased, and faster DNT cell reconstitution in the first month after UCBT was an independent protective factor for aGVHD (HR = 0.46, 95% confidence interval [CI]: 0.23-0.93; P = 0.031). CONCLUSIONS Compared to the number of DNT cells in Chinese healthy people, the reconstitution of DNT cells in adults with hematological malignancies after UCBT was slow. In addition, the faster reconstitution of DNT cells in the early stage after transplantation was associated with a lower incidence of aGVHD.
Collapse
Affiliation(s)
- Tianzhong Pan
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Peng Ding
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Aijie Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Baolin Tang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Kaidi Song
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Guangyu Sun
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yue Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shiying Yang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xingchi Chen
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dongyao Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoyu Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, Anhui, 230001, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
11
|
Rodríguez-Hernández MÁ, Baena-Bustos M, Carneros D, Zurita-Palomo C, Muñoz-Pinillos P, Millán J, Padillo FJ, Smerdou C, von Kobbe C, Rose-John S, Bustos M. Targeting IL-6 trans-signalling by sgp130Fc attenuates severity in SARS-CoV-2 -infected mice and reduces endotheliopathy. EBioMedicine 2024; 103:105132. [PMID: 38677182 PMCID: PMC11061249 DOI: 10.1016/j.ebiom.2024.105132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND SARS-CoV-2 infection is considered as a relapsing inflammatory process with a dysregulation of IL-6 signalling. Classic IL-6 signalling is thought to represent a defence mechanism against pathogens. In contrast, IL-6 trans-signalling has pro-inflammatory effects. In severe COVID-19, therapeutic strategies have focused on global inhibition of IL-6, with controversial results. We hypothesized that specific blockade of IL-6 trans-signalling could inhibit inflammatory response preserving the host defence activity inherent to IL-6 classic signalling. METHODS To test the role of the specific IL-6 trans-signalling inhibition by sgp130Fc in short- and long-term consequences of COVID-19, we used the established K18-hACE2 transgenic mouse model. Histological as well as immunohistochemical analysis, and pro-inflammatory marker profiling were performed. To investigate IL-6 trans-signalling in human cells we used primary lung microvascular endothelial cells and fibroblasts in the presence/absence of sgp130Fc. FINDINGS We report that targeting IL-6 trans-signalling by sgp130Fc attenuated SARS-CoV-2-related clinical symptoms and mortality. In surviving mice, the treatment caused a significant decrease in lung damage. In vitro, IL-6 trans-signalling induced strong and persisting JAK1/STAT3 activation in endothelial cells and lung fibroblasts with proinflammatory effects, which were attenuated by sgp130Fc. Our data also suggest that in those cells with scant amounts of IL-6R, the induction of gp130 and IL-6 by IL-6:sIL-6R complex sustains IL-6 trans-signalling. INTERPRETATION IL-6 trans-signalling fosters progression of COVID-19, and suggests that specific blockade of this signalling mode could offer a promising alternative to mitigate both short- and long-term consequences without affecting the beneficial effects of IL-6 classic signalling. These results have implications for the development of new therapies of lung injury and endotheliopathy in COVID-19. FUNDING The project was supported by ISCIII, Spain (COV-20/00792 to MB, PI23/01351 to MARH) and the European Commission-Next generation EU (European Union) (Regulation EU 2020/2094), through CSIC's Global Health Platform (PTI Salud Global, SGL2103029 to MB). PID2019-110587RB-I00 (MB) supported by MICIN/AEI/10.13039/501100011033/and PID2022-143034OB-I00 (MB) by MICIN/AEI/10.13039/501100011033/FEDER. MAR-H acknowledges support from ISCIII, Spain and the European Commission-Next generation EU (European Union), through CSIC's Global Health PTI.
Collapse
Affiliation(s)
- María Ángeles Rodríguez-Hernández
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain.
| | - Mercedes Baena-Bustos
- Pneumology Unit, Institute of Biomedicine of Seville (IBiS), Virgen Macarena University Hospital (HUVM), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - David Carneros
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Carola Zurita-Palomo
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Pablo Muñoz-Pinillos
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Francisco Javier Padillo
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), and CCUN, Pamplona, Spain
| | - Cayetano von Kobbe
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | | | - Matilde Bustos
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain.
| |
Collapse
|
12
|
Braga YLL, do Carmo Neto JR, Franco PIR, Helmo FR, Dos Reis MA, de Oliveira FA, Celes MRN, da Silva MV, Machado JR. The Influence of IL-11 on Cardiac Fibrosis in Experimental Models: A Systematic Review. J Cardiovasc Dev Dis 2024; 11:65. [PMID: 38392279 PMCID: PMC10888948 DOI: 10.3390/jcdd11020065] [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: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Fibrosis is one of the main factors that impair the function of many organs. In the heart, fibrosis leads to contractile dysfunction and arrhythmias, which are important in the development of heart failure. Interleukin (IL)-11 is regulated in various heart diseases and has recently been reported to be an important cytokine in fibrosis in this organ. However, this topic has been little explored, and many questions persist. Thus, this systematic review aimed to report on possible IL-11 therapies evaluated in rodent model-induced cardiac fibrosis. Inclusion criteria were experimental in vivo studies that used different rodent models for cardiac fibrosis associated with IL-11 interventions, without year and language restrictions. The search in PubMed, Web of Science, and Embase databases was performed in October 2022. The risk of bias assessment of the studies was based on the guidelines of the SYRCLE tool, and data from the selected articles were also presented in a table as a narrative description. This review was based on eight studies in which five different interventions were used: recombinant human IL-11 (rhIL-11), anti-IL11 (X203), recombinant mouse IL-11 (rmIL-11), lentivirus (LV)-IL-11 + lutein, and anti-IL11RA (X209). Based on the included studies, the results were variable, with IL-11 overexpression inducing cardiac fibrosis, while inhibition protected against this process, preserving the function of this organ. Therefore, IL-11 stands out as a promising therapeutic target for cardiac fibrosis. However, further studies are needed to understand the mechanisms triggered by each treatment, as well as its safety and immunogenicity.
Collapse
Affiliation(s)
- Yarlla Loyane Lira Braga
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Pablo Igor Ribeiro Franco
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | | | | | - Flávia Aparecida de Oliveira
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Mara Rúbia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Marcos Vinícius da Silva
- Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Juliana Reis Machado
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
- General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| |
Collapse
|
13
|
Luo X, He X, Zhang X, Zhao X, Zhang Y, Shi Y, Hua S. Hepatocellular carcinoma: signaling pathways, targeted therapy, and immunotherapy. MedComm (Beijing) 2024; 5:e474. [PMID: 38318160 PMCID: PMC10838672 DOI: 10.1002/mco2.474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 02/07/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer with a high mortality rate. It is regarded as a significant public health issue because of its complicated pathophysiology, high metastasis, and recurrence rates. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Traditional treatment methods such as surgical resection, radiotherapy, chemotherapy, and interventional therapies have limited therapeutic effects for HCC patients with recurrence or metastasis. With the development of molecular biology and immunology, molecular signaling pathways and immune checkpoint were identified as the main mechanism of HCC progression. Targeting these molecules has become a new direction for the treatment of HCC. At present, the combination of targeted drugs and immune checkpoint inhibitors is the first choice for advanced HCC patients. In this review, we mainly focus on the cutting-edge research of signaling pathways and corresponding targeted therapy and immunotherapy in HCC. It is of great significance to comprehensively understand the pathogenesis of HCC, search for potential therapeutic targets, and optimize the treatment strategies of HCC.
Collapse
Affiliation(s)
- Xiaoting Luo
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Xin He
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Xingmei Zhang
- Department of NeurobiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Xiaohui Zhao
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Yuzhe Zhang
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Yusheng Shi
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Shengni Hua
- Department of Radiation OncologyZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| |
Collapse
|
14
|
Wang Z, Liu N, Yang Y, Tu Z. The novel mechanism facilitating chronic hepatitis B infection: immunometabolism and epigenetic modification reprogramming. Front Immunol 2024; 15:1349867. [PMID: 38288308 PMCID: PMC10822934 DOI: 10.3389/fimmu.2024.1349867] [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: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Hepatitis B Virus (HBV) infections pose a global public health challenge. Despite extensive research on this disease, the intricate mechanisms underlying persistent HBV infection require further in-depth elucidation. Recent studies have revealed the pivotal roles of immunometabolism and epigenetic reprogramming in chronic HBV infection. Immunometabolism have identified as the process, which link cell metabolic status with innate immunity functions in response to HBV infection, ultimately contributing to the immune system's inability to resolve Chronic Hepatitis B (CHB). Within hepatocytes, HBV replication leads to a stable viral covalently closed circular DNA (cccDNA) minichromosome located in the nucleus, and epigenetic modifications in cccDNA enable persistence of infection. Additionally, the accumulation or depletion of metabolites not only directly affects the function and homeostasis of immune cells but also serves as a substrate for regulating epigenetic modifications, subsequently influencing the expression of antiviral immune genes and facilitating the occurrence of sustained HBV infection. The interaction between immunometabolism and epigenetic modifications has led to a new research field, known as metabolic epigenomics, which may form a mutually reinforcing relationship with CHB. Herein, we review the recent studies on immunometabolism and epigenetic reprogramming in CHB infection and discuss the potential mechanisms of persistent HBV infection. A deeper understanding of these mechanisms will offer novel insights and targets for intervention strategies against chronic HBV infection, thereby providing new hope for the treatment of related diseases.
Collapse
Affiliation(s)
- Zhengmin Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Liu
- Institute of Epigenetic Medicine, First Hospital of Jilin University, Changchun, China
| | - Yang Yang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengkun Tu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
15
|
Mu L, Qi L, Long H, Huang J, Zhong Z, Shi X, Chen C, Ye Q. Photothermal Fibrous Chitosan/Polydopamine Sponge for Intraoperative Hemostasis and Prevention of Tumor Recurrence in Hepatocellular Carcinoma Resection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304053. [PMID: 38029340 PMCID: PMC10797464 DOI: 10.1002/advs.202304053] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/06/2023] [Indexed: 12/01/2023]
Abstract
Hepatectomy, a surgical procedure for liver cancer, is often plagued by high recurrence rates worldwide. The recurrence of liver cancer is primarily attributed to microlesions in the liver, changes in the immune microenvironment, and circulating tumor cells in the bloodstream. To address this issue, a novel intervention method that combines intraoperative hemostasis with mild photothermal therapy is proposed, which has the potential to ablate microlesions and improve the immune microenvironment simultaneously. Specifically, the integrated strategy is realized based on the fibrous chitosan/polydopamine sponge (CPDS), which is constructed from shearing-flow-induced oriented hybrid chitosan fibers and subsequent self-assembly of polydopamine. The CPDS demonstrates high elasticity, excellent water absorption, and photothermal conversion performance. The results confirm the efficient hemostatic properties of the fibrous CPDS in various bleeding models. Notably, in subcutaneous and orthotopic postoperative recurrence and metastasis models of hepatocellular carcinoma, the fibrous CPDS significantly inhibits local tumor recurrence and distant metastasis. Moreover, the combination with lenvatinib can substantially enhance the antitumor effect. This comprehensive treatment strategy offers new insights into hepatectomy of liver cancer, representing a promising approach for clinical management.
Collapse
Affiliation(s)
- Lanxin Mu
- National Quality Control Center for Donated Organ ProcurementHubei Key Laboratory of Medical Technology on TransplantationHubei Clinical Research Center for Natural Polymer Biological LiverHubei Engineering Center of Natural Polymer‐based Medical Materials, Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan UniversityWuhan430071China
| | - Luhe Qi
- School of Resource and Environmental ScienceHubei International Scientific and Technological Cooperation Base of Sustainable Resource and EnergyHubei Engineering Center of Natural Polymers‐Based Medical MaterialsHubei Biomass‐Resource Chemistry and Environmental Biotechnology Key LaboratoryWuhan UniversityWuhan430079China
| | - Haitao Long
- National Quality Control Center for Donated Organ ProcurementHubei Key Laboratory of Medical Technology on TransplantationHubei Clinical Research Center for Natural Polymer Biological LiverHubei Engineering Center of Natural Polymer‐based Medical Materials, Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan UniversityWuhan430071China
| | - Jing Huang
- School of Resource and Environmental ScienceHubei International Scientific and Technological Cooperation Base of Sustainable Resource and EnergyHubei Engineering Center of Natural Polymers‐Based Medical MaterialsHubei Biomass‐Resource Chemistry and Environmental Biotechnology Key LaboratoryWuhan UniversityWuhan430079China
| | - Zibiao Zhong
- National Quality Control Center for Donated Organ ProcurementHubei Key Laboratory of Medical Technology on TransplantationHubei Clinical Research Center for Natural Polymer Biological LiverHubei Engineering Center of Natural Polymer‐based Medical Materials, Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan UniversityWuhan430071China
| | - Xiaowen Shi
- School of Resource and Environmental ScienceHubei International Scientific and Technological Cooperation Base of Sustainable Resource and EnergyHubei Engineering Center of Natural Polymers‐Based Medical MaterialsHubei Biomass‐Resource Chemistry and Environmental Biotechnology Key LaboratoryWuhan UniversityWuhan430079China
| | - Chaoji Chen
- School of Resource and Environmental ScienceHubei International Scientific and Technological Cooperation Base of Sustainable Resource and EnergyHubei Engineering Center of Natural Polymers‐Based Medical MaterialsHubei Biomass‐Resource Chemistry and Environmental Biotechnology Key LaboratoryWuhan UniversityWuhan430079China
| | - Qifa Ye
- National Quality Control Center for Donated Organ ProcurementHubei Key Laboratory of Medical Technology on TransplantationHubei Clinical Research Center for Natural Polymer Biological LiverHubei Engineering Center of Natural Polymer‐based Medical Materials, Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan UniversityWuhan430071China
| |
Collapse
|
16
|
Maspero M, Yilmaz S, Cazzaniga B, Raj R, Ali K, Mazzaferro V, Schlegel A. The role of ischaemia-reperfusion injury and liver regeneration in hepatic tumour recurrence. JHEP Rep 2023; 5:100846. [PMID: 37771368 PMCID: PMC10523008 DOI: 10.1016/j.jhepr.2023.100846] [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: 02/17/2023] [Revised: 06/20/2023] [Accepted: 07/01/2023] [Indexed: 09/30/2023] Open
Abstract
The risk of cancer recurrence after liver surgery mainly depends on tumour biology, but preclinical and clinical evidence suggests that the degree of perioperative liver injury plays a role in creating a favourable microenvironment for tumour cell engraftment or proliferation of dormant micro-metastases. Understanding the contribution of perioperative liver injury to tumour recurrence is imperative, as these pathways are potentially actionable. In this review, we examine the key mechanisms of perioperative liver injury, which comprise mechanical handling and surgical stress, ischaemia-reperfusion injury, and parenchymal loss leading to liver regeneration. We explore how these processes can trigger downstream cascades leading to the activation of the immune system and the pro-inflammatory response, cellular proliferation, angiogenesis, anti-apoptotic signals, and release of circulating tumour cells. Finally, we discuss the novel therapies under investigation to decrease ischaemia-reperfusion injury and increase regeneration after liver surgery, including pharmaceutical agents, inflow modulation, and machine perfusion.
Collapse
Affiliation(s)
- Marianna Maspero
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
- General Surgery and Liver Transplantation Unit, IRCCS Istituto Tumori, Milan, Italy
| | - Sumeyye Yilmaz
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Beatrice Cazzaniga
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Roma Raj
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Khaled Ali
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vincenzo Mazzaferro
- General Surgery and Liver Transplantation Unit, IRCCS Istituto Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Andrea Schlegel
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
17
|
Wu J, Chan YT, Lu Y, Wang N, Feng Y. The tumor microenvironment in the postsurgical liver: Mechanisms and potential targets of postoperative recurrence in human hepatocellular carcinoma. Med Res Rev 2023; 43:1946-1973. [PMID: 37102365 DOI: 10.1002/med.21967] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Surgery remains to be the mainstay of treatment for hepatocellular carcinoma (HCC). Nonetheless, its therapeutic efficacy is significantly impaired by postoperative recurrence, which occurs in more than half of cases as a result of intrahepatic metastasis or de novo tumorigenesis. For decades, most therapeutic strategies on inhibiting postoperative HCC recurrence have been focused on the residual tumor cells but satisfying therapeutic outcomes are barely observed in the clinic. In recent years, a better understanding of tumor biology allows us to shift our focus from tumor cells toward the postoperative tumor microenvironment (TME), which is gradually identified to play a pivotal role in tumor recurrence. In this review, we describe various surgical stress and surgical perturbation on postoperative TME. Besides, we discuss how such alternations in TME give rise to postoperative recurrence of HCC. Based on its clinical significance, we additionally highlight the potential of the postoperative TME as a target for postoperative adjuvant therapeutics.
Collapse
Affiliation(s)
- Junyu Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanjun Lu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Zhang Y, Wang N, Hao F, Chen Y, Fei X, Wang J. Attenuation of binuclear hepatocytes in the paracancerous liver tissue is associated with short-term recurrence of hepatocellular carcinoma post-radical surgery. FASEB J 2023; 37:e23271. [PMID: 37882195 DOI: 10.1096/fj.202301219r] [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: 06/18/2023] [Revised: 09/03/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
Short-term recurrence of hepatocellular carcinoma (HCC) after radical resection leads to dismal outcomes. To screen high-recurrence risk patients to provide adjuvant treatment is necessary. Herein, based on our previous research, we further focused on the changes in the abundance of binuclear hepatocytes (ABH) in the paracancerous liver tissue to discuss the relationship between the attenuation of binuclear hepatocytes and postoperative short-term recurrence, by combining with the assessment of the value of a reported independent early recurrence risk factor in HCC, protein induced by vitamin K absence or antagonist-II (PIVKA-II). A cohort of 142 paracancerous liver tissues from HCC patients who received radical resection was collected. Binuclear hepatocytes were reduced in the paracancerous liver tissues, compared with the liver tissues from normal donors. ABH was negatively correlated with clinical features such as tumor size, TNM stages, tumor microsatellite formation, venous invasion, and Alpha-fetoprotein (AFP) level, as well as the expression of E2F7 and Anillin, which are two critical regulators concerning the hepatocyte polyploidization. According to the short-term recurrence information, ABH value was laminated, and univariate and multivariate logistic regression was performed to analyze the relationship between paracancerous ABH and short-term tumor relapse. Simultaneously, the predictive effectiveness of the ABH value was compared with the preoperative PIVKA-II value. As observed, the paracancerous ABH value below 1.5% was found to be an independent risk factor for recurrence. In conclusion, the paracancerous ABH is a credible indicator of short-term recurrence of HCC patients after radical resection, and regular assessment of ABH might help to prevent short-term HCC recurrence.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Nan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| |
Collapse
|
19
|
Liu Y, Tian C, Zhang C, Liu Z, Li J, Li Y, Zhang Q, Ma S, Jiao D, Han X, Zhao Y. "One-stop" synergistic strategy for hepatocellular carcinoma postoperative recurrence. Mater Today Bio 2023; 22:100746. [PMID: 37564266 PMCID: PMC10410525 DOI: 10.1016/j.mtbio.2023.100746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Residual tumor recurrence after surgical resection of hepatocellular carcinoma (HCC) remains a considerable challenge that imperils the prognosis of patients. Notably, intraoperative bleeding and postoperative infection are potential risk factors for tumor recurrence. However, the biomaterial strategy for the above problems has rarely been reported. Herein, a series of cryogels (coded as SQ-n) based on sodium alginate (SA) and quaternized chitosan (QC) were synthesized and selected for optimal ratios. The in vitro assays showed that SQ-50 possessed superior hemostasis, excellent antibacterial property, and great cytocompatibility. Subsequently, SQAP was constructed by loading black phosphorus nanosheets (BPNSs) and anlotinib hydrochloride (AL3818) based on SQ-50. Physicochemical experiments confirmed that near-infrared (NIR)-assisted SQAP could control the release of AL3818 in photothermal response, significantly inhibiting the proliferation and survival of HUVECs and H22 cells. Furthermore, in vivo studies indicated that the NIR-assisted SQAP prevented local recurrence of ectopic HCC after surgical resection, achieved through the synergistic effect of mPTT and molecular targeted therapy. Thus, the multifunctional SQAP provides a "one-stop" synergistic strategy for HCC postoperative recurrence, showing great potential for clinical application.
Collapse
Affiliation(s)
- Yiming Liu
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Chuan Tian
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
- Department of Interventional Medical Center, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Shandong, 266000, Qingdao, PR China
| | - Chengzhi Zhang
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zaoqu Liu
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Jing Li
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yahua Li
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Quanhui Zhang
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Shengnan Ma
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, PR China
| | - Dechao Jiao
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xinwei Han
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yanan Zhao
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| |
Collapse
|
20
|
Hu Y, Huang Y, Xie X, Li L, Zhang Y, Zhang X. ARF6 promotes hepatocellular carcinoma proliferation through activating STAT3 signaling. Cancer Cell Int 2023; 23:205. [PMID: 37716993 PMCID: PMC10505330 DOI: 10.1186/s12935-023-03053-y] [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: 04/03/2023] [Accepted: 09/03/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) possesses the high mortality in cancers worldwide. Nevertheless, the concrete mechanism underlying HCC proliferation remains obscure. In this study, we show that high expression of ARF6 is associated with a poor clinical prognosis, which could boost the proliferation of HCC. METHODS Immunohistochemistry and western blotting were used to detect the expression level of ARF6 in HCC tissues. We analyzed the clinical significance of ARF6 in primary HCC patients. We estimated the effect of ARF6 on tumor proliferation with in vitro CCK8, colony formation assay, and in vivo nude mouse xenograft models. Immunofluorescence was conducted to investigate the ARF6 localization. western blotting was used to detect the cell cycle-related proteins with. Additionally, we examined the correlation between ARF6 and STAT3 signaling in HCC with western blotting, immunohistochemistry and xenograft assay. RESULTS ARF6 was upregulated in HCC tissues compared to adjacent normal liver tissues. The increased expression of ARF6 correlated with poor tumor differentiation, incomplete tumor encapsulation, advanced tumor TNM stage and poor prognosis. ARF6 obviously promoted HCC cell proliferation, colony formation, and cell cycle progression. In vivo nude mouse xenograft models showed that ARF6 enhanced tumor growth. Furthermore, ARF6 activated the STAT3 signaling and ARF6 expression was positively correlated with phosphorylated STAT3 level in HCC tissues. Furthermore, after intervening of STAT3, the effect of ARF6 on tumor-promoting was weakened, which demonstrated ARF6 functioned through STAT3 signaling in HCC. CONCLUSIONS Our results indicate that ARF6 promotes HCC proliferation through activating STAT3 signaling, suggesting that ARF6 may serve as potential prognostic and therapeutic targets for HCC patients.
Collapse
Affiliation(s)
- Yabing Hu
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, China
| | - Yongchu Huang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohang Xie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longshan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochao Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Qiu X, Zhou J, Xu H, Li Y, Ma S, Qiao H, Zeng K, Wang Q, Ouyang J, Liu Y, Ding J, Liu Y, Zhang J, Shi M, Liao Y, Liao W, Lin L. Alcohol reshapes a liver premetastatic niche for cancer by extra- and intrahepatic crosstalk-mediated immune evasion. Mol Ther 2023; 31:2662-2680. [PMID: 37469143 PMCID: PMC10492032 DOI: 10.1016/j.ymthe.2023.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer metastatic organotropism is still a mystery. The liver is known to be susceptible to cancer metastasis and alcoholic injury. However, it is unclear whether and how alcohol facilitates liver metastasis and how to intervene. Here, we show that alcohol preferentially promotes liver metastasis in colon-cancer-bearing mice and post-surgery pancreatic cancer patients. The mechanism is that alcohol triggers an extra- and intrahepatic crosstalk to reshape an immunosuppressive liver microenvironment. In detail, alcohol upregulates extrahepatic IL-6 and hepatocellular IL-6 receptor expression, resulting in hepatocyte STAT3 signaling activation and downstream lipocalin-2 (Lcn2) upregulation. Furthermore, LCN2 promotes T cell-exhaustion neutrophil recruitment and cancer cell epithelial plasticity. In contrast, knocking out hepatocellular Stat3 or systemic Il6 in alcohol-treated mice preserves the liver microenvironment and suppresses liver metastasis. This mechanism is reflected in hepatocellular carcinoma patients, in that alcohol-associated signaling elevation in noncancerous liver tissue indicates adverse prognosis. Accordingly, we discover a novel application for BBI608, a small molecular STAT3 inhibitor that can prevent liver metastasis. BBI608 pretreatment protects the liver and suppresses alcohol-triggered premetastatic niche formation. In conclusion, under extra- and intrahepatic crosstalk, the alcoholic injured liver forms a favorable niche for cancer cell metastasis, while BBI608 is a promising anti-metastatic agent targeting such microenvironments.
Collapse
Affiliation(s)
- Xiaofang Qiu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiaqi Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hong Xu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yongyin Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shudong Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hang Qiao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kangxin Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiongqiong Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiahe Ouyang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuanhan Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Ding
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yantan Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junhao Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
22
|
Hu Z, Zhao Y, Li L, Jiang J, Li W, Mang Y, Gao Y, Dong Y, Zhu J, Yang C, Ran J, Li L, Zhang S. Metformin promotes ferroptosis and sensitivity to sorafenib in hepatocellular carcinoma cells via ATF4/STAT3. Mol Biol Rep 2023; 50:6399-6413. [PMID: 37326750 PMCID: PMC10374833 DOI: 10.1007/s11033-023-08492-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/28/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common cancer worldwide, and sorafenib is a first-line drug for the treatment of advanced liver cancer. Resistance to sorafenib has become a major challenge in the treatment of hepatocellular carcinoma, however, studies have shown that metformin can promote ferroptosis and sorafenib sensitivity. Therefore, the aim of this study was to investigate the promotion of ferroptosis and sorafenib sensitivity by metformin via ATF4/STAT3 in hepatocellular carcinoma cells. METHODS Hepatocellular carcinoma cells Huh7 and Hep3B and induced sorafenib resistance (SR) Huh7/SR and Hep3B/SR cells were used as in vitro cell models. Cells were injected subcutaneously to establish a drug-resistant mouse model. CCK-8 was used to detect cell viability and sorafenib IC50. Western blotting was used to detect the expression of relevant proteins. BODIPY staining was used to analyze the lipid peroxidation level in cells. A scratch assay was used to detect cell migration. Transwell assays were used to detect cell invasion. Immunofluorescence was used to localize the expression of ATF4 and STAT3. RESULTS Metformin promoted ferroptosis in hepatocellular carcinoma cells through ATF4/STAT3, decreased sorafenib IC50, increased ROS and lipid peroxidation levels, decreased cell migration and invasion, inhibited the expression of the drug-resistant proteins ABCG2 and P-GP in hepatocellular carcinoma cells, and thus inhibited sorafenib resistance in hepatocellular carcinoma cells. Downregulating ATF4 inhibited the phosphorylated nuclear translocation of STAT3, promoted ferroptosis, and increased the sensitivity of Huh7 cells to sorafenib. Metformin was also shown in animal models to promote ferroptosis and sorafenib sensitivity in vivo via ATF4/STAT3. CONCLUSION Metformin promotes ferroptosis and sensitivity to sorafenib in hepatocellular carcinoma cells via ATF4/STAT3, and it inhibits HCC progression.
Collapse
Affiliation(s)
- Zongqiang Hu
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yingpeng Zhao
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Laibang Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jie Jiang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Wang Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yuanyi Mang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yang Gao
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yun Dong
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jiashun Zhu
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Chaomin Yang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jianghua Ran
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| | - Li Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| | - Shengning Zhang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
23
|
Li J, Wang X, Ren M, He S, Zhao Y. Advances in experimental animal models of hepatocellular carcinoma. Cancer Med 2023; 12:15261-15276. [PMID: 37248746 PMCID: PMC10417182 DOI: 10.1002/cam4.6163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with insidious early symptoms, easy metastasis, postoperative recurrence, poor drug efficacy, and a high drug resistance rate when surgery is missed, leading to a low 5-year survival rate. Research on the pathogenesis and drugs is particularly important for clinical treatment. Animal models are crucial for basic research, which is conducive to studying pathogenesis and drug screening more conveniently and effectively. An appropriate animal model can better reflect disease occurrence and development, and the process of anti-tumor immune response in the human body. This review summarizes the classification, characteristics, and advances in experimental animal models of HCC to provide a reference for researchers on model selection.
Collapse
Affiliation(s)
- Jing Li
- Department of GastroenterologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Xin Wang
- Department of GastroenterologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Mudan Ren
- Department of GastroenterologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Shuixiang He
- Department of GastroenterologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Yan Zhao
- Department of GastroenterologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| |
Collapse
|
24
|
Wang S, Zhou L, Ji N, Sun C, Sun L, Sun J, Du Y, Zhang N, Li Y, Liu W, Lu W. Targeting ACYP1-mediated glycolysis reverses lenvatinib resistance and restricts hepatocellular carcinoma progression. Drug Resist Updat 2023; 69:100976. [PMID: 37210811 DOI: 10.1016/j.drup.2023.100976] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Acylphosphatase 1 (ACYP1), a protein located in the mammalian cell cytoplasm, has been shown to be associated with tumor initiation and progression by functioning as a metabolism-related gene. Here we explored the potential mechanisms by which ACYP1 regulates the development of HCC and participates in the resistance to lenvatinib. ACYP1 can promote the proliferation, invasion, and migration capacities of HCC cells in vitro and in vivo. RNA sequencing reveals that ACYP1 markedly enhances the expression of genes related to aerobic glycolysis, and LDHA is identified as the downstream gene of ACYP1. Overexpression of ACYP1 upregulates LDHA levels, which then increases the malignancy potential of HCC cells. GSEA data analysis reveals the enrichment of differentially expressed genes in the MYC pathway, indicating a positive correlation between MYC and ACYP1 levels. Mechanistically, ACYP1 exerts its tumor-promoting roles by regulating the Warburg effect through activating the MYC/LDHA axis. Mass spectrometry analysis and Co-IP assays confirm that ACYP1 can bind to HSP90. The regulation of c-Myc protein expression and stability by ACYP1 is HSP90 dependent. Importantly, lenvatinib resistance is associated with ACYP1, and targeting ACYP1 remarkably decreases lenvatinib resistance and inhibits progression of HCC tumors with high ACYP1 expression when combined with lenvatinib in vitro and in vivo. These results illustrate that ACYP1 has a direct regulatory role in glycolysis and drives lenvatinib resistance and HCC progression via the ACYP1/HSP90/MYC/LDHA axis. Targeting ACYP1 could synergize with lenvatinib to treat HCC more effectively.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China; Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lingyi Zhou
- Department of Blood Transfusion, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Ning Ji
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China
| | - Chengtao Sun
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong, PR China
| | - Linlin Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China
| | - Jiao Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China
| | - Yawei Du
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China
| | - Ningning Zhang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China.
| | - Yueguo Li
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China
| | - Weishuai Liu
- Department of Pain Relief, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin 300060, PR China.
| | - Wei Lu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin 300060, PR China.
| |
Collapse
|
25
|
Jin J, Bai L, Wang D, Ding W, Cao Z, Yan P, Li Y, Xi L, Wang Y, Zheng X, Wei H, Ding C, Wang Y. SIRT3-dependent delactylation of cyclin E2 prevents hepatocellular carcinoma growth. EMBO Rep 2023; 24:e56052. [PMID: 36896611 PMCID: PMC10157311 DOI: 10.15252/embr.202256052] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Lysine lactylation (Kla) is a recently discovered histone mark derived from metabolic lactate. The NAD+ -dependent deacetylase SIRT3, which can also catalyze removal of the lactyl moiety from lysine, is expressed at low levels in hepatocellular carcinoma (HCC) and has been suggested to be an HCC tumor suppressor. Here we report that SIRT3 can delactylate non-histone proteins and suppress HCC development. Using SILAC-based quantitative proteomics, we identify cyclin E2 (CCNE2) as one of the lactylated substrates of SIRT3 in HCC cells. Furthermore, our crystallographic study elucidates the mechanism of CCNE2 K348la delactylation by SIRT3. Our results further suggest that lactylated CCNE2 promotes HCC cell growth, while SIRT3 activation by Honokiol induces HCC cell apoptosis and prevents HCC outgrowth in vivo by regulating Kla levels of CCNE2. Together, our results establish a physiological function of SIRT3 as a delactylase that is important for suppressing HCC, and our structural data could be useful for the future design of activators.
Collapse
Affiliation(s)
- Jing Jin
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Lin Bai
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Human Phenome Institute, School of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Dongyao Wang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Wei Ding
- Beijing National Laboratory for Condensed Matter Physics, Institute of PhysicsChinese Academy of SciencesBeijingChina
| | - Zhuoxian Cao
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Peidong Yan
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Yunjia Li
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Lulu Xi
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Yuxin Wang
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Xiaohu Zheng
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Human Phenome Institute, School of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yi Wang
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
26
|
Gao YX, Ning QQ, Yang PX, Guan YY, Liu PX, Liu ML, Qiao LX, Guo XH, Yang TW, Chen DX. Recent advances in recurrent hepatocellular carcinoma therapy. World J Hepatol 2023; 15:460-476. [PMID: 37206651 PMCID: PMC10190692 DOI: 10.4254/wjh.v15.i4.460] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer, accounting for 75%-85% of cases. Although treatments are given to cure early-stage HCC, up to 50%-70% of individuals may experience a relapse of the illness in the liver after 5 years. Research on the fundamental treatment modalities for recurrent HCC is moving significantly further. The precise selection of individuals for therapy strategies with established survival advantages is crucial to ensuring better outcomes. These strategies aim to minimize substantial morbidity, support good life quality, and enhance survival for patients with recurrent HCC. For individuals with recurring HCC after curative treatment, no approved therapeutic regimen is currently available. A recent study presented novel approaches, like immunotherapy and antiviral medication, to improve the prognosis of patients with recurring HCC with the apparent lack of data to guide the clinical treatment. The data supporting several neoadjuvant and adjuvant therapies for patients with recurring HCC are outlined in this review. We also discuss the potential for future clinical and translational investigations.
Collapse
Affiliation(s)
- Yu-Xue Gao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Qi-Qi Ning
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Peng-Xiang Yang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Yuan-Yue Guan
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Peng-Xiang Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Meng-Lu Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Lu-Xin Qiao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Xiang-Hua Guo
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Tong-Wang Yang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan Province, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, Hunan Province, China
| | - De-Xi Chen
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| |
Collapse
|
27
|
Wang H, Yang C, Li D, Wang R, Li Y, Lv L. Bioinformatics analysis and experimental validation of a novel autophagy-related signature relevant to immune infiltration for recurrence prediction after curative hepatectomy. Aging (Albany NY) 2023; 15:2610-2630. [PMID: 37014321 PMCID: PMC10120910 DOI: 10.18632/aging.204632] [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: 01/31/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Hepatocellular carcinoma (HCC) remains imposing an enormous economic and healthcare burden worldwide. In this present study, we constructed and validated a novel autophagy-related gene signature to predict the recurrence of HCC patients. A total of 29 autophagy-related differentially expressed genes were identified. A five-gene signature (CLN3, HGF, TRIM22, SNRPD1, and SNRPE) was constructed for HCC recurrence prediction. Patients in high-risk groups exhibited a significantly poor prognosis compared with low-risk patients both in the training set (GSE14520 dataset) and the validation set (TCGA and GSE76427 dataset). Multivariate cox regression analysis demonstrated that the 5-gene signature was an independent risk factor for recurrence-free survival (RFS) in HCC patients. The nomograms incorporating 5-gene signature and clinical prognostic risk factors were able to effectively predict RFS. KEGG and GSEA analysis revealed that the high-risk group was enriched with multiple oncology characteristics and invasive-related pathways. Besides, the high-risk group had a higher level of immune cells and higher levels of immune checkpoint-related gene expression in the tumor microenvironment, suggesting that they might be more likely to benefit from immunotherapy. Finally, the immunohistochemistry and cell experiments confirmed the role of SNRPE, the most significant gene in the gene signature. SNRPE was significantly overexpressed in HCC. After SNRPE knockdown, the proliferation, migration and invasion ability of the HepG2 cell line were significantly inhibited. Our study established a novel five-gene signature and nomogram to predict RFS of HCC, which may help in clinical decision-making for individual treatment.
Collapse
Affiliation(s)
- Huaxiang Wang
- Department of Hepatobiliary Surgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350025, China
- Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistic Team, Fuzhou, Fujian 350025, China
- Department of Hepatobiliary and pancreatic Surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Chengkai Yang
- Department of Hepatobiliary Surgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350025, China
- Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistic Team, Fuzhou, Fujian 350025, China
| | - Dong Li
- Department of Anesthesiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China
| | - Ruling Wang
- Department of Hepatobiliary and pancreatic Surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yanbing Li
- Department of Hepatobiliary and pancreatic Surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lizhi Lv
- Department of Hepatobiliary Surgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350025, China
- Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistic Team, Fuzhou, Fujian 350025, China
| |
Collapse
|
28
|
Ait-Ahmed Y, Lafdil F. Novel insights into the impact of liver inflammatory responses on primary liver cancer development. LIVER RESEARCH 2023; 7:26-34. [PMID: 39959704 PMCID: PMC11791919 DOI: 10.1016/j.livres.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/05/2022] [Accepted: 01/27/2023] [Indexed: 03/10/2023]
Abstract
Primary liver cancers rank among the deadliest cancers worldwide and often develop in patients with chronic liver diseases in an inflammatory context. This review highlights recent reports on the mechanisms of inflammatory-mediated hepatic cell transformation that trigger the tumorigenic process (initiation steps) and the impact of the immune response favoring tumor cell expansion (progression steps). Several cytokines, namely interleukin (IL)-6, IL-17, IL-1beta, and tumor necrosis factor-alpha, have been described to play a prominent role in the initiation of liver cancers. Additionally, inflammation contributes to cancer progression by favoring tumor escape from anti-tumor immune response, angiogenesis, and metastasis through tumor growth factor-beta and matrix metalloprotease upregulation. These recent studies allowed the development of novel therapeutic strategies aiming at regulating liver inflammation. These strategies are based on the use of anti-inflammatory agents, antibodies targeting immune checkpoint molecules such as programmed death ligand 1 and molecules targeting angiogenic factors, metastasis key factors, and microRNAs involved in tumor development. This review aims at summarizing the recent studies reporting different mechanisms by which the liver inflammatory responses could contribute to liver cancer development.
Collapse
Affiliation(s)
- Yeni Ait-Ahmed
- Université Paris-Est, UMR-S955, UPEC, Créteil, France
- Institut National de la Sante et de la Recherche Medicale (INSERM), U955, Créteil, France
| | - Fouad Lafdil
- Université Paris-Est, UMR-S955, UPEC, Créteil, France
- Institut National de la Sante et de la Recherche Medicale (INSERM), U955, Créteil, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
29
|
Sun S, Shen B, Wang N. Concurrent chemoradiotherapy improves prognosis and quality of life of patients with metastatic and recurrent hepatocellular carcinoma. Am J Transl Res 2023; 15:233-240. [PMID: 36777829 PMCID: PMC9908489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/30/2022] [Indexed: 02/14/2023]
Abstract
OBJECTIVE This study retrospectively analyzed the effect of concurrent chemoradiotherapy on prognosis and quality of life of patients with metastatic and recurrent hepatocellular carcinoma (HCC). METHODS This is a retrospective analysis. Data from 60 patients with metastatic and recurrent HCC admitted from Oct. 2020 to Feb. 2021 were chosen and grouped according to the treatment plans. Each group contained 30 cases. The control group was treated with chemotherapy, and the observation group received concurrent chemoradiotherapy. The two groups were treated continuously for two rounds, with 21 days in each round. The therapeutic efficacy, toxic side effects, pre- and post-treatment quality of life, changes in vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2), and survival during follow-up were compared between the two groups. RESULTS The total therapeutic efficacy of the observation group was higher than that of the control group (P<0.05). The post-treatment Karnofsky score in the observation group was higher than that in the control group (P<0.05). The post-treatment protein expressions of VEGF and COX-2 and peripheral blood mononuclear cells were lower than those before treatment in the two groups (P<0.05), and were lower in the observation group than those in the control group (P<0.05). The observation group had superior survival times than the control group (P<0.05). CONCLUSION Concurrent chemoradiotherapy has good short-term and long-term efficacy for patients with metastatic and recurrent HCC. It greatly improves patients' quality of life and down-regulates VEGF and COX-2 expression.
Collapse
Affiliation(s)
- Shugang Sun
- Department of General Surgery, Affiliated Hospital of Hebei Engineering UniversityHandan 056000, Hebei, China
| | - Bin Shen
- Department of General Surgery, Affiliated Hospital of Hebei Engineering UniversityHandan 056000, Hebei, China
| | - Ning Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei Engineering UniversityHandan 056000, Hebei, China
| |
Collapse
|
30
|
Yang Z, Xu H, Yang Y, Duan C, Zhang P, Wang Y, Fu K, Shen Y, Xu MX. Synthesis and evaluation of naphthalene derivatives as potent STAT3 inhibitors and agents against triple-negative breast cancer growth and metastasis. Breast Cancer Res Treat 2023; 197:255-267. [PMID: 36369502 DOI: 10.1007/s10549-022-06790-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) represents the worst prognostic subtype of breast cancer and lacks targeted therapeutic drugs. Signal transducer and activator of transcription 3 (STAT3) is overexpressed and constitutively activated in TNBCs and associated with poor patient outcomes. However, no agents targeting STAT3 have been successfully developed and marketed. Selective Estrogen Receptor Modulators (SERMs) have been reported as potential inhibitors of the IL-6/STAT3 signaling pathway. Naphthalene compounds have good pharmacological activity and significant anti-cancer activity. In this study, we synthesized a new series of naphthalene derivatives with the general structure of SERM and evaluated their effects on TNBC and STAT3 signals. METHODS A new series of compounds based on the scaffold of SERMs and an amino group were designed and screened based on the structure-activity relationship by MTT assay. The binding activity of SMY002 to STAT3 was predicted and validated by docking and SPR. The STAT3 signaling target and anti-cancer effects of SMY002 were evaluated with three TNBC cell lines and the mice transplanted tumor model. RESULTS Among the compounds, SMY002 displayed the most potent activity, which could directly interact with STAT3 SH2-domain, and strongly inhibit the phosphorylation, dimerization, nuclear distribution, transcriptional activity, and target genes expression of STAT3. Furthermore, SMY002 markedly suppressed migration, invasion, survival, growth, and metastasis of TNBC cells in vitro and in vivo via down-regulating the expression of Cyclin D1 and MMP9. CONCLUSIONS SMY002 can significantly inhibit the growth and metastasis of TNBC cells by targeting the STAT3 signal.
Collapse
Affiliation(s)
- Zhengyan Yang
- Henan International Joint Lab for Anti-Cancer Drug Design Based On Biological Target and Drug Discovery, Henan Ruida Bio-Tech Medicine Co. Ltd, Kaifeng, 475000, China
- School of Basic Medical Science, Henan University, Kaifeng, 475004, China
| | - Hongyun Xu
- Henan International Joint Lab for Anti-Cancer Drug Design Based On Biological Target and Drug Discovery, Henan Ruida Bio-Tech Medicine Co. Ltd, Kaifeng, 475000, China
| | - Yupo Yang
- Henan International Joint Lab for Anti-Cancer Drug Design Based On Biological Target and Drug Discovery, Henan Ruida Bio-Tech Medicine Co. Ltd, Kaifeng, 475000, China
| | - Chaoqun Duan
- Henan International Joint Lab for Anti-Cancer Drug Design Based On Biological Target and Drug Discovery, Henan Ruida Bio-Tech Medicine Co. Ltd, Kaifeng, 475000, China
| | - Pai Zhang
- School of Basic Medical Science, Henan University, Kaifeng, 475004, China
| | - Yang Wang
- Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Kai Fu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York, 14263-0001, USA
| | - Yonghong Shen
- Institute of Microbial Engineering, Engineering Research Center for Applied Microbiology of Henan Province, School of Life Sciences of Henan University, Kaifeng, 475004, China.
| | - Marvin Xuejun Xu
- Henan International Joint Lab for Anti-Cancer Drug Design Based On Biological Target and Drug Discovery, Henan Ruida Bio-Tech Medicine Co. Ltd, Kaifeng, 475000, China.
- Translational Medical Center, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, China.
| |
Collapse
|
31
|
Cao Q, Yang L, Zhou G, Hu Y. Clinical efficacy of laparoscopic radical hepatectomy and laparotomy for hepatocellular carcinoma and factors of postoperative recurrence. Front Oncol 2023; 13:1116984. [PMID: 37035189 PMCID: PMC10073724 DOI: 10.3389/fonc.2023.1116984] [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: 12/06/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Objective For exploring the clinical efficacy of laparoscopic radical hepatectomy and laparotomy for hepatocellular carcinoma and analysing related factors of postoperative recurrence. Methods Totally 212 patients with hepatocellular carcinoma admitted to our hospital between April 2017 and December 2020 were enrolled, and all of them were followed up after the operation. According to the treatment modes, the patients were assigned to a laparotomy group (n=106) and a laparoscopic group (n=106). Perioperative indicators, haematological examination results, complications and recurrence were compared between the two groups. The recurrence time of hepatocellular carcinoma after the operation was confirmed by imaging examination with definite mass, and logistic multivariate analysis was used for analyzing the risk factors associated with postoperative recurrence. Results Patients in both groups were comparable in terms of general baseline data. The laparoscopic group experienced longer operation time and shorter incision length, less intraoperative blood loss, early time to have the first off-bed activity and time to eat liquid and shorter hospital stay than the laparotomy group (all P<0.05). Seven days after the operation, the laparoscopic group showed notably lower levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), tumour necrosis factor-α (TNF-α), interleukin-8 (IL-8), and C-reactive protein (CRP) than the laparotomy group (all P<0.05), and also showed notably higher levels of immunoglobulin A (IgA), IgG and IgM than the laparotomy group (all P<0.05). Additionally, the laparotomy group showed a higher total incidence rate of complications than the laparoscopic group (19.81% vs. 9.43%, P<0.05). During the one-year follow-up, the laparotomy group was not greatly different from the laparoscopic group in recurrence rate (22.64% (24/106) vs. 16.98% (18/106), P>0.05). Multivariate analysis showed that average tumour diameter and microvascular invasion were risk factors for postoperative recurrence (P<0.05). Conclusion Laparoscopic radical hepatectomy for hepatocellular carcinoma can reduce the influence on liver function and immune function, with less damage to tissues, and can ameliorate postoperative inflammatory reaction, and promote postoperative recovery of patients as soon as possible. There are many factors influencing the postoperative recurrence of hepatocellular carcinoma, and average tumour diameter and microvascular invasion are the risk factors.
Collapse
|
32
|
Wang D, Sun Z, Zhu X, Zheng X, Zhou Y, Lu Y, Yan P, Wang H, Liu H, Jin J, Zhu H, Sun R, Wang Y, Fu B, Tian Z, Wei H. GARP-mediated active TGF-β1 induces bone marrow NK cell dysfunction in AML patients with early relapse post-allo-HSCT. Blood 2022; 140:2788-2804. [PMID: 35981475 PMCID: PMC10653097 DOI: 10.1182/blood.2022015474] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 01/05/2023] Open
Abstract
Relapse is a leading cause of death after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for acute myeloid leukemia (AML). However, the underlying mechanisms remain poorly understood. Natural killer (NK) cells play a crucial role in tumor surveillance and cancer immunotherapy, and NK cell dysfunction has been observed in various tumors. Here, we performed ex vivo experiments to systematically characterize the mechanisms underlying the dysfunction of bone marrow-derived NK (BMNK) cells isolated from AML patients experiencing early relapse after allo-HSCT. We demonstrated that higher levels of active transforming growth factor β1 (TGF-β1) were associated with impaired effector function of BMNK cells in these AML patients. TGF-β1 activation was induced by the overexpression of glycoprotein A repetitions predominant on the surface of CD4+ T cells. Active TGF-β1 significantly suppressed mTORC1 activity, mitochondrial oxidative phosphorylation, the proliferation, and cytotoxicity of BMNK cells. Furthermore, pretreatment with the clinical stage TGF-β1 pathway inhibitor, galunisertib, significantly restored mTORC1 activity, mitochondrial homeostasis, and cytotoxicity. Importantly, the blockade of the TGF-β1 signaling improved the antitumor activity of NK cells in a leukemia xenograft mouse model. Thus, our findings reveal a mechanism explaining BMNK cell dysfunction and suggest that targeted inhibition of TGF-β1 signaling may represent a potential therapeutic intervention to improve outcomes in AML patients undergoing allo-HSCT or NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Dongyao Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Zimin Sun
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoyu Zhu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaohu Zheng
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yonggang Zhou
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yichen Lu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Peidong Yan
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huiru Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Huilan Liu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Jing Jin
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huaiping Zhu
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Wang
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Binqing Fu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
33
|
Huang B, Lang X, Li X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front Oncol 2022; 12:1023177. [PMID: 36591515 PMCID: PMC9800921 DOI: 10.3389/fonc.2022.1023177] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine involved in immune regulation. It can activate janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. As one of the important signal transduction pathways in cells, JAK2/STAT3 signaling pathway plays a critical role in cell proliferation and differentiation by affecting the activation state of downstream effector molecules. The activation of JAK2/STAT3 signaling pathway is involved in tumorigenesis and development. It contributes to the formation of tumor inflammatory microenvironment and is closely related to the occurrence and development of many human tumors. This article focuses on the relationship between IL-6/JAK2/STAT3 signaling pathway and liver cancer, breast cancer, colorectal cancer, gastric cancer, lung cancer, pancreatic cancer and ovarian cancer, hoping to provide references for the research of cancer treatment targeting key molecules in IL-6/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Bei Huang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoling Lang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| |
Collapse
|
34
|
Li J, Tang M, Wu J, Qu H, Tu M, Pan Z, Gao C, Yang Y, Qu C, Huang W, Hong J. NUSAP1, a novel stemness-related protein, promotes early recurrence of hepatocellular carcinoma. Cancer Sci 2022; 113:4165-4180. [PMID: 36106345 DOI: 10.1111/cas.15585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/15/2022] [Accepted: 09/07/2022] [Indexed: 01/27/2023] Open
Abstract
Early recurrence (within 2 years after resection) is the primary cause of poor outcomes among hepatocellular carcinoma (HCC) patients, and liver cancer stem cells are the main contributors to postsurgical HCC recurrence. Nucleolar and spindle-associated protein 1 (NUSAP1) has been reported to be involved in tumor progression. We investigated the function and clinical value of NUSAP1 in early recurrence of HCC. Data from public datasets and our cohort were used to assess the association between NUSAP1 expression and early HCC recurrence. Gain- and loss-of-function experiments were carried out in vivo and in vitro. The predictive effect of NUSAP1 on early HCC recurrence was further evaluated by a validation cohort. We found that elevated NUSAP1 expression in HCC specimens was correlated with poor outcome, especially in cases with postoperative early recurrence. Functional studies indicated that NUSAP1 significantly promotes HCC progression. A postsurgical recurrence murine model further revealed that upregulated NUSAP1 dramatically increased the likelihood of HCC early recurrence. RNA sequencing data revealed that the gene sets of cancer stemness and the signal transducer and activator of transcription 3 (STAT3) pathway were enriched by NUSAP1 overexpression. Mechanistically, NUSAP1 enhanced cancer stemness through stimulating STAT3 nuclear translocation and activation through receptor of activated protein C kinase 1 (RACK1). In a validation cohort with 112 HCC patients, NUSAP1 effectively predicted HCC early recurrence. Our results indicated that NUSAP1 promotes early recurrence of HCC by sustaining cancer stemness and could serve as a valuable predictive indicator for postsurgical intervention in HCC patients.
Collapse
Affiliation(s)
- Jinying Li
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Ming Tang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Junru Wu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hengdong Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Mengxian Tu
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhaojie Pan
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chongqing Gao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yuping Yang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chen Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Wei Huang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China.,Department of Hepatological Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
35
|
Wang H, Liu H, Zhou L, Wang D, Wang S, Liu Q, Wu Y, Tu M, Sun Z, Zheng X, Fu B, Wang B, Wei H. Cytomegalovirus-specific neutralizing antibodies effectively prevent uncontrolled infection after allogeneic hematopoietic stem cell transplantation. iScience 2022; 25:105065. [PMID: 36147955 PMCID: PMC9485910 DOI: 10.1016/j.isci.2022.105065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/06/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Cytomegalovirus (CMV) infection remains one of the most frequent and life-threatening infectious complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Herein, we comprehensively compared the immune cells of patients with uncontrolled and controlled CMV infection post-allo-HSCT and found that B-cells were extraordinarily insufficient because of impaired B-cells reconstitution in the uncontrolled infection group. Furthermore, in the controlled infection group, reconstructed B-cells showed signatures of mature B-cells, high expression of CXCR4 and IFITM1, and enrichment of CMV-associated B-cell receptors, which were lacking in the uncontrolled infection group. Consistently, sera from the uncontrolled infection group failed to inhibit CMV infection via neutralizing virus in vitro because of its lower content of anti-CMV-specific immunoglobulin G (IgG) than the controlled infection group. Overall, these results highlighted the contribution of B cells and anti-CMV-specific neutralizing IgGs to the restraint of CMV infection post-allo-HSCT, suggesting their potential as a supplementary treatment to improve outcomes.
Collapse
Affiliation(s)
- Huiru Wang
- Department of Transfusion, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Huilan Liu
- Department of Transfusion, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Li Zhou
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Dongyao Wang
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Shushu Wang
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qian Liu
- Organ Transplant Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Yun Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Meijuan Tu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zimin Sun
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaohu Zheng
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Binqing Fu
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Haiming Wei
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
36
|
Fibroblast Common Serum Response Signature-Related Classification Affects the Tumour Microenvironment and Predicts Prognosis in Bladder Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5645944. [PMID: 36312898 PMCID: PMC9606836 DOI: 10.1155/2022/5645944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022]
Abstract
Abnormal oncogenic signatures provide important clues regarding cancer prognosis and treatment. We analysed the variations in 189 oncogenic signature gene sets between normal and tumourous tissues from The Cancer Genome Atlas (TCGA) and found that the “CSR_LATE_UP” signature was the most upregulated oncogenic signature gene set in bladder cancer. Next, we developed a common serum response (CSR) risk score (CRS) model based on fibroblast CSR genes and systematically analysed the correlations of these genes or the CRSs with survival, previously reported molecular subtypes, clinicopathological features, cancer signalling pathways, chemotherapeutic responses, and the tumour microenvironment using TCGA and validation cohorts. The CRS could predict the malignant phenotype, chemotherapeutic efficacy, immune invasion, and disease prognosis. Inflammatory signalling pathways (e.g., inflammatory response, TNFA signalling via NFƘB, IFNα response, and IL2-STAT5 signalling) were markedly upregulated in patients with high CRS. Notably, the CSR-related gene ANLN was positively correlated with CD8+ immune cell infiltration, PD-L1 expression, and sensitivity to PD-L1 inhibitors and could thus provide guidance for clinical immunotherapy. This study highlights the crucial role of the CSR signature in bladder cancer and provides a CRS model for accurate predictions of the disease prognosis and chemotherapy and immunotherapy responses.
Collapse
|
37
|
Wang D, Fu B, Wei H. Advances in Immunotherapy for Hepatitis B. Pathogens 2022; 11:1116. [PMID: 36297173 PMCID: PMC9612046 DOI: 10.3390/pathogens11101116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2023] Open
Abstract
Hepatitis B virus (HBV) is a hepatotropic virus with the potential to cause chronic infection, and it is one of the common causes of liver disease worldwide. Chronic HBV infection leads to liver cirrhosis and, ultimately, hepatocellular carcinoma (HCC). The persistence of covalently closed circular DNA (cccDNA) and the impaired immune response in patients with chronic hepatitis B (CHB) has been studied over the past few decades. Despite advances in the etiology of HBV and the development of potent virus-suppressing regimens, a cure for HBV has not been found. Both the innate and adaptive branches of immunity contribute to viral eradication. However, immune exhaustion and evasion have been demonstrated during CHB infection, although our understanding of the mechanism is still evolving. Recently, the successful use of an antiviral drug for hepatitis C has greatly encouraged the search for a cure for hepatitis B, which likely requires an approach focused on improving the antiviral immune response. In this review, we discuss our current knowledge of the immunopathogenic mechanisms and immunobiology of HBV infection. In addition, we touch upon why the existing therapeutic approaches may not achieve the goal of a functional cure. We also propose how combinations of new drugs, and especially novel immunotherapies, contribute to HBV clearance.
Collapse
Affiliation(s)
- Dongyao Wang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China
- Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei 230001, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu 233030, China
| | - Binqing Fu
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China
| | - Haiming Wei
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
38
|
Liu Y, Liang W, Chang Y, He Z, Wu M, Zheng H, Ke X, Lv M, Liu Q, Liu Q, Tang W, Huang Q, Lu Y, He M, Yang Q, Mo C, Wang J, Peng K, Min Z, Su H, Chen J. CEP192 is a novel prognostic marker and correlates with the immune microenvironment in hepatocellular carcinoma. Front Immunol 2022; 13:950884. [PMID: 36238304 PMCID: PMC9551108 DOI: 10.3389/fimmu.2022.950884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) responds poorly to standard chemotherapy or targeted therapy; hence, exploration for novel therapeutic targets is urgently needed. CEP192 protein is indispensable for centrosome amplification, which has been extensively characterized in both hematological malignancies and solid tumors. Here, we combined bioinformatics and experimental approaches to assess the potential of CEP192 as a prognostic and therapeutic target in HCC. CEP192 expression increased with tumor stage and was associated with poor clinicopathologic features, frequent recurrence, and higher mortality. Upon single-cell RNA sequencing, CEP192 was found to be involved in the proliferation and self-renewal of hepatic progenitor-like cells. This observation was further evidenced using CEP192 silencing, which prevented tumor cell proliferation and self-renewal by arresting cells in the G0/G1 phase of the cell cycle. Notably, CEP192 was highly correlated with multiple tumor-associated cytokine ligand–receptor axes, including IL11–IL11RA, IL6–IL6R, and IL13–IL13RA1, which could promote interactions between hepatic progenitor-like cells, PLVAP+ endothelial cells, tumor-associated macrophages, and CD4+ T cells. Consequently, CEP192 expression was closely associated with an immunosuppressive tumor microenvironment and low immunophenoscores, making it a potential predictor of response to immune checkpoint inhibitors. Taken together, our results unravel a novel onco-immunological role of CEP192 in establishing the immunosuppressive tumor microenvironment and provide a novel biomarker, as well as a potential target for therapeutic intervention of HCC.
Collapse
Affiliation(s)
- Yanli Liu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wanmei Liang
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yabin Chang
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zehui He
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Meijian Wu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haozhi Zheng
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xinrong Ke
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Minjia Lv
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qingqian Liu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qinyu Liu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Waner Tang
- Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiaoling Huang
- Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yu Lu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Min He
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qijun Yang
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunpan Mo
- The Second Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Jiefan Wang
- The Second Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Kunwei Peng
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiqun Min
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hang Su
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingqi Chen
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology and Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jingqi Chen,
| |
Collapse
|
39
|
Wang A, Fang M, Jiang H, Wang D, Zhang X, Tang B, Zhu X, Hu W, Liu X. Palbociclib promotes the antitumor activity of Venetoclax plus Azacitidine against acute myeloid leukemia. Biomed Pharmacother 2022; 153:113527. [DOI: 10.1016/j.biopha.2022.113527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022] Open
|
40
|
Fu J, Chen F, Lin Y, Gao J, Chen A, Yang J. Discovery and characterization of tumor antigens in hepatocellular carcinoma for mRNA vaccine development. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04325-2. [PMID: 36038676 PMCID: PMC9423891 DOI: 10.1007/s00432-022-04325-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/24/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND mRNA vaccines are emerging as new targets for cancer immunotherapy. However, the potential tumor antigens for mRNA vaccine design in hepatocellular carcinoma (HCC) remain to be elucidated. METHODS Genetic and RNA-Seq data were obtained from TCGA and ICGC. Tumor-specific antigens (TSAs) were identified by differential expression, mutation status, HLA binding, antigen-presenting cell (APC) correlation, immune checkpoint (ICP) relevance and prognosis. Consensus clustering was used for patient classification. The molecular and immune status of TSAs and clustered patients, including prognostic ability, tumor microenvironment, tumor-related signature and tumor immune dysfunction and exclusion (TIDE), were further characterized. RESULTS Five dysregulated and mutated TSAs were identified in HCC (TSA5): FXYD6, JAM2, GALNT16, C7, and CCDC146. Seven immune gene modules and five immune subtypes (IS1-IS5) of HCC were identified. The immune subtypes and TSA5-related modules showed distinct molecular, cellular and clinical characteristics. According to our study, IS1 patients may be suitable for vaccination.
Collapse
Affiliation(s)
- Jiantao Fu
- Department of Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
| | - Feng Chen
- Department of Blood Bank, Blood Center of Zhejiang Province, Hangzhou, 310052, Zhejiang, China
| | - Yuanji Lin
- Department of Research, Hangzhou MC Life Sciences Co., Ltd, Hangzhou, 311500, Zhejiang, China
| | - Jin Gao
- Department of Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
| | - Anna Chen
- Department of Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
| | - Jin Yang
- Department of Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China.
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
41
|
Liu X, Jin Y, Wan X, Liang X, Wang K, Liu J, Jiang J, Meng B, Han S, Zhou L, Cai S, Zou F. SALIS transcriptionally represses IGFBP3/Caspase-7-mediated apoptosis by associating with STAT5A to promote hepatocellular carcinoma. Cell Death Dis 2022; 13:642. [PMID: 35871161 PMCID: PMC9308799 DOI: 10.1038/s41419-022-05094-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common subtype of liver cancer and the second most fatal cancer in the world despite the great therapeutic advances in the past two decades, which reminds us of the gap in fully understanding the oncogenic mechanism of HCC. To explore the key factors contributing to the progression of HCC, we identified a LncRNA, termed SALIS (Suppression of Apoptosis by LINC01186 Interacting with STAT5A), functions in promoting the proliferation, colony formation, migration and invasion while suppressing apoptosis in HCC cells. Mechanistic study indicated SALIS physically associates with transcription factor STAT5A and binds to the promoter regions of IGFBP3 and Caspase-7 to transcriptionally repress their expression and further inhibit apoptosis. Our findings identified SALIS as an oncogene to promote HCC by physically binding with STAT5A to inhibit the expression of pro-apoptotic IGFBP3 and Caspase-7, which suggests novel therapeutic targets for HCC treatments.
Collapse
Affiliation(s)
- Xingyuan Liu
- grid.284723.80000 0000 8877 7471Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yi Jin
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuan Wan
- grid.284723.80000 0000 8877 7471Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoting Liang
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ke Wang
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jieyu Liu
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiale Jiang
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Bingyao Meng
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shuo Han
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Liang Zhou
- grid.284723.80000 0000 8877 7471Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- grid.284723.80000 0000 8877 7471Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fei Zou
- grid.284723.80000 0000 8877 7471Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Leung JH, Ng B, Lim WW. Interleukin-11: A Potential Biomarker and Molecular Therapeutic Target in Non-Small Cell Lung Cancer. Cells 2022; 11:cells11142257. [PMID: 35883698 PMCID: PMC9318853 DOI: 10.3390/cells11142257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 85% of lung cancer and is a fast progressive disease when left untreated. Identification of potential biomarkers in NSCLC is an ongoing area of research that aims to detect, diagnose, and prognosticate patients early to optimize treatment. We review the role of interleukin-11 (IL11), a stromal-cell derived pleiotropic cytokine with profibrotic and cellular remodeling properties, as a potential biomarker in NSCLC. This review identifies the need for biomarkers in NSCLC, the potential sources of IL11, and summarizes the available information leveraging upon published literature, publicly available datasets, and online tools. We identify accumulating evidence suggesting IL11 to be a potential biomarker in NSCLC patients. Further in-depth studies into the pathophysiological effects of IL11 on stromal-tumor interaction in NSCLC are warranted and current available literature highlights the potential value of IL11 detection as a diagnostic and prognostic biomarker in NSCLC.
Collapse
Affiliation(s)
- Jason Hongting Leung
- Department of Cardiothoracic Surgery, National Heart Center Singapore, Singapore 169609, Singapore
- Correspondence:
| | - Benjamin Ng
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore 169609, Singapore; (B.N.); (W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169609, Singapore
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore 169609, Singapore; (B.N.); (W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169609, Singapore
| |
Collapse
|
43
|
Huang JY, Lin YC, Chen HM, Lin JT, Kao SH. RETRACTED: Adenine Combined with Cisplatin Promotes Anticancer Activity against Hepatocellular Cancer Cells through AMPK-Mediated p53/p21 and p38 MAPK Cascades. Pharmaceuticals (Basel) 2022; 15:795. [PMID: 35890094 PMCID: PMC9322617 DOI: 10.3390/ph15070795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/24/2022] Open
Abstract
Cisplatin has been widely used in cancer treatments. Recent evidence indicates that adenine has potential anticancer activities against various types of cancers. However, the effects of the combination of adenine and cisplatin on hepatocellular carcinoma (HCC) cells remain sketchy. Here, our objective was to elucidate the anticancer activity of adenine in combination with cisplatin in HCC cells and its mechanistic pathways. Cell viability and cell cycle progression were assessed by the SRB assay and flow cytometry, respectively. Apoptosis was demonstrated by PI/annexin V staining and flow cytometric analysis. Protein expression, signaling cascade, and mRNA expression were analyzed by Western blotting and quantitative RT-PCR, respectively. Our results showed that adenine jointly potentiated the inhibitory effects of cisplatin on the cell viability of SK-Hep1 and Huh7 cells. Further investigation showed that adenine combined with cisplatin induced higher S phase arrest and apoptosis in HCC cells. Mechanically, adenine induced AMPK activation, reduced mTOR phosphorylation, and increased p53 and p21 levels. The combination of adenine and cisplatin synergistically reduced Bcl-2 and increased PUMA, cleaved caspase-3, and PARP in HCC cells. Adenine also upregulated the mRNA expression of p53, p21, PUMA, and PARP, while knockdown of AMPK reduced the increased expression of these genes. Furthermore, adenine also induced the activation of p38 MAPK through AMPK signaling, and the inhibition of p38 MAPK reduced the apoptosis of HCC cells with exposure to adenine combined with cisplatin. Collectively, these findings reveal that the combination of adenine and cisplatin synergistically enhances apoptosis of HCC cells, which may be attributed to the AMPK-mediated p53/p21 and p38 MAPK cascades. It suggests that adenine may be a potential adjuvant for the treatment of HCC in combination with cisplatin.
Collapse
Affiliation(s)
- Jhen-Yu Huang
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung City 402306, Taiwan;
| | - You-Cian Lin
- Cardiovascular Division, Surgical Department, China Medical University Hospital, Taichung City 404332, Taiwan;
- School of Medicine, College of Medicine, China Medical University, Taichung City 404332, Taiwan
| | - Han-Min Chen
- Institute of Applied Science and Engineering, Catholic Fu Jen University, New Taipei 242048, Taiwan;
| | - Jiun-Tsai Lin
- Energenesis Biomedical Co., Ltd., Taipei 114694, Taiwan;
| | - Shao-Hsuan Kao
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung City 402306, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402306, Taiwan
| |
Collapse
|
44
|
Huang ZL, Xu B, Li TT, Xu YH, Huang XY, Huang XY. Integrative Analysis Identifies Cell-Type-Specific Genes Within Tumor Microenvironment as Prognostic Indicators in Hepatocellular Carcinoma. Front Oncol 2022; 12:878923. [PMID: 35707353 PMCID: PMC9190278 DOI: 10.3389/fonc.2022.878923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, but effective early detection and prognostication methods are lacking. Methods The Cox regression model was built to stratify the HCC patients. The single-cell RNA sequencing data analysis and gene set enrichment analysis were employed to investigate the biological function of identified markers. PLCB1 gain- or loss-of-function experiments were performed, and obtained HCC samples were analyzed using quantitative real-time PCR and immunohistochemistry assay to validate the biological function of identified markers. Results In this study, we developed a model using optimized markers for HCC recurrence prediction. Specifically, we screened out 8 genes through a series of data analyses, and built a multivariable Cox model based on their expression. The risk stratifications using the Eight-Gene Cox (EGC) model were closely associated with the recurrence-free survivals (RFS) in both training and three validation cohorts. We further demonstrated that this risk stratification could serve as an independent predictor in predicting HCC recurrence, and that the EGC model could outperform other models. Moreover, we also investigated the cell-type-specific expression patterns of the eight recurrence-related genes in tumor microenvironment using single-cell RNA sequencing data, and interpreted their functional roles from correlation and gene set enrichment analyses, in vitro and in vivo experiments. Particularly, PLCB1 and SLC22A7 were predominantly expressed in malignant cells, and they were predicted to promote angiogenesis and to help maintain normal metabolism in liver, respectively. In contrast, both FASLG and IL2RB were specifically expressed in T cells, and were highly correlated with T cell marker genes, suggesting that these two genes might assist in maintaining normal function of T cell-mediated immune response in tumor tissues. Conclusion In conclusion, the EGC model and eight identified marker genes could not only facilitate the accurate prediction of HCC recurrence, but also improve our understanding of the mechanisms behind HCC recurrence.
Collapse
Affiliation(s)
- Zi-Li Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Xu
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of General Surgery, The Tenth People's Hospital of Tongji University, Shanghai, China
| | - Ting-Ting Li
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yong-Hua Xu
- Department of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Yu Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiu-Yan Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
45
|
Dai XK, Ding ZX, Tan YY, Bao HR, Wang DY, Zhang H. Neutrophils inhibit CD8 + T cells immune response by arginase-1 signaling in patients with sepsis. World J Emerg Med 2022; 13:266-273. [PMID: 35837557 PMCID: PMC9233973 DOI: 10.5847/wjem.j.1920-8642.2022.068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/24/2022] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Patients with sepsis often exhibit an acute inflammatory response, followed by an immunosuppressive phase with a poor immune response. However, the underlying mechanisms have not been fully elucidated. METHODS We sought to comprehensively characterize the transcriptional changes in neutrophils of patients with sepsis by transcriptome sequencing. Additionally, we conducted a series of experiments, including real-time quantitative polymerase chain reaction (RT-qPCR) and flow cytometry to investigate the role of arginase-1 signaling in sepsis. RESULTS Through the analysis of gene expression profiles, we identified that the negative regulation of T cell activation signaling was enriched, and the expression of arginase-1 was high in neutrophils from patients with sepsis. Furthermore, we conducted flow cytometry and found that the function of CD8+ T cells in septic patients was impaired. Moreover, neutrophils from septic patients inhibited the percentage of polyfunctional effector CD8+ T cells through arginase-1. Additionally, the proportions of granzyme B+IFN-γ+CD8+ T and TNF-α+IFN-γ+CD8+ T cells increased after inhibition of arginase-1 signaling. CONCLUSION The impaired effector function of CD8+ T cells could be restored by blocking arginase-1 signaling in patients with sepsis.
Collapse
Affiliation(s)
- Xiao-kang Dai
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, Hefei 230027, China
| | - Zhen-xing Ding
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, Hefei 230027, China
| | - Yuan-yuan Tan
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, Hefei 230027, China
| | - Hua-rui Bao
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, Hefei 230027, China
| | - Dong-yao Wang
- Department of Hematology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, Hefei 230001, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230001, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu 233030, China
| | - Hong Zhang
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, Hefei 230027, China
| |
Collapse
|
46
|
Zhang C, Liu S, Yang M. Hepatocellular Carcinoma and Obesity, Type 2 Diabetes Mellitus, Cardiovascular Disease: Causing Factors, Molecular Links, and Treatment Options. Front Endocrinol (Lausanne) 2021; 12:808526. [PMID: 35002979 PMCID: PMC8733382 DOI: 10.3389/fendo.2021.808526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, which will affect more than a million people by the year 2025. However, current treatment options have limited benefits. Nonalcoholic fatty liver disease (NAFLD) is the fastest growing factor that causes HCC in western countries, including the United States. In addition, NAFLD co-morbidities including obesity, type 2 diabetes mellitus (T2DM), and cardiovascular diseases (CVDs) promote HCC development. Alteration of metabolites and inflammation in the tumor microenvironment plays a pivotal role in HCC progression. However, the underlying molecular mechanisms are still not totally clear. Herein, in this review, we explored the latest molecules that are involved in obesity, T2DM, and CVDs-mediated progression of HCC, as they share some common pathologic features. Meanwhile, several therapeutic options by targeting these key factors and molecules were discussed for HCC treatment. Overall, obesity, T2DM, and CVDs as chronic metabolic disease factors are tightly implicated in the development of HCC and its progression. Molecules and factors involved in these NAFLD comorbidities are potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO, United States
| |
Collapse
|
47
|
Muller X, Mohkam K, Mabrut JY. Inflammation is king in liver resection for hepatocellular carcinoma. Hepatobiliary Surg Nutr 2021; 10:839-841. [PMID: 35004949 DOI: 10.21037/hbsn-21-435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022]
Affiliation(s)
- Xavier Muller
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France.,Cancer Research Center of Lyon, INSERM U1052, Lyon, France
| | - Kayvan Mohkam
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France.,Cancer Research Center of Lyon, INSERM U1052, Lyon, France.,Department of Pediatric Surgery and Liver Transplantation, Hôpital Femme Mère Enfant, Université Claude Bernard Lyon 1, Hospices Civils de Lyon, Bron, France
| | - Jean-Yves Mabrut
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France.,Cancer Research Center of Lyon, INSERM U1052, Lyon, France
| |
Collapse
|
48
|
Neganova M, Liu J, Aleksandrova Y, Klochkov S, Fan R. Therapeutic Influence on Important Targets Associated with Chronic Inflammation and Oxidative Stress in Cancer Treatment. Cancers (Basel) 2021; 13:6062. [PMID: 34885171 PMCID: PMC8657135 DOI: 10.3390/cancers13236062] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 01/17/2023] Open
Abstract
Chronic inflammation and oxidative stress are the interconnected pathological processes, which lead to cancer initiation and progression. The growing level of oxidative and inflammatory damage was shown to increase cancer severity and contribute to tumor spread. The overproduction of reactive oxygen species (ROS), which is associated with the reduced capacity of the endogenous cell defense mechanisms and/or metabolic imbalance, is the main contributor to oxidative stress. An abnormal level of ROS was defined as a predisposing factor for the cell transformation that could trigger pro-oncogenic signaling pathways, induce changes in gene expression, and facilitate accumulation of mutations, DNA damage, and genomic instability. Additionally, the activation of transcription factors caused by a prolonged oxidative stress, including NF-κB, p53, HIF1α, etc., leads to the expression of several genes responsible for inflammation. The resulting hyperactivation of inflammatory mediators, including TNFα, TGF-β, interleukins, and prostaglandins can contribute to the development of neoplasia. Pro-inflammatory cytokines were shown to trigger adaptive reactions and the acquisition of resistance by tumor cells to apoptosis, while promoting proliferation, invasion, and angiogenesis. Moreover, the chronic inflammatory response leads to the excessive production of free radicals, which further aggravate the initiated reactions. This review summarizes the recent data and progress in the discovery of mechanisms that associate oxidative stress and chronic inflammation with cancer onset and metastasis. In addition, the review provides insights for the development of therapeutic approaches and the discovery of natural substances that will be able to simultaneously inhibit several key oncological and inflammation-related targets.
Collapse
Affiliation(s)
- Margarita Neganova
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Sergey Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
49
|
Qu C, He L, Yao N, Li J, Jiang Y, Li B, Peng S, Hu K, Chen D, Chen G, Huang W, Cao M, Fan J, Yuan Y, Ye W, Hong J. Myofibroblast-Specific Msi2 Knockout Inhibits HCC Progression in a Mouse Model. Hepatology 2021; 74:458-473. [PMID: 33609283 DOI: 10.1002/hep.31754] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Myofibroblasts play a pivotal role in the development and progression of HCC. Here, we aimed to explore the role and mechanism of myofibroblast Musashi RNA binding protein 2 (MSI2) in HCC progression. APPROACH AND RESULTS Myofibroblast infiltration and collagen deposition were detected and assessed in the tissues from 117 patients with HCC. Transgenic mice (Msi2ΔCol1a1 ) with floxed Msi2 allele and collagen type I alpha 1 chain (Col1a1)-ligand inducible Cre recombinases (CreER) were constructed to generate a myofibroblast-specific Msi2 knockout model. Mouse HCC cells were orthotopically transplanted into the Msi2ΔCol1a1 or the control mice (Msi2F/F ). We found that the deposition of collagen fibers, the main product of myofibroblasts, predicted a poor prognosis for HCC; meanwhile, we detected high MSI2 expression in the peritumoral infiltrated myofibroblasts. Conditional deletion of Msi2 in myofibroblasts significantly inhibited the growth of orthotopically implanted HCC, reduced both intrahepatic and lung metastasis, and prolonged the overall survival of tumor-bearing mice (P = 0.002). In vitro analysis demonstrated that myofibroblasts promoted cell proliferation, invasion, and epithelial-mesenchymal transformation of HCC cells, whereas Msi2 deletion in myofibroblasts reversed these effects. Mechanically, Msi2 knockout decreased myofibroblast-derived IL-6 and IL-11 secretion by inhibiting the extracellular signal-regulated kinase 1/2 pathway, and thus attenuated the cancer stem cell-promoting effect of myofibroblasts. Interestingly, we found that the simultaneous knockout of Msi2 in myofibroblasts and knockdown of Msi2 in HCC cells could not further attenuate the implanted HCC progression. CONCLUSIONS Myofibroblast-specific Msi2 knockout abrogated the tumor-promoting function of myofibroblasts and inhibited HCC progression in mouse models. Targeting myofibroblast MSI2 expression may therefore prove to be a therapeutic strategy for HCC treatment in the future.
Collapse
Affiliation(s)
- Chen Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Lu He
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Nan Yao
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jinying Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuchuan Jiang
- Department of Hepatological Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuang Peng
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Kunpeng Hu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guo Chen
- Department of Biochemistry, School of Medicine, Jinan University, Guangzhou, China
| | - Wei Huang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mingrong Cao
- Department of Hepatological Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Fan
- Department of Biochemistry, School of Medicine, Jinan University, Guangzhou, China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Department of Hepatological Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
50
|
Zhang H, Qiu C, Zeng H, Zhu W, Lyu W, Lao X. Upregulation of Stress-Induced Protein Kinase CK1 Delta is associated with a Poor Prognosis for patients with Hepatocellular Carcinoma. Genet Test Mol Biomarkers 2021; 25:504-514. [PMID: 34280005 DOI: 10.1089/gtmb.2020.0093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: This study was designed to analyze the expression of CSNK1D in hepatocellular carcinoma (HCC) and investigate the relationship between the expression of CSNK1D and the prognosis of HCC patients. Methods: The CSNK1D and alpha-fetoprotein (AFP) expression levels in patients with HCC and their corresponding clinical data were downloaded from The Cancer Genome Atlas (TCGA) and sorted with a Perl program. CSNK1D and AFP expression differences in liver tissue and liver cancer were compared and analyzed, based on the online database human cancer metastasis database, the relationships between the expression levels of CSNK1D and AFP and the proliferation and metastases of HCC were explored. The immunohistochemical data obtained from the Human Protein Atlas Database further verified the differences in the expression levels of CSNK1D and AFP in liver tissues and liver cancer tissues. Through Kaplan-Meier survival analysis, the effects of CSNK1D and AFP expression levels on the prognosis of patients with HCC were investigated, and the influences of and patients' gender, age and grades of cancer cells, tumor size, the status of lymph node metastasis, distant metastasis, and tumor stage on the expression of CSNK1D were analyzed with R language. The influence of differential expressions of CSNK1D on survival time was compared and the prognostic factors influencing the survival of HCC patients were statistically explored by univariate analysis and multivariate analysis. The potential influencing mechanism of CSNK1D on the prognosis of HCC patients was explored by Gene Set Enrichment Analysis (GSEA) enrichment. Results: The expression level of CSNK1D and AFP in cancer foci was significantly higher than that in normal tissues, However, in the same patient, the expression levels of AFP in paracarcinoma tissues and cancer tissues showed no significant difference. The expression level of CSNK1D in HCC with distant metastases was higher than that in those without metastasis, but the expression level of AFP in metastatic HCC was lower than that in those HCC without metastases. In immunohistochemical tests, CSNK1D was moderately positive in normal liver tissues, slightly positive in normal bile duct tissues, and highly positive in HCC. AFP was slightly positive in normal liver tissues and negative in HCC, but it was not detected in normal intrahepatic bile duct tissue. Survival analysis results suggested that the higher expression level of CSNK1D corresponded to the shorter survival period, whereas the expression level of AFP showed no significant influence on survival time. The expression level of CSNK1D was not correlated with gender, age, the status of lymph node metastasis status, or distant metastasis of patients. The main factors influencing the expression level of CSNK1D included tumor size, cancer cell grade, and tumor stage. The expression levels of CSNK1D in T2 and T3 were higher than that in T1. The expression levels of CSNK1D in G3 and G4 were higher than that in G1. The expression levels of CSNK1D in Stage II and Stage III were higher than that in Stage I. Univariate analysis suggested that tumor size, cell grade, distant metastasis, clinical stage, and CSNK1D expression level were the prognostic factors influencing the survival of patients. Multivariate analysis suggested that CSNK1D expression level was an independent factor influencing the prognosis of HCC patients. GSEA enrichment analysis indicated that CSNK1D mainly affected the prognosis of HCC patients through cell cycle, WNT signaling pathway, amino acid degradation metabolism, and other pathways. Conclusion: CSNK1D is an independent influencing factor for the prognosis of HCC patients and has the potential to be developed as a potential therapeutic target for HCC, and better than AFP in predicting the prognosis of HCC.
Collapse
Affiliation(s)
| | | | - Haifeng Zeng
- Zhaoqing First People's Hospital, Zhaoqing, China
| | - Wentian Zhu
- Zhaoqing First People's Hospital, Zhaoqing, China
| | - Weidong Lyu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Xuejun Lao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|