1
|
Zhang X, Li S, Hao L, Jia F, Yu F, Hu X. Influencing factors and mechanism of hepatocyte regeneration. J Transl Med 2025; 23:493. [PMID: 40307789 PMCID: PMC12042435 DOI: 10.1186/s12967-025-06278-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/20/2025] [Indexed: 05/02/2025] Open
Abstract
As a research hotspot in the field of regenerative medicine, hepatocyte regeneration has great potential in the treatment of liver diseases. This paper comprehensively summarizes the diverse sources of hepatocyte regeneration and its complex influencing factors, and deeply discusses the typical mechanism. According to the existing research, we observed that Wnt signaling pathway and Notch signaling pathway can play a synergistic role in the process of hepatocyte regeneration. So we further analyzed the crosstalk between Wnt and Notch signal pathway and the cross mechanism with TGF-β, YAP/TAZ pathway during regeneration. Despite the remarkable progress in the study of liver regeneration at the cellular and molecular levels, the comprehensive understanding of the fine regulation of influencing factors and the interaction between mechanisms still needs to be deepened. This paper aims to systematically analyze the interaction between influencing factors and classical mechanisms of hepatocyte regeneration by integrating multi-group data and advanced bioinformatics methods, so as to provide feasible ideas for the treatment of liver diseases and lay a solid theoretical foundation for the future development of regenerative medicine. It is believed that focusing on the rational development of innovative means such as inducing gene tendentiousness expression and anti-aging therapy, and in-depth analysis of the complex interactive network between hepatocyte regeneration mechanisms are expected to open up a new road for the development of more effective treatment strategies for liver diseases.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Fukang Jia
- Henan University of Traditional Chinese, Zhengzhou, China
| | - Fei Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, Bao H, Yam JWP, Xu Y. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol 2025; 13:315-326. [PMID: 40206274 PMCID: PMC11976435 DOI: 10.14218/jcth.2024.00401] [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: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.
Collapse
Affiliation(s)
- Zixin Liang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Li
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyu Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junting Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiehan Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
3
|
Planells-Cases R, Vorobeva V, Kar S, Schmitt FW, Schulte U, Schrecker M, Hite RK, Fakler B, Jentsch TJ. Endosomal chloride/proton exchangers need inhibitory TMEM9 β-subunits for regulation and prevention of disease-causing overactivity. Nat Commun 2025; 16:3117. [PMID: 40169677 PMCID: PMC11962092 DOI: 10.1038/s41467-025-58546-3] [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: 12/10/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
The function of endosomes critically depends on their ion homeostasis. A crucial role of luminal Cl-, in addition to that of H+, is increasingly recognized. Both ions are transported by five distinct endolysosomal CLC chloride/proton exchangers. Dysfunction of each of these transporters entails severe disease. Here we identified TMEM9 and TMEM9B as obligatory β-subunits for endosomal ClC-3, ClC-4, and ClC-5. Mice lacking both β-subunits displayed severely reduced levels of all three CLCs and died embryonically or shortly after birth. TMEM9 proteins regulate trafficking of their partners. Surprisingly, they also strongly inhibit CLC ion transport. Tonic inhibition enables the regulation of CLCs and prevents toxic Cl- accumulation and swelling of endosomes. Inhibition requires a carboxy-terminal TMEM9 domain that interacts with CLCs at multiple sites. Disease-causing CLCN mutations that weaken inhibition by TMEM9 proteins cause a pathogenic gain of ion transport. Our work reveals the need to suppress, in a regulated manner, endolysosomal chloride/proton exchange. Several aspects of endosomal ion transport must be revised.
Collapse
Affiliation(s)
- Rosa Planells-Cases
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Viktoriia Vorobeva
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Graduate Program of the Free University Berlin, Berlin, Germany
| | - Sumanta Kar
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Franziska W Schmitt
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Graduate Program of the Humboldt University Berlin, Berlin, Germany
| | - Uwe Schulte
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Logopharm GmbH, March-Buchheim, Breisgau, Germany
| | - Marina Schrecker
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard K Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Wang W, Wang Z, Li R, Huang W, Ling Q, Li X, Li Z, Cao M, Zhang Z, Sun Q, Liang Z, Zhang HA, Jiang X, Lin C, Chen Y, Zhao B, Zhao Y, Pan JA, Peng X. RNF138 regulates skeletal muscle differentiation via the Wnt/β-catenin signaling pathway. Theranostics 2025; 15:4446-4464. [PMID: 40225576 PMCID: PMC11984406 DOI: 10.7150/thno.110925] [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: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Myogenesis is a strictly regulated process driven by signaling pathways activating muscle-specific gene expression. During myogenesis, muscle stem cells exhibit DNA damage response (DDR) features, which are essential for myoblast differentiation and skeletal muscle regeneration. However, the specific roles of DDR-associated proteins in these processes are not yet fully understood. Methods: Gene knockdown and knockout were used in cell and animal models to study RNF138's function in myoblast differentiation and skeletal muscle regeneration. Multi-omics profiling, including transcriptomics and proteomics, was conducted to identify the key proteins regulated by RNF138 in myogenesis. Protein turnover assays were utilized to investigate RNF138's role in APC protein turnover. Immunofluorescence microscopy was performed to confirm the protein colocalization and subcellular localization. Results: RNF138 expression increases during myoblast differentiation and in regenerating myofibers following muscle injury. Knockdown of RNF138 in C2C12 myoblasts impairs myogenic differentiation and fusion. Additionally, Rnf138-deficient mice exhibit delayed muscle regeneration following cardiotoxin-induced injury. Multi-omics profiling, including transcriptomics and proteomics, reveals that Wnt/β-catenin signaling, a key driver of myogenic differentiation, is enhanced by RNF138. Mechanistically, RNF138 stabilizes β-catenin and enhances its nuclear localization by facilitating lysosomal degradation of APC, a component of the β-catenin degradation complex responsible for mediating the export of β-catenin from the nucleus to the cytoplasm for further ubiquitin-proteasome degradation. Conclusions: We reveal a noncanonical role for RNF138, an E3 ubiquitin ligase, as a positive regulator of myoblast differentiation and skeletal muscle regeneration via the Wnt/β-catenin pathway. This finding highlights the noncanonical function of RNF138 beyond its known roles in DDR and other cellular processes. Therefore, RNF138 provides a potential link between DDR and myoblast differentiation, offering new insights into the molecular regulation of muscle regeneration.
Collapse
Affiliation(s)
- Wenhao Wang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhuohua Wang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Rourong Li
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Weiyi Huang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Qiao Ling
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xiaoxiao Li
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zan Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Manqi Cao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhihui Zhang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Qingrong Sun
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhijuan Liang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Hua-an Zhang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xuan Jiang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Chuwen Lin
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Yaoqing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Bo Zhao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Yu Zhao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Ji-An Pan
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- The Center for Infection and Immunity Study, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xiaoxue Peng
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- The Center for Infection and Immunity Study, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| |
Collapse
|
5
|
Zhang X, Zhou L, Wei S, Zhang X. Comprehensive analysis of TMEM9 in human tumors. Discov Oncol 2025; 16:276. [PMID: 40053281 DOI: 10.1007/s12672-025-02040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND TMEM9, a transmembrane protein, has emerged as a significant player in tumor progression, yet its comprehensive role across various cancers remains unclear. This study investigates the expression patterns, genetic alterations, immune associations, and prognostic implications of TMEM9 across multiple cancer types using large-scale bioinformatics approaches. METHODS TMEM9 expression was analyzed in normal and tumor tissues using data from the TCGA and GTEx databases, with protein expression verified via CPTAC datasets. The prognostic impact of TMEM9 was assessed using overall survival (OS) and disease-free survival (DFS) analyses across various cancers. Genetic alterations, including mutation types and copy number alterations, were explored using the cBioPortal platform. We also examined DNA methylation, RNA modifications, and immune infiltration correlations using bioinformatics tools, including TIMER2 and UALCAN. TMEM9's relationship with tumor mutational burden (TMB) and microsatellite instability (MSI) was analyzed, and gene enrichment analyses were performed using the STRING database and GO/KEGG pathway analyses. RESULTS TMEM9 was significantly overexpressed in several cancers, including ACC, BLCA, BRCA, CHOL, COAD, GBM, and others. Elevated TMEM9 expression correlated with worse OS and DFS in ACC, CESC, KICH, UVM, and additional tumor types. Genetic alterations, predominantly amplifications, were frequent in BRCA, LIHC, and UCEC. DNA methylation analysis revealed hypermethylation in tumors like HNSC and KIRC, while RNA modification analyses showed TMEM9 associations with m6A and m1A-related genes. TMEM9 expression was strongly correlated with immune infiltration, particularly cancer-associated fibroblasts in tumors like THYM and HNSC. Positive associations between TMEM9 and TMB/MSI were observed in several cancers, suggesting genomic instability. Enrichment analyses identified TMEM9 involvement in pathways related to the endoplasmic reticulum, Wnt signaling, and protein processing. CONCLUSION TMEM9 plays a crucial role in cancer progression, influencing gene expression, immune modulation, and genomic instability. These findings highlight TMEM9 as a potential prognostic biomarker and therapeutic target across multiple cancer types.
Collapse
Affiliation(s)
- Xuezhong Zhang
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, 255036, China
| | - Lele Zhou
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, 255036, China
| | - Shumin Wei
- Department of Anesthesiology (Operation Room),, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, 257091, China
| | - Xuebin Zhang
- Department of Anorectal Surgery, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, 257091, China.
| |
Collapse
|
6
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
7
|
Li S, Li M, Li G, Li L, Yang X, Zuo Z, Zhang L, Hu X, He X. Physical Exercise Decreases Complement-Mediated Synaptic Loss and Protects Against Cognitive Impairment by Inhibiting Microglial Tmem9-ATP6V0D1 in Alzheimer's Disease. Aging Cell 2025:e14496. [PMID: 39871402 DOI: 10.1111/acel.14496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/25/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
Physical exercise is known to slow synaptic neurodegeneration and cognitive aging in Alzheimer's disease (AD). The benefits of physical exercise are related to reduced amyloid beta (Aβ) deposition and increased synaptic plasticity. Yet little is known about the mechanisms that mediate these effects. Here, we show that physical exercise down-regulated the microglial Tmem9 protein, inhibited C1q activation, and decreased C1q-dependent microglial synapse engulfment, eventually ameliorating cognitive impairment in 5xFAD mice. Furthermore, using oAβ cultured-BV2 in vitro, we show that downregulation of microglial Tmem9 was sufficient to restrain complement activity and decrease microglia-mediated synaptic loss, whereas overexpression of microglial Tmem9 tended to promote complement activation and induced synaptic loss, abolishing exercise-associated protection. Finally, we show that microglial Tmem9 contributed to complement activation by regulating ATP6V0D1, a vesicular (H+) ATP-dependent proton pump (V-ATPase) subunit that regulates V-ATPase assembly. Together, our results demonstrate that exercise is a potential treatment for AD patients. In an AD mouse model, it decreased the levels of microglial Tmem9 to inhibit the activation of complement, alleviated complement-dependent synaptic loss, and eventually ameliorated emotional and cognitive disorders.
Collapse
Affiliation(s)
- Shiyin Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mingyue Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Lili Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaofeng Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaofei He
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Li X, Feng J, Cheng H, Jin N, Jin S, Liu Z, Xu J, Xie J. Human umbilical cord mesenchymal stem cells enhance liver regeneration and decrease collagen content in fibrosis mice after partial hepatectomy by activating Wnt/β-catenin signaling. Acta Biochim Biophys Sin (Shanghai) 2024; 57:604-615. [PMID: 39716885 PMCID: PMC12040747 DOI: 10.3724/abbs.2024207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/17/2024] [Indexed: 12/25/2024] Open
Abstract
Liver fibrosis is a critical stage in the progression of various chronic liver diseases to cirrhosis and liver cancer. Early inhibition of liver fibrosis is crucial for the treatment of liver disease. Hepatectomy, a common treatment for liver-related diseases, promotes liver regeneration. However, in the context of liver fibrosis, liver regeneration is hindered. Many studies have shown that mesenchymal stem cells (MSCs) can promote liver regeneration after partial hepatectomy (PH). However, there are few reports on the impact of MSC therapy on liver regeneration post-PH in the context of hepatic fibrosis. The objective of this study is to examine the impact of MSCs on liver regeneration following PH in the fibrotic liver and uncover the related molecular mechanisms. This study reveals that MSC therapy significantly enhances liver function and mitigates liver inflammation after PH in the context of hepatic fibrosis. MSCs also significantly promote liver regeneration and alleviate liver fibrosis. In addition, this study identifies the role of MSCs in promoting liver regeneration and alleviating liver fibrosis via the activation of Wnt/β-catenin signaling. The combination of MSCs with hepatectomy may offer a novel approach for the treatment of liver fibrotic diseases.
Collapse
Affiliation(s)
- Xuewei Li
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Jinghui Feng
- Academy of Medical SciencesShanxi Medical UniversityTaiyuan030001China
| | - Haiqin Cheng
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Ning Jin
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Shanshan Jin
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplant Centerthe First Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Jun Xie
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
9
|
Tejeda-Muñoz N, Binder G, Mei KC. Emerging therapeutic strategies for Wnt-dependent colon cancer targeting macropinocytosis. Cells Dev 2024; 180:203974. [PMID: 39528157 PMCID: PMC12009640 DOI: 10.1016/j.cdev.2024.203974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Aberrations in the Wnt signaling pathway, particularly mutations in genes like APC and β-catenin, are pivotal in initiating and driving the progression of colorectal cancer (CRC), establishing this pathway as a crucial target for therapeutic intervention. Membrane trafficking plays a key role in regulating Wnt signaling by controlling the activation, modulation, and secretion of essential signaling molecules that contribute to CRC progression. This review explores the connection between membrane trafficking and Wnt signaling, with a specific focus on macropinocytosis-an endocytic process involved in nutrient uptake that also plays a role in Wnt signal regulation. The relationship between Wnt signaling and macropinocytosis, critical in both embryonic development and cancer onset, reveals a new dimension for therapeutic intervention. Targeting Wnt signaling through the modulation of macropinocytosis and broader membrane trafficking pathways presents a promising therapeutic strategy, with several candidates already in early clinical trials. These emerging approaches underscore the potential of targeting Wnt and its associated membrane trafficking processes for CRC treatment, aligning with the development of innovative therapies.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Grace Binder
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - Kuo-Ching Mei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Binghamton, NY, USA
| |
Collapse
|
10
|
Zhou Y, Lin X, Jiao Y, Yang D, Li Z, Zhu L, Li Y, Yin S, Li Q, Xu S, Tang D, Zhang S, Yu W, Gao P, Yang L. A brain-to-liver signal mediates the inhibition of liver regeneration under chronic stress in mice. Nat Commun 2024; 15:10361. [PMID: 39609433 PMCID: PMC11605118 DOI: 10.1038/s41467-024-54827-5] [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: 02/08/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
As the ability of liver regeneration is pivotal for liver disease patients, it will be of high significance and importance to identify the missing piece of the jigsaw influencing the liver regeneration. Here, we report that chronic stress impairs the liver regeneration capacity after partial hepatectomy with increased mortality in male mice. Anatomical tracing and functional mapping identified a neural circuit from noradrenergic neurons in the locus coeruleus (LC) to serotonergic neurons in the rostral medullary raphe region (rMR), which critically contributes to the inhibition of liver regeneration under chronic stress. In addition, hepatic sympathetic nerves were shown to be critical for the inhibitory effects on liver regeneration by releasing norepinephrine (NE), which acts on adrenergic receptor β2 (ADRB2) to block the proinflammatory macrophage activation. Collectively, we reveal a "brain-to-liver" neural connection that mediates chronic stress-evoked deficits in liver regeneration, thus shedding important insights into hepatic disease therapy.
Collapse
Affiliation(s)
- Yanyu Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Xiaoqi Lin
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Dan Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Zhengyu Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yixuan Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Suqing Yin
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Quanfu Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Song Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| | - Liqun Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| |
Collapse
|
11
|
Tsakiroglou M, Evans A, Doce-Carracedo A, Little M, Hornby R, Roberts P, Zhang E, Miyajima F, Pirmohamed M. Gene Expression Dysregulation in Whole Blood of Patients with Clostridioides difficile Infection. Int J Mol Sci 2024; 25:12653. [PMID: 39684365 DOI: 10.3390/ijms252312653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Clostridioides difficile (C. difficile) is a global threat and has significant implications for individuals and health care systems. Little is known about host molecular mechanisms and transcriptional changes in peripheral immune cells. This is the first gene expression study in whole blood from patients with C. difficile infection. We took blood and stool samples from patients with toxigenic C. difficile infection (CDI), non-toxigenic C. difficile infection (GDH), inflammatory bowel disease (IBD), diarrhea from other causes (DC), and healthy controls (HC). We performed transcriptome-wide RNA profiling on peripheral blood to identify diarrhea common and CDI unique gene sets. Diarrhea groups upregulated innate immune responses with neutrophils at the epicenter. The common signature associated with diarrhea was non-specific and shared by various other inflammatory conditions. CDI had a unique 45 gene set reflecting the downregulation of humoral and T cell memory functions. Dysregulation of immunometabolic genes was also abundant and linked to immune cell fate during differentiation. Whole transcriptome analysis of white cells in blood from patients with toxigenic C. difficile infection showed that there is an impairment of adaptive immunity and immunometabolism.
Collapse
Affiliation(s)
- Maria Tsakiroglou
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Anthony Evans
- Computational Biology Facility, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Alejandra Doce-Carracedo
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Clinical Directorate, GCP Laboratories, University of Liverpool, Liverpool L7 8TX, UK
| | - Margaret Little
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Rachel Hornby
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Paul Roberts
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Faculty of Science and Engineering, School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LZ, UK
| | - Eunice Zhang
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Fabio Miyajima
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Oswaldo Cruz Foundation (Fiocruz), Branch Ceara, Eusebio 61773-270, Brazil
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| |
Collapse
|
12
|
Jin X, Wang S, Luo L, Yan F, He Q. Targeting the Wnt/β-catenin signal pathway for the treatment of gastrointestinal cancer: Potential for advancement. Biochem Pharmacol 2024; 227:116463. [PMID: 39102994 DOI: 10.1016/j.bcp.2024.116463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
Collapse
Affiliation(s)
- Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Fangjie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
13
|
Tümen D, Heumann P, Huber J, Hahn N, Macek C, Ernst M, Kandulski A, Kunst C, Gülow K. Unraveling Cancer's Wnt Signaling: Dynamic Control through Protein Kinase Regulation. Cancers (Basel) 2024; 16:2686. [PMID: 39123414 PMCID: PMC11312265 DOI: 10.3390/cancers16152686] [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: 07/08/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Since the initial identification of oncogenic Wnt in mice and Drosophila, the Wnt signaling pathway has been subjected to thorough and extensive investigation. Persistent activation of Wnt signaling exerts diverse cancer characteristics, encompassing tumor initiation, tumor growth, cell senescence, cell death, differentiation, and metastasis. Here we review the principal signaling mechanisms and the regulatory influence of pathway-intrinsic and extrinsic kinases on cancer progression. Additionally, we underscore the divergences and intricate interplays of the canonical and non-canonical Wnt signaling pathways and their critical influence in cancer pathophysiology, exhibiting both growth-promoting and growth-suppressing roles across diverse cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (N.H.)
| |
Collapse
|
14
|
de Pellegars-Malhortie A, Picque Lasorsa L, Mazard T, Granier F, Prévostel C. Why Is Wnt/β-Catenin Not Yet Targeted in Routine Cancer Care? Pharmaceuticals (Basel) 2024; 17:949. [PMID: 39065798 PMCID: PMC11279613 DOI: 10.3390/ph17070949] [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: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
Collapse
Affiliation(s)
- Auriane de Pellegars-Malhortie
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Laurence Picque Lasorsa
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Thibault Mazard
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
- Medical Oncology Department, ICM, University of Montpellier, CEDEX 5, 34298 Montpellier, France
| | | | - Corinne Prévostel
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| |
Collapse
|
15
|
Gao H, Wei L, Indulkar S, Nguyen TTL, Liu D, Ho MF, Zhang C, Li H, Weinshilboum RM, Ingle JN, Wang L. Androgen receptor-mediated pharmacogenomic expression quantitative trait loci: implications for breast cancer response to AR-targeting therapy. Breast Cancer Res 2024; 26:111. [PMID: 38965614 PMCID: PMC11225427 DOI: 10.1186/s13058-024-01861-2] [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: 11/17/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Shreya Indulkar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Thanh Thanh L Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Tejeda-Muñoz N, Mei KC. Wnt signaling in cell adhesion, development, and colon cancer. IUBMB Life 2024; 76:383-396. [PMID: 38230869 DOI: 10.1002/iub.2806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/03/2023] [Indexed: 01/18/2024]
Abstract
Wnt signaling is essential for embryonic development, influencing processes such as axis formation, cell proliferation and differentiation, cell fate decisions, and axon guidance. It also plays a role in maintaining tissue homeostasis in adult organisms. The loss of normal cell polarity and adhesion caused by Wnt signaling activation is a fundamental step for tumor progression and metastasis. Activating the canonical Wnt pathway is a driving force in many human cancers, especially colorectal, hepatocellular, and mammary carcinomas. Wnt causes the stabilization and nuclear transport of newly synthesized transcriptional regulator β-catenin. The generally accepted view is that the canonical effects of Wnt growth factors are caused by the transcription of β-catenin target genes. Here, we review recent findings that indicate Wnt is a regulator of many other cellular physiological activities, such as macropinocytosis, endosome trafficking, protein stability, focal adhesions, and lysosomal activity. Some of these regulatory responses occur within minutes and do not require new protein synthesis, indicating that there is much more to Wnt beyond the well-established transcriptional role of β-catenin. The main conclusion that emerges from these studies is that in basal cell conditions, the activity of the key protein kinase GSK3, which is inhibited by Wnt pathway activation, normally represses the actin machinery that orchestrates macropinocytosis with implications in cancer. These contributions expand our understanding of the multifaceted roles of Wnt signaling in cellular processes, development, and cancer, providing insights into potential therapeutic targets and strategies.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Kuo-Ching Mei
- School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, USA
| |
Collapse
|
17
|
Li T, Zhong W, Li M, Shao Z, Zhang G, Wang W, Gao Z, Tan X, Xu Z, Luo F, Song G. TRIM26 deficiency enhancing liver regeneration through macrophage polarization and β-catenin pathway activation. Cell Death Dis 2024; 15:453. [PMID: 38926362 PMCID: PMC11208526 DOI: 10.1038/s41419-024-06798-0] [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] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Liver regeneration is a complex process involving the crosstalk between parenchymal and non-parenchymal cells, especially macrophages. However, the underlying mechanisms remain incompletely understood. Here, we identify the E3 ubiquitin ligase TRIM26 as a crucial regulator of liver regeneration. Following partial hepatectomy or acute liver injury induced by carbon tetrachloride, Trim26 knockout mice exhibit enhanced hepatocyte proliferation compared to wild-type controls, while adeno-associated virus (AAV)-mediated overexpression of Trim26 reverses the promotional effects. Mechanistically, Trim26 deficiency promotes the recruitment of macrophages to the liver and their polarization towards pro-inflammatory M1 phenotype. These M1 macrophages secrete Wnts, including Wnt2, which subsequently stimulate hepatocyte proliferation through the activation of Wnt/β-catenin signaling. In hepatocytes, Trim26 knockdown reduces the ubiquitination and degradation of β-catenin, thereby further enhancing Wnt/β-catenin signaling. Pharmacological inhibition of Wnt/β-catenin pathway by ICG-001 or depletion of macrophages by clodronate liposomes diminishes the pro-regenerative effects of Trim26 deficiency. Moreover, bone marrow transplantation experiments provide evidence that Trim26 knockout in myeloid cells alone can also promote liver regeneration, highlighting the critical role of macrophage Trim26 in this process. Taken together, our study uncovers TRIM26 as a negative regulator of liver regeneration by modulating macrophage polarization and Wnt/β-catenin signaling in hepatocytes, providing a potential therapeutic target for promoting liver regeneration in clinical settings.
Collapse
Affiliation(s)
- Tingting Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Mengqi Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zile Shao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Gongye Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Weiwei Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zhixing Gao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Xuemei Tan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Ziyi Xu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Fanghong Luo
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Song
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
18
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
19
|
Wang Z, Zhao P, Tian K, Qiao Z, Dong H, Li J, Guan Z, Su H, Song Y, Ma X. TMEM9 promotes lung adenocarcinoma progression via activating the MEK/ERK/STAT3 pathway to induce VEGF expression. Cell Death Dis 2024; 15:295. [PMID: 38664392 PMCID: PMC11045738 DOI: 10.1038/s41419-024-06669-8] [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: 08/06/2023] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Abnormal Transmembrane protein 9 (TMEM9) expression has been identified in various human tumors. However, the prognostic potential and mechanistic role of TMEM9 in lung adenocarcinoma (LUAD) remain unclear. Here, we first found a significant upregulation of TMEM9 in LUAD tissues, and TMEM9 expression was positively correlated with microvessel density (MVD), T stage, and clinical stage. Survival analysis demonstrated TMEM9 was an independent indicator of poor prognosis in LUAD patients. In addition, downregulation of TMEM9 suppressed tumor growth and metastasis in vitro and in vivo models, and reduced HUVEC proliferation, migration, and tube formation in a cancer cell/HUVEC coculture model. Furthermore, TMEM9 upregulated VEGF expression, and VEGF-neutralizing antibodies reversed HUVEC angiogenesis and cancer cell migration ability caused by overexpression of TMEM9. In contrast, recombinant VEGF (rVEGF) abolished the inhibitory effect of TMEM9-knockdown LUAD cells on HUVEC angiogenesis and tumor cell migration. Moreover, we showed that TMEM9 upregulated VEGF expression by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase/STAT3 (MEK/ERK/STAT3) pathway. Together, our study provides mechanistic insights into the role of TMEM9 in LUAD and highlights the potential of targeting the TMEM9/MEK/ERK/STAT3/VEGF pathway as a novel therapy for preventing LUAD progression.
Collapse
Affiliation(s)
- Zhiqian Wang
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Peng Zhao
- Biotherapy Center, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Kaihua Tian
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhongshi Qiao
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hao Dong
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jie Li
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zitong Guan
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Su
- Department of Oncology, LiaochengPeople's Hospital, Liaocheng, Shandong, China
| | - Yang Song
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Qingdao University, Qingdao, Shandong, China.
| | - Xuezhen Ma
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
20
|
Wang Y, Xu M, Yao Y, Li Y, Zhang S, Fu Y, Wang X. Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions. Oncol Lett 2024; 27:167. [PMID: 38449793 PMCID: PMC10915806 DOI: 10.3892/ol.2024.14300] [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: 08/14/2023] [Accepted: 01/10/2024] [Indexed: 03/08/2024] Open
Abstract
Tumor invasion and metastasis are the processes that primarily cause adverse outcomes in patients with cervical cancer. Cancer-associated fibroblasts (CAFs), which participate in cancer progression and metastasis, are novel targets for the treatment of tumors. The present study aimed to assess the heterogeneity of CAFs in the cervical cancer microenvironment through single-cell RNA sequencing. After collecting five cervical cancer samples and obtaining the CAF-associated gene sets, the CAFs in the cervical cancer microenvironment were divided into myofibroblastic CAFs and extracellular (ec)CAFs. The ecCAFs appeared with more robust pro-tumorigenic effects than myCAFs according to enrichment analysis. Subsequently, through combining the ecCAF hub genes and bulk gene expression data for cervical cancer obtained from The Cancer Genome Atlas and Gene Ontology databases, univariate Cox regression and least absolute shrinkage and selection operator analyses were performed to establish a CAF-associated risk signature for patients with cancer. The established risk signature demonstrated a stable and strong prognostic capability in both the training and validation cohorts. Subsequently, the association between the risk signature and clinical data was evaluated, and a nomogram to facilitate clinical application was established. The risk score was demonstrated to be associated with both the tumor immune microenvironment and the therapeutic responses. Moreover, the signature also has predictive value for the prognosis of head and neck squamous cell carcinoma, and bladder urothelial carcinoma, which were also associated with human papillomavirus infection. In conclusion, the present study assessed the heterogeneity of CAFs in the cervical cancer microenvironment, and a subgroup of CAFs that may be closely associated with tumor progression was defined. Moreover, a signature based on the hub genes of ecCAFs was shown to have biomarker functionality in terms of predicting survival rates, and therefore this CAF subgroup may become a therapeutic target for cervical cancer in the future.
Collapse
Affiliation(s)
- Yuehan Wang
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Mingxia Xu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yeli Yao
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Ying Li
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Songfa Zhang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yunfeng Fu
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xinyu Wang
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
21
|
Hsiao CY, Lu CY, Su HJ, Huang KW. Plasma Cell-Free Adenomatous Polyposis Coli Gene Promoter Methylation as a Prognostic Biomarker for Hepatocellular Carcinoma. Oncology 2024; 102:935-943. [PMID: 38527449 PMCID: PMC11548096 DOI: 10.1159/000538455] [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: 12/27/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a leading cause of cancer death worldwide. Lack of biomarkers for follow-up after treatment is a clinical challenge. DNA methylation has been proposed to be a potential biomarker in HCC. However, there is still a lack of evidence of its clinical use. This study aimed to evaluate the value of using plasma Adenomatous Polyposis Coli promoter methylation level (APC-MET) as a potential biomarker in HCC treatment. METHOD A total of 96 patients with HCC at BCLC stage B who underwent local tumor ablation treatment were prospectively included in this study. APC-MET was examined in the plasma of each patient before and 1 month after treatment. The prediction value of APC-MET for survival outcome and disease status after treatment was analyzed and adjusted with alpha-fetoprotein and protein induced by vitamin K absence-II using Cox regression analysis. RESULTS Univariate Cox regression analysis showed preoperative APC-MET >0 (HR, 2.9, 95% CI: 1.05-8.05, p = 0.041) and postoperative APC-MET >0 (HR, 3.47, 95% CI: 1.16-10.4, p = 0.026) were both predictors of death, and preoperative APC-MET >0 was a predictor of disease progression after treatment (HR, 2.04, 95% CI: 1.21-3.44, p = 0.007). In multivariate models, preoperative APC-MET >0 was a significant predictor of disease progression after adjusting with the other two traditional biomarkers (HR, 1.82, 95% CI: 1.05-3.17, p = 0.034). CONCLUSIONS Hypermethylation of APC promoter appears to be a potential biomarker that could predict patient survival and disease progression outcomes in patients with intermediate-stage HCC after local ablation treatment.
Collapse
Affiliation(s)
- Chih-Yang Hsiao
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan,
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan,
- Department of Traumatology, National Taiwan University Hospital, Taipei, Taiwan,
| | | | | | - Kai-Wen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
22
|
Liu L, Chen X, Wu L, Huang K, Wang Z, Zheng Y, Zheng C, Zhang Z, Chen J, Wei J, Chen S, Jin W, Chen J, Wei D, Xu Y. Ubiquitin ligase subunit FBXO9 inhibits V-ATPase assembly and impedes lung cancer metastasis. Exp Hematol Oncol 2024; 13:32. [PMID: 38486234 PMCID: PMC10938814 DOI: 10.1186/s40164-024-00497-4] [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: 11/26/2023] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND The evolutionarily conserved protein FBXO9 acts as a substrate receptor for the SKP1-cullin-1-RBX1 ubiquitin ligase and is implicated in cancer, exhibiting either tumor-suppressive or oncogenic effects depending on the specific tumor type. However, their role in lung cancer metastasis remains unclear. METHODS Lentiviral vectors carrying miRNA-based shRNA sequences for gene-specific knockdown were generated, and Lenti-CRISPR-Cas9 vectors containing gene-specific sgRNA sequences were designed. Gene overexpression was achieved using doxycycline-inducible lentiviral constructs, while gene knockdown or knockout cells were generated using shRNA and CRISPR-Cas9, respectively. Functional assays included migration, clonogenic survival assays, tumor sphere assays, and protein interaction studies using mass spectrometry, immunoprecipitation, and immunoblot analysis. RESULTS This study identified FBXO9 as a crucial regulator that suppresses lung cancer cell migration, tumor sphere growth and restricts metastasis. We showed that FBXO9 facilitates the ubiquitination of the catalytic subunit A (ATP6V1A) of the Vacuolar-type H+-ATPase (V-ATPase), resulting in its interaction with the cytoplasmic chaperone HSPA8 and subsequent sequestration within the cytoplasm. This process hinders the assembly of functional V-ATPase, resulting in reduced vesicular acidification. In contrast, depletion of FBXO9 reduced ATP6V1A ubiquitination, resulting in increased V-ATPase assembly and vesicular acidification, thus promoting pro-metastatic Wnt signaling and metastasis of lung cancer cells. Furthermore, we demonstrated the effectiveness of inhibitors targeting V-ATPase in inhibiting lung cancer metastasis in a mouse model. Finally, we established a correlation between lower FBXO9 levels and poorer survival outcomes in patients with lung cancer. CONCLUSION These findings collectively elucidate the critical role of FBXO9 in regulating V-ATPase assembly and provide a molecular basis for FBXO9's function in inhibiting lung cancer metastasis. This highlights the potential therapeutic opportunities of FBXO9 supplementation.
Collapse
Affiliation(s)
- Liang Liu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Institute of Clinical Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaodong Chen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Leilei Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Kaizong Huang
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zhenyi Wang
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Yaolin Zheng
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Cheng Zheng
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, 200032, China
| | - Jiayan Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiaming Wei
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, 223300, Jiangsu, China
| | - Song Chen
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, 223300, Jiangsu, China
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Weilin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Jinfei Chen
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 730000, Zhejiang Province, China
| | - Dongping Wei
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China.
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
23
|
Zhang C, Sun C, Zhao Y, Ye B, Yu G. Signaling pathways of liver regeneration: Biological mechanisms and implications. iScience 2024; 27:108683. [PMID: 38155779 PMCID: PMC10753089 DOI: 10.1016/j.isci.2023.108683] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2023] Open
Abstract
The liver possesses a unique regenerative ability to restore its original mass, in this regard, partial hepatectomy (PHx) and partial liver transplantation (PLTx) can be executed smoothly and safely, which has important implications for the treatment of liver disease. Liver regeneration (LR) can be the very complicated procedure that involves multiple cytokines and transcription factors that interact with each other to activate different signaling pathways. Activation of these pathways can drive the LR process, which can be divided into three stages, namely, the initiation, progression, and termination stages. Therefore, it is important to investigate the pathways involved in LR to elucidate the mechanism of LR. This study reviews the latest research on the key signaling pathways in the different stages of LR.
Collapse
Affiliation(s)
- Chunyan Zhang
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Caifang Sun
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Yabin Zhao
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Bingyu Ye
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - GuoYing Yu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
24
|
García-Trevijano ER, Ortiz-Zapater E, Gimeno A, Viña JR, Zaragozá R. Calpains, the proteases of two faces controlling the epithelial homeostasis in mammary gland. Front Cell Dev Biol 2023; 11:1249317. [PMID: 37795261 PMCID: PMC10546029 DOI: 10.3389/fcell.2023.1249317] [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: 06/28/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Calpain-1 and calpain-2 are calcium-dependent Cys-proteases ubiquitously expressed in mammalian tissues with a processive, rather than degradative activity. They are crucial for physiological mammary gland homeostasis as well as for breast cancer progression. A growing number of evidences indicate that their pleiotropic functions depend on the cell type, tissue and biological context where they are expressed or dysregulated. This review considers these standpoints to cover the paradoxical role of calpain-1 and -2 in the mammary tissue either, under the physiological conditions of the postlactational mammary gland regression or the pathological context of breast cancer. The role of both calpains will be examined and discussed in both conditions, followed by a brief snapshot on the present and future challenges for calpains, the two-gateway proteases towards tissue homeostasis or tumor development.
Collapse
Affiliation(s)
- Elena R. García-Trevijano
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- INLIVA Biomedical Research Institute, Valencia, Spain
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- INLIVA Biomedical Research Institute, Valencia, Spain
| | - Amparo Gimeno
- Department of Anatomy and Human Embryology, Universitat de Valencia, Valencia, Spain
| | - Juan R. Viña
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- INLIVA Biomedical Research Institute, Valencia, Spain
| | - Rosa Zaragozá
- INLIVA Biomedical Research Institute, Valencia, Spain
- Department of Anatomy and Human Embryology, Universitat de Valencia, Valencia, Spain
| |
Collapse
|
25
|
Siapoush S, Rezaei R, Alavifard H, Hatami B, Zali MR, Vosough M, Lorzadeh S, Łos MJ, Baghaei K, Ghavami S. Therapeutic implications of targeting autophagy and TGF-β crosstalk for the treatment of liver fibrosis. Life Sci 2023; 329:121894. [PMID: 37380126 DOI: 10.1016/j.lfs.2023.121894] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023]
Abstract
Liver fibrosis is characterized by the excessive deposition and accumulation of extracellular matrix components, mainly collagens, and occurs in response to a broad spectrum of triggers with different etiologies. Under stress conditions, autophagy serves as a highly conserved homeostatic system for cell survival and is importantly involved in various biological processes. Transforming growth factor-β1 (TGF-β1) has emerged as a central cytokine in hepatic stellate cell (HSC) activation and is the main mediator of liver fibrosis. A growing body of evidence from preclinical and clinical studies suggests that TGF-β1 regulates autophagy, a process that affects various essential (patho)physiological aspects related to liver fibrosis. This review comprehensively highlights recent advances in our understanding of cellular and molecular mechanisms of autophagy, its regulation by TGF-β, and the implication of autophagy in the pathogenesis of progressive liver disorders. Moreover, we evaluated crosstalk between autophagy and TGF-β1 signalling and discussed whether simultaneous inhibition of these pathways could represent a novel approach to improve the efficacy of anti-fibrotic therapy in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Samaneh Siapoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramazan Rezaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Alavifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland; Autophagy Research Center, Department of Biochemistry; Shiraz University of Medical Sciences, Shiraz, Iran; LinkoCare Life Sciences AB, Linkoping, Sweden
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
26
|
Qiao X, Zhang Y, Zhang Z, Niu N, Li H, Sun L, Ma Q, Bu J, Liu J, Chen G, Xue J, Yang Y, Liu C. KCNJ15 deficiency promotes drug resistance via affecting the function of lysosomes. Asian J Pharm Sci 2023; 18:100814. [PMID: 37274925 PMCID: PMC10238847 DOI: 10.1016/j.ajps.2023.100814] [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: 11/04/2022] [Revised: 03/19/2023] [Accepted: 04/22/2023] [Indexed: 06/07/2023] Open
Abstract
The altered lysosomal function can induce drug redistribution which leads to drug resistance and poor prognosis for cancer patients. V-ATPase, an ATP-driven proton pump positioned at lysosomal surfaces, is responsible for maintaining the stability of lysosome. Herein, we reported that the potassium voltage-gated channel subfamily J member 15 (KCNJ15) protein, which may bind to V-ATPase, can regulate the function of lysosome. The deficiency of KCNJ15 protein in breast cancer cells led to drug aggregation as well as reduction of drug efficacy. The application of the V-ATPase inhibitor could inhibit the binding between KCNJ15 and V-ATPase, contributing to the amelioration of drug resistance. Clinical data analysis revealed that KCNJ15 deficiency was associated with higher histological grading, advanced stages, more metastases of lymph nodes, and shorter disease free survival of patients with breast cancer. KCNJ15 expression level is positively correlated with a high response rate after receiving neoadjuvant chemotherapy. Moreover, we revealed that the small molecule drug CMA/BAF can reverse drug resistance by disrupting the interaction between KCNJ15 and lysosomes. In conclusion, KCNJ15 could be identified as an underlying indicator for drug resistance and survival of breast cancer, which might guide the choice of therapeutic strategies.
Collapse
Affiliation(s)
- Xinbo Qiao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Yixiao Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Nan Niu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Haonan Li
- School of Bioengineering, Dalian University of Technology, Dalian 116000, China
| | - Lisha Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Qingtian Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Jinchi Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| | - Guanglei Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yongliang Yang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- School of Bioengineering, Dalian University of Technology, Dalian 116000, China
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shenyang 110000, China
| |
Collapse
|
27
|
Hou X, Luan J, Fu S. Multi-functional gene ZNF281 identified as a molecular biomarker in soft tissue regeneration and pan-cancer progression. Front Genet 2023; 13:1082654. [PMID: 36685971 PMCID: PMC9849369 DOI: 10.3389/fgene.2022.1082654] [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: 10/28/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Regeneration and tumorigenesis are indicated as related processes, while regeneration leads to life and the outcome of tumorigenesis is death. Here, we show the upregulation of zfp281 (zinc finger 281) in our adipose de novo regeneration model through RNA-seq analysis. Then, we validated the upregulation of zfp281 in adipose regeneration via immunofluorescence. Following that, we found that ZNF281 (the human homolog of Zfp281) was upregulated in most types of cancer and related to worse prognosis in 10 tumors. We further investigated the role of ZNF281 in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), pancreatic adenocarcinoma (PAAD), and stomach adenocarcinoma (STAD) and confirmed the high accuracy in the clinical diagnostic feature. Beyond that, based on these three types of cancers, we analyzed the ZNF281-related tumor immune infiltration and DNA methylation sites and finally built risk prediction models for future disease diagnosis. Taken together, our findings provide new insights into the dual role of ZNF281, and we found that it was a potential biomarker for regeneration and tumor prognosis.
Collapse
Affiliation(s)
| | - Jie Luan
- *Correspondence: Jie Luan, ; Su Fu,
| | - Su Fu
- *Correspondence: Jie Luan, ; Su Fu,
| |
Collapse
|
28
|
Zou G, Park JI. Wnt signaling in liver regeneration, disease, and cancer. Clin Mol Hepatol 2023; 29:33-50. [PMID: 35785913 PMCID: PMC9845677 DOI: 10.3350/cmh.2022.0058] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/30/2022] [Indexed: 02/02/2023] Open
Abstract
The liver exhibits the highest recovery rate from acute injuries. However, in chronic liver disease, the long-term loss of hepatocytes often leads to adverse consequences such as fibrosis, cirrhosis, and liver cancer. The Wnt signaling plays a pivotal role in both liver regeneration and tumorigenesis. Therefore, manipulating the Wnt signaling has become an attractive approach to treating liver disease, including cancer. Nonetheless, given the crucial roles of Wnt signaling in physiological processes, blocking Wnt signaling can also cause several adverse effects. Recent studies have identified cancer-specific regulators of Wnt signaling, which would overcome the limitation of Wnt signaling target approaches. In this review, we discussed the role of Wnt signaling in liver regeneration, precancerous lesion, and liver cancer. Furthermore, we summarized the basic and clinical approaches of Wnt signaling blockade and proposed the therapeutic prospects of cancer-specific Wnt signaling blockade for liver cancer treatment.
Collapse
Affiliation(s)
- Gengyi Zou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Corresponding author : Gengyi Zou Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd Unit 1054, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA,Jae-Il Park Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd. Unit 1052, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| |
Collapse
|
29
|
Zhao Z, Su J, Zhao J, Chen J, Cui X, Sun M, Zhang X. Curcumin inhibits invasion and metastasis of human hepatoma cells through Bclaf1-mediated Wnt/β-catenin signalling. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2113864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Zhongwei Zhao
- Medical College, Yanbian University, Yanji, People’s Republic of China
- College of Special Education, Changchun University, Changchun, People’s Republic of China
| | - Jielin Su
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Jiaqi Zhao
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Jiaxin Chen
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Xinmu Cui
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Manqing Sun
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Xuewu Zhang
- Medical College, Yanbian University, Yanji, People’s Republic of China
| |
Collapse
|
30
|
Zhang S, Lee SH, Nie L, Huang Y, Zou G, Jung Y, Jun S, Zhang J, Lien EM, Chen J, Park J. Lysosomal TMEM9-LAMTOR4-controlled mTOR signaling integrity is required for mammary tumorigenesis. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 43:159-163. [PMID: 36336962 PMCID: PMC9859727 DOI: 10.1002/cac2.12382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/16/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Shengzhe Zhang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Sung Ho Lee
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Litong Nie
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Yuanjian Huang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Gengyi Zou
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Youn‐Sang Jung
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Sohee Jun
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jie Zhang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Esther M. Lien
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Junjie Chen
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jae‐Il Park
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA,Graduate School of Biomedical Sciencesthe University of Texas MD Anderson Cancer CenterHoustonTX77030USA,Program in Genetics and Epigeneticsthe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| |
Collapse
|
31
|
Wang Q, Mao Z, Liu Z, Xu M, Huang S, Wang Y, Xu Y, Qi L, Liu M, Liu Y. Akt/mTOR integrate energy metabolism with Wnt signal to influence wound epithelium growth in Gekko Japonicus. Commun Biol 2022; 5:1018. [PMID: 36167813 PMCID: PMC9515156 DOI: 10.1038/s42003-022-04004-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
The formation of wound epithelium initiates regeneration of amputated tail in Gekko japonicus. Energy metabolism is indispensable for the growth of living creatures and typically influenced by temperature. In this study, we reveal that low temperature lowers energy metabolism level and inhibits the regeneration of amputated tails of Gekko japonicus. We further find that low temperature attenuates the activation of protein kinase B (Akt) and mammalian target of rapamycin (mTOR) in regenerated tissues upon injury signals, and the inhibition of Akt hinders proliferation of the wound epithelium. Additionally, wingless/integrated (Wnt) inhibition suppresses epithelium proliferation and formation by inhibiting Akt activation. Finally, low temperature elevates the activity of adenylate-activated kinase (AMPK) pathway and in turn attenuates wound epithelium formation. Meanwhile, either mTOR downregulation or AMPK upregulation is associated with worse wound epithelium formation. Summarily, low temperature restricts wound epithelium formation by influencing energy sensory pathways including Akt/mTOR and AMPK signaling, which is also modulated by injury induced Wnt signal. Our results provide a mechanism that incorporates the injury signals with metabolic pathway to facilitate regeneration. Low temperature inhibits the regeneration of amputated tails of Gekko japonicus by influencing the energy sensory Akt/mTOR pathway, which is also modulated by injury-induced Wnt signal.
Collapse
Affiliation(s)
- Qinghua Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Comparative Medicine Research Institution, Nantong University, Nantong, 226001, China
| | - Zuming Mao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Zhuang Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Man Xu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuai Huang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yin Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yanran Xu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong University, Nantong, 226001, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
32
|
Zhang L, Li Y, Wang Y, Qiu Y, Mou H, Deng Y, Yao J, Xia Z, Zhang W, Zhu D, Qiu Z, Lu Z, Wang J, Yang Z, Mao G, Chen D, Sun L, Liu L, Ju Z. mTORC2 Facilitates Liver Regeneration Through Sphingolipid-Induced PPAR-α-Fatty Acid Oxidation. Cell Mol Gastroenterol Hepatol 2022; 14:1311-1331. [PMID: 35931382 PMCID: PMC9703135 DOI: 10.1016/j.jcmgh.2022.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS During liver regeneration after partial hepatectomy, the function and metabolic pathways governing transient lipid droplet accumulation in hepatocytes remain obscure. Mammalian target of rapamycin 2 (mTORC2) facilitates de novo synthesis of hepatic lipids. Under normal conditions and in tumorigenesis, decreased levels of triglyceride (TG) and fatty acids (FAs) are observed in the mTORC2-deficient liver. However, during liver regeneration, their levels increase in the absence of mTORC2. METHODS Rictor liver-specific knockout and control mice underwent partial hepatectomy, followed by measurement of TG and FA contents during liver regeneration. FA metabolism was evaluated by analyzing the expression of FA metabolism-related genes and proteins. Intraperitoneal injection of the peroxisome proliferator-activated receptor α (PPAR-α) agonist, p53 inhibitor, and protein kinase B (AKT) activator was performed to verify the regulatory pathways involved. Lipid mass spectrometry was performed to identify the potential PPAR-α activators. RESULTS The expression of FA metabolism-related genes and proteins suggested that FAs are mainly transported into hepatocytes during liver regeneration. The PPAR-α pathway is down-regulated significantly in the mTORC2-deficient liver, resulting in the accumulation of TGs. The PPAR-α agonist WY-14643 rescued deficient liver regeneration and survival in mTORC2-deficient mice. Furthermore, lipidomic analysis suggested that mTORC2 deficiency substantially reduced glucosylceramide (GluCer) content. GluCer activated PPAR-α. GluCer treatment in vivo restored the regenerative ability and survival rates in the mTORC2-deficient group. CONCLUSIONS Our data suggest that FAs are mainly transported into hepatocytes during liver regeneration, and their metabolism is facilitated by mTORC2 through the GluCer-PPAR-α pathway, thereby establishing a novel role for mTORC2 in lipid metabolism.
Collapse
Affiliation(s)
- Lingling Zhang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China,Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China,Correspondence Address correspondence to: Lingling Zhang, MD, PhD, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China.
| | - Yanqiu Li
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Ying Wang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yugang Qiu
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Hanchuan Mou
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Yuanyao Deng
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Jiyuan Yao
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhiqing Xia
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Wenzhe Zhang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Di Zhu
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Zeyu Qiu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Zhongjie Lu
- Department of Thoracic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jirong Wang
- Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Zhouxin Yang
- Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - GenXiang Mao
- Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Dan Chen
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Leimin Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Leiming Liu
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China,Leiming Liu, PhD, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China,Zhenyu Ju, MD, PhD, Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
33
|
Yang Z, Chen W, Zhu H, Zhang L, Zhou K, Tang H, Sun R, Huang Y, Xie H, Zheng S, Jia C. Methylation site APC112043544 as a potential biomarker for post-transplant hepatocellular carcinoma recurrence. Future Oncol 2022; 18:2401-2413. [PMID: 35502765 DOI: 10.2217/fon-2021-1608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/13/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: To investigate the prognostic value of DNA methylation of tumor suppressor genes for hepatocellular carcinoma (HCC) recurrence after liver transplantation. Methods: APC gene was selected according to The Cancer Genome Atlas dataset. Tumor tissues and clinical data of 85 HCC patients who received a liver transplantation were retrospectively enrolled and next-generation methylation sequencing was performed. Risk factors were determined using the Cox proportional-hazard-regression model. Results: The APC methylation site (chromosome 5, position 112043544) was an independent predictor of post-transplant HCC recurrence. Patients with hyper-methylated APC112043544 experienced superior recurrence-free survival (p = 0.021) and had a decreased proportion of microvascular invasion (p = 0.017). APC112043544 also predicted recurrence risk in patients beyond selection criteria. Conclusions: APC112043544 methylation may serve as a potential biomarker for post-transplant HCC recurrence.
Collapse
Affiliation(s)
- Zhentao Yang
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Wei Chen
- Department of Hepatopancreatobiliary Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Research Center of Diagnosis & Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, 310000, China
| | - Hai Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, China
| | - Liang Zhang
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Ke Zhou
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Hong Tang
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Ruiqi Sun
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Yiqian Huang
- Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Haiyang Xie
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis & Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310000, China
| | - Shusen Zheng
- Department of Surgery, Division of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis & Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310000, China
| | - Changku Jia
- Department of Hepatopancreatobiliary Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Research Center of Diagnosis & Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
34
|
Tejeda-Muñoz N, Mei KC, Sheladiya P, Monka J. Targeting Membrane Trafficking as a Strategy for Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10050790. [PMID: 35632546 PMCID: PMC9144176 DOI: 10.3390/vaccines10050790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/07/2022] [Accepted: 05/15/2022] [Indexed: 12/17/2022] Open
Abstract
Membrane trafficking is emerging as an attractive therapeutic strategy for cancer. Recent reports have found a connection between Wnt signaling, receptor-mediated endocytosis, V-ATPase, lysosomal activity, and macropinocytosis through the canonical Wnt pathway. In macropinocytic cells, a massive internalization of the plasma membrane can lead to the loss of cell-surface cadherins, integrins, and other antigens that mediate cell–cell adhesion, favoring an invasive phenotype. V-ATPase is a key regulator in maintaining proper membrane trafficking, homeostasis, and the earliest developmental decisions in the Xenopus vertebrate development model system. Here, we review how the interference of membrane trafficking with membrane trafficking inhibitors might be clinically relevant in humans.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
- Correspondence:
| | - Kuo-Ching Mei
- Division of Pharmacoengineering and Molecular School Pharmaceutics, Eshelman of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Pooja Sheladiya
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| |
Collapse
|
35
|
Pathogenesis of Liver Fibrosis and Its TCM Therapeutic Perspectives. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5325431. [PMID: 35529927 PMCID: PMC9071861 DOI: 10.1155/2022/5325431] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/15/2022] [Indexed: 12/16/2022]
Abstract
Liver fibrosis is a pathological process of abnormal tissue proliferation in the liver caused by various pathogenic factors, which will further develop into cirrhosis or even hepatocellular carcinoma if liver injury is not intervened in time. As a diffuse progressive liver disease, its clinical manifestations are mostly excessive deposition of collagen-rich extracellular matrix resulting in scar formation due to liver injury. Hepatic fibrosis can be caused by hepatitis B and C, fatty liver, alcohol, and rare diseases such as hemochromatosis. As the metabolic center of the body, the liver regulates various vital activities. During the development of fibrosis, it is influenced by many other factors in addition to the central event of hepatic stellate cell activation. Currently, with the increasing understanding of TCM, the advantages of TCM with multiple components, pathways, and targets have been demonstrated. In this review, we will describe the factors influencing liver fibrosis, focusing on the effects of cells, intestinal flora, iron death, signaling pathways, autophagy and angiogenesis on liver fibrosis, and the therapeutic effects of herbal medicine on liver fibrosis.
Collapse
|
36
|
Abstract
Lysosomes are the digestive center of the cell and play important roles in human diseases, including cancer. Previous work has suggested that late endosomes, also known as multivesicular bodies (MVBs), and lysosomes are essential for canonical Wnt pathway signaling. Sequestration of Glycogen Synthase 3 (GSK3) and of β‐catenin destruction complex components in MVBs is required for sustained canonical Wnt signaling. Little is known about the role of lysosomes during early development. In the Xenopus egg, a Wnt-like cytoplasmic determinant signal initiates formation of the body axis following a cortical rotation triggered by sperm entry. Here we report that cathepsin D was activated in lysosomes specifically on the dorsal marginal zone of the embryo at the 64-cell stage, long before zygotic transcription starts. Expansion of the MVB compartment with low-dose hydroxychloroquine (HCQ) greatly potentiated the dorsalizing effects of the Wnt agonist lithium chloride (LiCl) in embryos, and this effect required macropinocytosis. Formation of the dorsal axis required lysosomes, as indicated by brief treatments with the vacuolar ATPase (V-ATPase) inhibitors Bafilomycin A1 or Concanamycin A at the 32-cell stage. Inhibiting the MVB-forming machinery with a dominant-negative point mutation in Vacuolar Protein Sorting 4 (Vps4-EQ) interfered with the endogenous dorsal axis. The Wnt-like activity of the dorsal cytoplasmic determinant Huluwa (Hwa), and that of microinjected xWnt8 messenger RNA, also required lysosome acidification and the MVB-forming machinery. We conclude that lysosome function is required for early dorsal axis development in Xenopus. The results highlight the intertwining between membrane trafficking, lysosomes, and vertebrate axis formation.
Collapse
|
37
|
Zhang D, Dai J, Cao Y, Wang Z, Qiao Z, Qiao Z. Nicotine exposure of male mice protects offspring against carbon tetrachloride-induced acute liver injury. J Biochem Mol Toxicol 2022; 36:e23069. [PMID: 35411647 DOI: 10.1002/jbt.23069] [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: 05/14/2021] [Revised: 11/19/2021] [Accepted: 03/23/2022] [Indexed: 11/09/2022]
Abstract
Paternal nicotine exposure can cause a phenotypic change in offspring. To study whether paternal nicotine exposure influences acute liver injury and repair of the offspring, we established a paternal nicotine exposure model in mice and treated the offspring mice with carbon tetrachloride (CCl4 ) to induce acute liver injury. After the treatment of CCl4 , the levels of alanine aminotransferase and aspartate aminotransferase in offspring serum of paternal nicotine exposed mice are about 37.44%, and 30.21% lower than the control mice, respectively. Transcription profiling screen and bioinformatics analysis of differently expressed genes in F1 mice liver revealed that the Wnt pathway was altered. The results demonstrate that nicotine exposure in male mice could enhance the activation of the Wnt pathway in F1 mice liver. The Wnt pathway facilitates cell proliferation and tissue repair. In conclusion, our findings showed that nicotine exposure of male mice protects hepatic against CCl4 -induced acute injury in offspring by activating the Wnt pathway in the F1 liver.
Collapse
Affiliation(s)
- Dong Zhang
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingbo Dai
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, Illinois, USA
| | - Yong Cao
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoxia Wang
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiguang Qiao
- Renji Hospital
- South Campus, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongdong Qiao
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Ageing related thyroid deficiency increases brain-targeted transport of liver-derived ApoE4-laden exosomes leading to cognitive impairment. Cell Death Dis 2022; 13:406. [PMID: 35468877 PMCID: PMC9039072 DOI: 10.1038/s41419-022-04858-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer’s disease (AD) is the prevalent cause of dementia in the ageing world population. Apolipoprotein E4 (ApoE4) allele is the key genetic risk factor for AD, although the mechanisms linking ApoE4 with neurocognitive impairments and aberrant metabolism remains to be fully characterised. We discovered a significant increase in the ApoE4 content of serum exosomes in old healthy subjects and AD patients carrying ApoE4 allele as compared with healthy adults. Elevated exosomal ApoE4 demonstrated significant inverse correlation with serum level of thyroid hormones and cognitive function. We analysed effects of ApoE4-containing peripheral exosomes on neural cells and neurological outputs in aged or thyroidectomised young mice. Ageing-associated hypothyroidism as well as acute thyroidectomy augmented transport of liver-derived ApoE4 reach exosomes into the brain, where ApoE4 activated nucleotide-binding oligomerisation domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome by increasing cholesterol level in neural cells. This, in turn, affected cognition, locomotion and mood. Our study reveals pathological potential of exosomes-mediated relocation of ApoE4 from the periphery to the brain, this process can represent potential therapeutic target.
Collapse
|
39
|
Dong Y, Liu X, Jiang B, Wei S, Xiang B, Liao R, Wang Q, He X. A Genome-Wide Investigation of Effects of Aberrant DNA Methylation on the Usage of Alternative Promoters in Hepatocellular Carcinoma. Front Oncol 2022; 11:780266. [PMID: 35111672 PMCID: PMC8803206 DOI: 10.3389/fonc.2021.780266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The alternative usage of promoters provides a way to regulate gene expression, has a significant influence on the transcriptome, and contributes to the cellular transformation of cancer. However, the function of alternative promoters (APs) in hepatocellular carcinoma (HCC) has not been systematically studied yet. In addition, the potential mechanism of regulation to the usage of APs remains unclear. DNA methylation, one of the most aberrant epigenetic modifications in cancers, is known to regulate transcriptional activity. Whether DNA methylation regulates the usage of APs needs to be explored. Here, we aim to investigate the effects of DNA methylation on usage of APs in HCC. METHODS Promoter activities were calculated based on RNA-seq data. Functional enrichment analysis was implemented to conduct GO terms. Correlation tests were used to detect the correlation between promoter activity and methylation status. The LASSO regression model was used to generate a diagnostic model. Kaplan-Meier analysis was used to compare the overall survival between high and low methylation groups. RNA-seq and whole-genome bisulfite sequencing (WGBS) in HCC samples were performed to validate the correlation of promoter activity and methylation. RESULTS We identified 855 APs in total, which could be well used to distinguish cancer from normal samples. The correlation of promoter activity and DNA methylation in APs was observed, and the APs with negative correlation were defined as methylation-regulated APs (mrAPs). Six mrAPs were identified to generate a diagnostic model with good performance (AUC = 0.97). Notably, the majority of mrAPs had CpG sites that could be used to predict clinical outcomes by methylation status. Finally, we verified 85.6% of promoter activity variation and 92.3% of methylation changes in our paired RNA-seq and WGBS samples, respectively. The negative correlation between promoter activity and methylation status was further confirmed in our HCC samples. CONCLUSION The aberrant methylation status plays a critical role in the precision usage of APs in HCC, which sheds light on the mechanism of cancer development and provides a new insight into cancer screening and treatment.
Collapse
Affiliation(s)
- Yuting Dong
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhao Liu
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Bijun Jiang
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Siting Wei
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ruichu Liao
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi, Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Ximiao He
- Department of Physiology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Zhao X, Yang L, Qin L. Methyltransferase-like 3 (METTL3) attenuates cardiomyocyte apoptosis with myocardial ischemia-reperfusion (I/R) injury through miR-25-3p and miR-873-5p. Cell Biol Int 2021; 46:992. [PMID: 34553450 DOI: 10.1002/cbin.11706] [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: 12/20/2022]
Abstract
Methyltransferase-like 3 (METTL3) mediated N6 -methyladenosine (m6A) promotes microRNAs (miRNAs) maturation by processing the primary miRNAs, and METTL3 involves in regulating the development of various diseases, including myocardial ischemia-reperfusion (I/R) injury. However, up until now, the association between METTL3 regulated miRNAs and I/R injury is not fully investigated, which makes investigations on this academic issue necessary. In this study, we showed that METTL3 was downregulated in mice I/R myocardial tissues and hypoxic/re-oxygenated (H/R) cardiomyocytes, and upregulation of METTL3 attenuated I/R and H/R-induced cell apoptosis. In addition, we screened out that two miRNAs, including miR-25-3p and miR-873-5p, were positively regulated by METTL3 in cardiomyocytes in a DGCR8-dependent manner. In addition, both miR-25-3p and miR-873-5p were significantly downregulated by I/R and H/R treatments in mice tissues and cardiomyocytes, and overexpression of the above two miRNAs were effective to improve cell viability in cardiomyocytes under H/R stress. Next, we evidenced that METTL3 suppressed H/R-induced cell death via upregulating miR-25-3p and miR-873-5p. Finally, the potential downstream mechanisms were investigated, and we expectedly found that METTL3 activated the PI3K/Akt pathway in H/R-treated cardiomyocytes through modulating miR-25-3p and miR-873-5p, and the PI3K/Akt pathway inhibitor (LY294002) abrogated the protective effects of METTL3 overexpression in cardiomyocytes with H/R treatment. Collectively, we concluded that METTL3 upregulated miR-25-3p and miR-873-5p to activate the PI3K/Akt pathway, resulting in the suppression of I/R injury.
Collapse
Affiliation(s)
- Xiangmei Zhao
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Lei Yang
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Lijie Qin
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| |
Collapse
|
41
|
Abstract
Mucins are high molecular-weight epithelial glycoproteins and are implicated in many physiological processes, including epithelial cell protection, signaling transduction, and tissue homeostasis. Abnormality of mucus expression and structure contributes to biological properties related to human cancer progression. Tumor growth sites induce inhospitable conditions. Many kinds of research suggest that mucins provide a microenvironment to avoid hypoxia, acidic, and other biological conditions that promote cancer progression. Given that the mucus layer captures growth factors or cytokines, we propose that mucin helps to ameliorate inhospitable conditions in tumor-growing sites. Additionally, the composition and structure of mucins enable them to mimic the surface of normal epithelial cells, allowing tumor cells to escape from immune surveillance. Indeed, human cancers such as mucinous carcinoma, show a higher incidence of invasion to adjacent organs and lymph node metastasis than do non-mucinous carcinoma. In this mini-review, we discuss how mucin provides a tumor-friendly environment and contributes to increased cancer malignancy in mucinous carcinoma.
Collapse
Affiliation(s)
- Dong-Han Wi
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| | - Jong-Ho Cha
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon 22212, Korea
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| |
Collapse
|
42
|
Abstract
The liver is uniquely bestowed with an ability to regenerate following a surgical or toxicant insult. One of the most researched models to demonstrate the regenerative potential of this organ is the partial hepatectomy model, where two thirds of the liver is surgically resected. The remnant liver replenishes the lost mass within 1014 days in mice. The distinctive ability of the liver to regenerate has allowed living donor and split liver transplantation. One signaling pathway shown to be activated during the process of regeneration to contribute toward the mass and functional recovery of the liver is the Wnt/-catenin pathway. Very early after any insult to the liver, the cellmolecule circuitry of the Wnt/-catenin pathway is set into motion with the release of specific Wnt ligands from sinusoidal endothelial cells and macrophages, which, in a paracrine manner, engage Frizzled and LDL-related protein-5/6 coreceptors on hepatocytes to stabilize -catenin inducing its nuclear translocation. Nuclear -catenin interacts with T-cell factor family of transcription factors to induce target genes including cyclin D1 for proliferation, and others for regulating hepatocyte function. Working in collaboration with other signaling pathways, Wnt/-catenin signaling contributes to the restoration process without any compromise of function at any stage. Also, stimulation of this pathway through innovative means induces liver regeneration when this process is exhausted or compromised and thus has applications in the treatment of end-stage liver disease and in the field of liver transplantation. Thus, Wnt/-catenin signaling pathway is highly relevant in the discipline of hepatic regenerative medicine.
Collapse
Affiliation(s)
- Shikai Hu
- *School of Medicine, Tsinghua University, Beijing, China
- †Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Satdarshan P. Monga
- †Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- ‡Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- §Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Inhibiting ATP6V0D2 Aggravates Liver Ischemia-Reperfusion Injury by Promoting NLRP3 Activation via Impairing Autophagic Flux Independent of Notch1/Hes1. J Immunol Res 2021; 2021:6670495. [PMID: 33860063 PMCID: PMC8024071 DOI: 10.1155/2021/6670495] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
At present, liver ischemia-reperfusion (IR) injury is still a great challenge for clinical liver partial resection and liver transplantation. The innate immunity regulated by liver macrophages orchestrates the cascade of IR inflammation and acts as a bridge. As a specific macrophage subunit of vacuolar ATPase, ATP6V0D2 (V-ATPase D2 subunit) has been shown to promote the formation of autophagolysosome in vitro. Our research fills a gap which has existed in the study of inflammatory stress about the V-ATPase subunit ATP6V0D2 in liver macrophages. We first found that the expression of specific ATP6V0D2 in liver macrophages was upregulated with the induction of inflammatory cascade after liver IR surgery, and knockdown of ATP6V0D2 resulted in increased secretion of proinflammatory factors and chemokines, which enhanced activation of NLRP3 and aggravation of liver injury. Further studies found that the exacerbated activation of NLRP3 was related to the autophagic flux regulated by ATP6V0D2. Knocking down ATP6V0D2 impaired the formation of autophagolysosome and aggravated liver IR injury through nonspecific V-ATPase activation independent of V-ATPase-Notchl-Hesl signal axis. In general, we illustrated that the expression of ATP6V0D2 in liver macrophages was upregulated after liver IR, and by gradually promoting the formation of autophagolysosomes to increase autophagy flux to limit the activation of liver inflammation, this regulation is independent of the Notch1-Hes1 signal axis.
Collapse
|
44
|
Colozza G, Koo BK. Wnt/β-catenin signaling: Structure, assembly and endocytosis of the signalosome. Dev Growth Differ 2021; 63:199-218. [PMID: 33619734 PMCID: PMC8251975 DOI: 10.1111/dgd.12718] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022]
Abstract
Wnt/β‐catenin signaling is an ancient pathway that regulates key aspects of embryonic development, cell differentiation, proliferation, and adult stem cell homeostasis. Work from different laboratories has shed light on the molecular mechanisms underlying the Wnt pathway, including structural details of ligand–receptor interactions. One key aspect that has emerged from multiple studies is that endocytosis of the receptor complex plays a crucial role in fine‐tuning Wnt/β‐catenin signaling. Endocytosis is a key process involved in both activation as well as attenuation of Wnt signaling, but how this is regulated is still poorly understood. Importantly, recent findings show that Wnt also regulates central metabolic pathways such as the acquisition of nutrients through actin‐driven endocytic mechanisms. In this review, we propose that the Wnt pathway displays diverse characteristics that go beyond the regulation of gene expression, through a connection with the endocytic machinery.
Collapse
Affiliation(s)
- Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
45
|
Kim MJ, Huang Y, Park JI. Targeting Wnt Signaling for Gastrointestinal Cancer Therapy: Present and Evolving Views. Cancers (Basel) 2020; 12:E3638. [PMID: 33291655 PMCID: PMC7761926 DOI: 10.3390/cancers12123638] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/19/2022] Open
Abstract
Wnt signaling governs tissue development, homeostasis, and regeneration. However, aberrant activation of Wnt promotes tumorigenesis. Despite the ongoing efforts to manipulate Wnt signaling, therapeutic targeting of Wnt signaling remains challenging. In this review, we provide an overview of current clinical trials to target Wnt signaling, with a major focus on gastrointestinal cancers. In addition, we discuss the caveats and alternative strategies for therapeutically targeting Wnt signaling for cancer treatment.
Collapse
Affiliation(s)
- Moon Jong Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and Health Science Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
46
|
Zhang Y, Sun Y, Liu J, Han Y, Yan J. MicroRNA-346-5p Regulates Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells by Inhibiting Transmembrane Protein 9. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8822232. [PMID: 33299881 PMCID: PMC7704134 DOI: 10.1155/2020/8822232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 12/27/2022]
Abstract
The molecular mechanisms how bone marrow-derived mesenchymal stem cells (BMSCs) differentiate into osteoblast need to be investigated. MicroRNAs (miRNAs) contribute to the osteogenic differentiation of BMSCs. However, the effect of miR-346-5p on osteogenic differentiation of BMSCs is not clear. This study is aimed at elucidating the underlying mechanism by which miR-346-5p regulates osteogenic differentiation of human BMSCs. Results of alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining indicated that upregulation of miR-346-5p suppressed osteogenic differentiation of BMSCs, whereas downregulation of miR-346-5p enhanced this process. The protein levels of the osteoblastic markers Osterix and Runt-related transcription factor 2 (Runx2) were decreased in cells treated with miR-346-5p mimic at day 7 and day 14 after being differentiated. By contrast, downregulation of miR-346-5p elevated the protein levels of Osterix and Runx2. Moreover, a dual-luciferase reporter assay revealed that Transmembrane Protein 9 (TMEM9) was a target of miR-346-5p. In addition, the Western Blot results demonstrated that the TMEM9 protein level was significantly reduced by the miR-346-5p mimic whereas downregulation of miR-346-5p improved the protein level of TMEM9. These results together demonstrated that miR-346-5p served a key role in BMSC osteogenic differentiation of through targeting TMEM9, which may provide a novel target for clinical treatments of bone injury.
Collapse
Affiliation(s)
- Yicai Zhang
- Department of Orthopedic, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150010, China
- Department of Orthopedic, The Third People's Hospital of Huizhou, Huizhou, Guangdong, 516002, China
| | - Yi Sun
- Department of Orthopedic, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150010, China
| | - Jinlong Liu
- Department of Orthopedic, The 80th Group Army Hospital of PLA, Weifang, Shandong 261042, China
| | - Yu Han
- Department of Orthopedic, The Third People's Hospital of Huizhou, Huizhou, Guangdong, 516002, China
| | - Jinglong Yan
- Department of Orthopedic, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150010, China
| |
Collapse
|
47
|
He S, Tang S. WNT/β-catenin signaling in the development of liver cancers. Biomed Pharmacother 2020; 132:110851. [PMID: 33080466 DOI: 10.1016/j.biopha.2020.110851] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
The WNT/β-catenin signaling pathway is a highly conserved and tightly controlled molecular mechanism that regulates embryonic development, cellular proliferation and differentiation. Of note, accumulating evidence has shown that the aberrant of WNT/β-catenin signaling promotes the development and/or progression of liver cancer, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), the two most prevalent primary liver tumours in adults. There are two different WNT signaling pathways have been identified, which were termed non-canonical and canonical pathways, the latter involving the activation of β-catenin. β-catenin, acting as an intracellular signal transducer in the WNT signaling pathway, is encoded by CTNNB1 and plays a critical role in tumorigenesis. In the past research, most liver tumors have mutations in genes encoding key components of the WNT/β-catenin signaling pathway. In addition, several of other signaling pathways also can crosswalk with β-catenin. In this review, we discuss the most relevant molecular mechanisms of action and regulation of WNT/β-catenin signaling in the development and pathophysiology of liver cancers, as well as in the development of therapeutics.
Collapse
Affiliation(s)
- Shuai He
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Shilei Tang
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| |
Collapse
|
48
|
Wen X, Wu Y, Awadasseid A, Tanaka Y, Zhang W. New Advances in Canonical Wnt/β-Catenin Signaling in Cancer. Cancer Manag Res 2020; 12:6987-6998. [PMID: 32821165 PMCID: PMC7418153 DOI: 10.2147/cmar.s258645] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/19/2020] [Indexed: 12/16/2022] Open
Abstract
Wnt/β-catenin-mediated signaling is a key pathway regulating tissue growth and development, and tumorigenesis, and has received increasing attention in recent years. In addition to participating in healthy tissue and organ development, ectopic activation of the pathway can cause a variety of tumors and other pathologies. The pathway plays a critical role in many processes such as proliferation, differentiation, apoptosis, migration, invasion, epithelial–mesenchymal transition and cancer cell stemness. The importance of the Wnt signal is self-evident. This review describes the underlying mechanism of Wnt signaling pathway and highlights the latest findings on the relationship between Wnt signaling pathway and tumorigenesis. In addition, the potential relationship between miRNAs and Wnt signaling is presented. Furthermore, we discuss the intrinsic link between Wnt signaling and cancer cell stemness, which shed light on the malignant progression of tumor cells. Finally, cancer treatment strategies based on the canonical Wnt signaling pathway are summarized, hoping to help clinical development.
Collapse
Affiliation(s)
- Xiaolan Wen
- Laboratory of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.,Laboratory of Molecular Immunology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, People's Republic of China
| | - Yanling Wu
- Laboratory of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.,Laboratory of Molecular Immunology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, People's Republic of China
| | - Annoor Awadasseid
- Laboratory of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.,Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, People's Republic of China
| | - Yoshimasa Tanaka
- Laboratory of Bioinformatics and Molecular Medicine, Center for Medical Innovation, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Wen Zhang
- Laboratory of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| |
Collapse
|