1
|
Zhang Y, Lu Y, Wang N, Hao F, Chen Y, Fei X, Wang J. Ascending E2F7a/b ratio facilitates KLF13 transcription in hepatocellular carcinoma and correlates with the abundance of binuclear hepatocytes (ABH) modulation for short-term recurrence. FASEB J 2025; 39:e70485. [PMID: 40116212 PMCID: PMC11926945 DOI: 10.1096/fj.202402520r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Short-term recurrence after surgery severely threatens patients' lives and leads to dismal outcomes in hepatocellular carcinoma (HCC). Our previous research proposed the abundance of binuclear hepatocytes (ABH) as an independent indicator related to the cytokinesis regulator Anillin and significantly associated with HCC recurrence. The exact mechanism of ABH modulation has not been clearly illustrated. In this study, we intensively investigated the probable regulation mechanism and noticed a contradiction between E2F7 upregulation and ABH attenuation. As we discovered, E2F7 has two isoforms, E2F7a and E2F7b, and we innovatively define a value of the E2F7a/b ratio using a cutoff value of 6.5. E2F7 upregulation in the paracancerous tissues was predominantly presented by the E2F7a isoform, leading to an increase in the E2F7a/b ratio, instead of E2F7b as a main component in non-cancerous tissues, and is associated with short-term recurrence. We further found that KLF13 transcriptionally promotes Anillin expression in HCC and was suppressively impacted by E2F7b, but not by the highly expressed E2F7a. Hence, the ascending E2F7a/b ratio induced significant upregulation of KLF13 and participated in the attenuation of ABH in the paracancerous liver tissues. In conclusion, E2F7 presents a particular expression status in HCC by predominantly upregulating E2F7a rather than E2F7b. The ascending E2F7a/b ratio weakens the suppressive effect on KLF13 transcription and sequentially participates in ABH attenuation, associated with short-term HCC recurrence post-operation.
Collapse
Affiliation(s)
- Yian Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yiquan Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Nan Wang
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Wang L, Wang J, Wang N, Wang X, Song M, Zhou Y, Wang Z, Meng H, Guo E, Miao S. ANLN promotes head and neck squamous cell carcinoma progression by upregulating PD-L1 via the ERK-MAPK pathway. iScience 2025; 28:111633. [PMID: 39967877 PMCID: PMC11834070 DOI: 10.1016/j.isci.2024.111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/27/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
Anillin (ANLN) is a highly conserved protein involved in cytokinesis and cytoskeletal remodeling. This study investigates the role of ANLN in head and neck squamous cell carcinoma (HNSCC) progression and its impact on the tumor immune microenvironment, with a focus on the combination of ANLN silencing and anti-programmed cell death protein 1 (PD-1) therapy. Through in vitro and in vivo experiments, along with clinical specimen analysis, we discovered that silencing ANLN not only inhibits the malignant progression of HNSCC but also reduces the activation of the extracellular signal-regulated kinase-mitogen-activated protein kinase (ERK-MAPK) signaling pathway and decreases programmed death ligand-1 (PD-L1) expression. Integrating ANLN silencing with anti-PD-1 monoclonal antibody treatment significantly enhances the activation of infiltrating CD8+ T cells, leading to marked tumor growth suppression. Our findings highlight the potential of ANLN as a therapeutic target in HNSCC, providing a foundation for developing innovative and effective combined treatment strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Junrong Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Nana Wang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China
| | - Xueying Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha 410008, Hunan, P.R. China
| | - Ming Song
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Yang Zhou
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Ziyuan Wang
- Department of Pathology, Harbin Medical University, Harbin 150081, Heilongjiang, P.R. China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| |
Collapse
|
3
|
Nguyen MT, Lee GJ, Kim B, Kim HJ, Tak J, Park MK, Kim EJ, Kang GJ, Rho SB, Lee H, Lee K, Kim SG, Lee CH. Penfluridol suppresses MYC-driven ANLN expression and liver cancer progression by disrupting the KEAP1-NRF2 interaction. Pharmacol Res 2024; 210:107512. [PMID: 39643070 DOI: 10.1016/j.phrs.2024.107512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 12/09/2024]
Abstract
Hepatocellular carcinoma (HCC) comprises the majority of primary liver cancers and possesses a low 5-year survival rate when in the advanced stages. Anillin (ANLN), a key player in cell growth and cytokinesis, is implicated in HCC development. Currently, no treatment agents are known to suppress ANLN. Analysis of The Cancer Genome Atlas data showed that high ANLN expression is associated with poor prognosis and survival in HCC patients. ANLN knockdown was shown to inhibit proliferation, cell cycle progression, and PD-L1 expression in liver cancer cells. The antipsychotic drug penfluridol was identified to suppress ANLN expression in the Connectivity Map analysis. Penfluridol downregulated ANLN at both the mRNA and protein levels, leading to G2/M cell cycle arrest and reduced colony formation in liver cancer cells. Mechanistically, penfluridol inhibited the transcription factor MYC from binding to an E-box motif in the ANLN promoter. This process was mediated by penfluridol-induced upregulation of NRF2, which competitively bound and sequestered MYC away from the ANLN promoter. Penfluridol inhibited the interaction between NRF2 and KEAP1, increasing NRF2. In a syngeneic mouse model, penfluridol suppressed liver tumour growth accompanied by increased NRF2 and decreased MYC and ANLN expression. These findings suggest penfluridol can be applied as the first ANLN blocker to modulate the MYC/NRF2/KEAP1 axis.
Collapse
Affiliation(s)
- Minh Tuan Nguyen
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Gi Jeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Boram Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Hyun Ji Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Jihoon Tak
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Mi Kyung Park
- Department of Bio-Healthcare, Hwasung Medi-Science University, Hwaseong-si 18274, Republic of Korea
| | - Eun Ji Kim
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gyeoung Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Seung Bae Rho
- National Cancer Center, Goyang 10408, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang 10408, Republic of Korea
| | - Kyung Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Sang Geon Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Chang Hoon Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
4
|
Li Y, Cao B, Lin M, Xu J, Qi S, Wang J, Xiao X, Li G, Li C. An integrative lipidomics and transcriptomics study revealing Bavachin and Icariin synergistically induce idiosyncratic liver injury. Immunopharmacol Immunotoxicol 2024; 46:924-934. [PMID: 39505304 DOI: 10.1080/08923973.2024.2424293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
Objectives: Reports of traditional Chinese medicine (TCM)-related liver injury have increased over recent years; however, identifying susceptibility-related components and biomarkers remains challenging due to the heterogeneous nature of TCM and idiosyncratic drug-induced liver injury (IDILI). Psoraleae Fructus (PF) and Epimedii Folium (EF), commonly found in TCM prescriptions, have been implicated in IDILI, but their constituents and underlying mechanisms are poorly understood. Methods: In this study, we identified bavachin (Bav) and icariin (Ica) as susceptibility components for IDILI in PF and EF using a TNF-α-mediated mouse model. Lipidomics and transcriptomics were used to investigate their related mechanism. Results: Liver biochemistry and histopathology analyses revealed that co-exposure to Bav, Ica, and a non-toxic dose of TNF-α prestimulation induced significant liver injury, while Bav and Ica alone did not. Lipidomics identified seven differentially abundant metabolites in the Bav/Ica/TNF-α group compared to the Ica/TNF-α or Bav/TNF-α groups, mainly enriched in alpha-linolenic acid (ALA), arachidonic acid (AA), and linoleic acid (LA) metabolic pathways. Additionally, transcriptomics revealed 49 differentially expressed genes (DEGs) in the Bav/TNF-α vs Bav/Ica/TNF-α and Ica/TNF-α vs Bav/Ica/TNF-α groups, primarily associated with the PI3K/AKT/mTOR signaling pathway and sphingolipid metabolism. Integrative lipidomics and transcriptomics analyses identified significant positive correlations between five differential metabolites (DMs) - PC (O-16:0_14:1), PG (22:1_20:3), PI (16:0_14:1), PS (18:0_19:2), and TG (17:0_18:2_22:5) - and ten DEGs - Nr0b2, Btbd19, Btg2, Fam222a, Fam83f, Gtse1, Anln, Gja4, Srrm4, and Zfp13. Conslusions: Collectively, these results suggest that alterations in intracellular metabolism and gene expression levels may contribute to the synergistic induction of IDILI by the incompatible pair Bav and Ica in the presence of TNF-α.
Collapse
Affiliation(s)
- Yingying Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Cao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Lin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuya Qi
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiabo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaohe Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guohui Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyu Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Morris JP, Baslan T, Soltis DE, Soltis PS, Fox DT. Integrating the Study of Polyploidy Across Organisms, Tissues, and Disease. Annu Rev Genet 2024; 58:297-318. [PMID: 39227132 PMCID: PMC11590481 DOI: 10.1146/annurev-genet-111523-102124] [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] [Indexed: 09/05/2024]
Abstract
Polyploidy is a cellular state containing more than two complete chromosome sets. It has largely been studied as a discrete phenomenon in either organismal, tissue, or disease contexts. Increasingly, however, investigation of polyploidy across disciplines is coalescing around common principles. For example, the recent Polyploidy Across the Tree of Life meeting considered the contribution of polyploidy both in organismal evolution over millions of years and in tumorigenesis across much shorter timescales. Here, we build on this newfound integration with a unified discussion of polyploidy in organisms, cells, and disease. We highlight how common polyploidy is at multiple biological scales, thus eliminating the outdated mindset of its specialization. Additionally, we discuss rules that are likely common to all instances of polyploidy. With increasing appreciation that polyploidy is pervasive in nature and displays fascinating commonalities across diverse contexts, inquiry related to this important topic is rapidly becoming unified.
Collapse
Affiliation(s)
- John P Morris
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA;
| | - Timour Baslan
- Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology and Translational Therapeutics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Sciences and Penn Vet Cancer Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Douglas E Soltis
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, USA;
- Biodiversity Institute, University of Florida, Gainesville, Florida, USA
- Polyploidy Integration and Innovation Institute
- Department of Biology, University of Florida, Gainesville, Florida, USA;
| | - Pamela S Soltis
- Biodiversity Institute, University of Florida, Gainesville, Florida, USA
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, USA;
- Polyploidy Integration and Innovation Institute
| | - Donald T Fox
- Department of Pharmacology and Cancer Biology, Duke Regeneration Center, and Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA;
- Polyploidy Integration and Innovation Institute
| |
Collapse
|
6
|
Yin K, Büttner M, Deligiannis IK, Strzelecki M, Zhang L, Talavera-López C, Theis F, Odom DT, Martinez-Jimenez CP. Polyploidisation pleiotropically buffers ageing in hepatocytes. J Hepatol 2024; 81:289-302. [PMID: 38583492 DOI: 10.1016/j.jhep.2024.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND & AIMS Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS We performed single-nucleus RNA sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent strains that were haploinsufficient for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS We observed that ageing in wild-type mice results in nuclei polyploidy and a marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in the CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A-haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbations. CONCLUSIONS Our results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome. IMPACT AND IMPLICATIONS The functional role of hepatocyte polyploidisation during ageing is poorly understood. Using single-nucleus RNA sequencing and BaseScope approaches, we have studied ploidy dynamics during ageing in murine livers with non-deleterious genetic perturbations. We have identified that hepatocytes present different cellular states and the ability to buffer ageing-associated dysfunctions. Tetraploid nuclei exhibit robust transcriptional networks and are better adapted to genomically overcome perturbations. Novel therapeutic interventions aimed at attenuating age-related changes in tissue function could be exploited by manipulation of ploidy dynamics during chronic liver conditions.
Collapse
Affiliation(s)
- Kelvin Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany
| | | | | | - Liwei Zhang
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Carlos Talavera-López
- Division of Infectious Diseases and Tropical Medicine, Ludwig-Maximilian-Universität Klinikum, Germany
| | - Fabian Theis
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching. Munich, Germany; German Cancer Research Centre, Heidelberg, Germany.
| | - Duncan T Odom
- German Cancer Research Center, Division of Regulatory Genomics and Cancer Evolution (B270), Heidelberg, Germany; Cancer Research UK Cambridge Institute, University of Cambridge, CB20RE, United Kingdom.
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany; Institute of Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
7
|
Kim J, Seki E. Unveiling the cancer risk nexus of the steatotic liver. Trends Endocrinol Metab 2024; 35:708-719. [PMID: 38531699 PMCID: PMC11321945 DOI: 10.1016/j.tem.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
Steatotic liver, characterized by the accumulation of fat in the liver, poses significant health risks including metabolic dysfunction-associated steatotic liver disease (MASLD) and an elevated risk of primary liver cancer. Emerging evidence indicates a robust association between steatotic liver and increased susceptibility to extrahepatic primary cancers and their metastases. The deposition of fat induces dynamic changes in hepatic microenvironments, thereby fostering inflammation and immune responses that enhance liver metastasis from extrahepatic primary cancers. This review explores the impact of steatotic liver on hepatic carcinogenesis and metastasis from extrahepatic cancers, with a specific focus on hepatocyte-derived factors and the immune microenvironment. By emphasizing novel conclusions, this article underscores the timely relevance of understanding these intricate connections.
Collapse
Affiliation(s)
- Jieun Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
8
|
Zhang Y, Lu Y, Wang N, Yang Y, Hao F, Fei X, Chen Y, Wang J. Alternative splicing-related long noncoding RNA ANRIL facilitates hepatocellular carcinoma by targeting the miR-199a-5p/SRSF1 axis and impacting Anillin. Mol Carcinog 2024; 63:1064-1078. [PMID: 38411272 DOI: 10.1002/mc.23709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by aberrant alternative splicing (AS), which plays an important part in the pathological process of this disease. However, available reports about genes and mechanisms involved in AS process are limited. Our previous research has identified ANRIL as a long noncoding RNA related to the AS process of HCC. Here, we investigated the exact effect and the mechanism of ANRIL on HCC progress. The ANRIL expression profile was validated using the real-time quantitative polymerase chain reaction assay. The western blot analysis and IHC assay were conducted on candidate targets, including SRSF1 and Anillin. The clinicopathological features of 97 patients were collected and analyzed. Loss-of and gain-of-function experiments were conducted. The dual-luciferase reporter assay was applied to verify the interaction between ANRIL, miR-199a-5p, and SRSF1. Anomalous upregulation of ANRIL in HCC was observed, correlating with worse clinicopathological features of HCC. HCC cell proliferation, mobility, tumorigenesis, and metastasis were impaired by depleting ANRIL. We found that ANRIL acts as a sponger of miRNA-199a-5p, resulting in an elevated level of its target protein SRSF1. The phenotypes induced by ANRIL/miR-199a-5p/SRSF1 alteration are associated with Anillin, a validated HCC promoter. ANRIL is an AS-related lncRNA promoting HCC progress by modulating the miR-199a-5p/SRSF1 axis. The downstream effector of this axis in the development of HCC is Anillin.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yiquan Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Nan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yuchen Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
9
|
Johnson JR, Martini RN, Yuan YC, Woods-Burnham L, Walker M, Ortiz-Hernandez GL, Kobeissy F, Galloway D, Gaddy A, Oguejiofor C, Allen B, Lewis D, Davis MB, Kimbro KS, Yates CC, Murphy AB, Kittles RA. 1,25-Dihydroxyvitamin D 3 Suppresses Prognostic Survival Biomarkers Associated with Cell Cycle and Actin Organization in a Non-Malignant African American Prostate Cell Line. BIOLOGY 2024; 13:346. [PMID: 38785827 PMCID: PMC11118023 DOI: 10.3390/biology13050346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Vitamin D3 is a steroid hormone that confers anti-tumorigenic properties in prostate cells. Serum vitamin D3 deficiency has been associated with advanced prostate cancer (PCa), particularly affecting African American (AA) men. Therefore, elucidating the pleiotropic effects of vitamin D on signaling pathways, essential to maintaining non-malignancy, may provide additional drug targets to mitigate disparate outcomes for men with PCa, especially AA men. We conducted RNA sequencing on an AA non-malignant prostate cell line, RC-77N/E, comparing untreated cells to those treated with 10 nM of vitamin D3 metabolite, 1α,25(OH)2D3, at 24 h. Differential gene expression analysis revealed 1601 significant genes affected by 1α,25(OH)2D3 treatment. Pathway enrichment analysis predicted 1α,25(OH)2D3- mediated repression of prostate cancer, cell proliferation, actin cytoskeletal, and actin-related signaling pathways (p < 0.05). Prioritizing genes with vitamin D response elements and associating expression levels with overall survival (OS) in The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA PRAD) cohort, we identified ANLN (Anillin) and ECT2 (Epithelial Cell Transforming 2) as potential prognostic PCa biomarkers. Both genes were strongly correlated and significantly downregulated by 1α,25(OH)2D3 treatment, where low expression was statistically associated with better overall survival outcomes in the TCGA PRAD public cohort. Increased ANLN and ECT2 mRNA gene expression was significantly associated with PCa, and Gleason scores using both the TCGA cohort (p < 0.05) and an AA non-malignant/tumor-matched cohort. Our findings suggest 1α,25(OH)2D3 regulation of these biomarkers may be significant for PCa prevention. In addition, 1α,25(OH)2D3 could be used as an adjuvant treatment targeting actin cytoskeleton signaling and actin cytoskeleton-related signaling pathways, particularly among AA men.
Collapse
Affiliation(s)
- Jabril R. Johnson
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Rachel N. Martini
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Yate-Ching Yuan
- Department of Computational Quantitative Medicine, Center for Informatics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Leanne Woods-Burnham
- Department of Physiology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Mya Walker
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Greisha L. Ortiz-Hernandez
- Department of Population Sciences, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr SW, Atlanta, GA 30310, USA
| | - Dorothy Galloway
- Department of Population Sciences, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Amani Gaddy
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Chidinma Oguejiofor
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Blake Allen
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Deyana Lewis
- Department of Community Health and Preventive Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Melissa B. Davis
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - K. Sean Kimbro
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| | - Clayton C. Yates
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Adam B. Murphy
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rick A. Kittles
- Department of Community Health and Preventive Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310, USA
| |
Collapse
|
10
|
Su Y, Lin H, Yu J, Mao L, Jin W, Liu T, Jiang S, Wu Y, Zhang S, Geng Q, Ge C, Zhao F, Chen T, Cui Y, Li J, Hou H, Zhou X, Li H. RIT1 regulates mitosis and promotes proliferation by interacting with SMC3 and PDS5 in hepatocellular carcinoma. J Exp Clin Cancer Res 2023; 42:326. [PMID: 38017479 PMCID: PMC10685607 DOI: 10.1186/s13046-023-02892-x] [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: 07/30/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND As a small G protein of Ras family, Ras-like-without-CAAX-1 (RIT1) plays a critical role in various tumors. Our previous study has demonstrated the involvement of RIT1 in promoting malignant progression of hepatocellular carcinoma (HCC). However, its underlying mechanism remains unclear. METHODS Gene set enrichment analysis (GSEA) was conducted in the TCGA LIHC cohort to investigate the underlying biological mechanism of RIT1. Live cell imaging, immunofluorescence (IF) and flow cytometry assays were used to verify biological function of RIT1 in HCC mitosis. Subcutaneous xenografting of human HCC cells in BALB/c nude mice was utilized to assess tumor proliferation in vivo. RNA-seq, co-immunoprecipitation (Co-IP), mass spectrometry analyses, western blot and IF assays were employed to elucidate the mechanisms by which RIT1 regulates mitosis and promotes proliferation in HCC. RESULTS Our findings demonstrate that RIT1 plays a crucial role in regulating mitosis in HCC. Knockdown of RIT1 disrupts cell division, leading to G2/M phase arrest, mitotic catastrophe, and apoptosis in HCC cells. SMC3 is found to interact with RIT1 and knockdown of SMC3 attenuates the proliferative effects mediated by RIT1 both in vitro and in vivo. Mechanistically, RIT1 protects and maintains SMC3 acetylation by binding to SMC3 and PDS5 during mitosis, thereby promoting rapid cell division and proliferation in HCC. Notably, we have observed an upregulation of SMC3 expression in HCC tissues, which is associated with poor patient survival and promotion of HCC cell proliferation. Furthermore, there is a significant positive correlation between the expression levels of RIT1, SMC3, and PDS5. Importantly, HCC patients with high expression of both RIT1 and SMC3 exhibit worse prognosis compared to those with high RIT1 but low SMC3 expression. CONCLUSIONS Our findings underscore the crucial role of RIT1 in regulating mitosis in HCC and further demonstrate its potential as a promising therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Yang Su
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Hechun Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Junming Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Lin Mao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Wenjiao Jin
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Tengfei Liu
- Department of Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Shuqing Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Yunyu Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Saihua Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Qin Geng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Chao Ge
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Fangyu Zhao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Taoyang Chen
- Qidong Liver Cancer Institute, Qidong, 226200, China
| | - Ying Cui
- Cancer Institute of Guangxi, Nanning, 530021, China
| | - Jinjun Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Xinli Zhou
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Hong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| |
Collapse
|
11
|
Zhang Y, Wang N, Hao F, Chen Y, Fei X, Wang J. Attenuation of binuclear hepatocytes in the paracancerous liver tissue is associated with short-term recurrence of hepatocellular carcinoma post-radical surgery. FASEB J 2023; 37:e23271. [PMID: 37882195 DOI: 10.1096/fj.202301219r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/03/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
Short-term recurrence of hepatocellular carcinoma (HCC) after radical resection leads to dismal outcomes. To screen high-recurrence risk patients to provide adjuvant treatment is necessary. Herein, based on our previous research, we further focused on the changes in the abundance of binuclear hepatocytes (ABH) in the paracancerous liver tissue to discuss the relationship between the attenuation of binuclear hepatocytes and postoperative short-term recurrence, by combining with the assessment of the value of a reported independent early recurrence risk factor in HCC, protein induced by vitamin K absence or antagonist-II (PIVKA-II). A cohort of 142 paracancerous liver tissues from HCC patients who received radical resection was collected. Binuclear hepatocytes were reduced in the paracancerous liver tissues, compared with the liver tissues from normal donors. ABH was negatively correlated with clinical features such as tumor size, TNM stages, tumor microsatellite formation, venous invasion, and Alpha-fetoprotein (AFP) level, as well as the expression of E2F7 and Anillin, which are two critical regulators concerning the hepatocyte polyploidization. According to the short-term recurrence information, ABH value was laminated, and univariate and multivariate logistic regression was performed to analyze the relationship between paracancerous ABH and short-term tumor relapse. Simultaneously, the predictive effectiveness of the ABH value was compared with the preoperative PIVKA-II value. As observed, the paracancerous ABH value below 1.5% was found to be an independent risk factor for recurrence. In conclusion, the paracancerous ABH is a credible indicator of short-term recurrence of HCC patients after radical resection, and regular assessment of ABH might help to prevent short-term HCC recurrence.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Nan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| |
Collapse
|
12
|
Bloomfield M, Cimini D. The fate of extra centrosomes in newly formed tetraploid cells: should I stay, or should I go? Front Cell Dev Biol 2023; 11:1210983. [PMID: 37576603 PMCID: PMC10413984 DOI: 10.3389/fcell.2023.1210983] [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: 04/23/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
An increase in centrosome number is commonly observed in cancer cells, but the role centrosome amplification plays along with how and when it occurs during cancer development is unclear. One mechanism for generating cancer cells with extra centrosomes is whole genome doubling (WGD), an event that occurs in over 30% of human cancers and is associated with poor survival. Newly formed tetraploid cells can acquire extra centrosomes during WGD, and a generally accepted model proposes that centrosome amplification in tetraploid cells promotes cancer progression by generating aneuploidy and chromosomal instability. Recent findings, however, indicate that newly formed tetraploid cells in vitro lose their extra centrosomes to prevent multipolar cell divisions. Rather than persistent centrosome amplification, this evidence raises the possibility that it may be advantageous for tetraploid cells to initially restore centrosome number homeostasis and for a fraction of the population to reacquire additional centrosomes in the later stages of cancer evolution. In this review, we explore the different evolutionary paths available to newly formed tetraploid cells, their effects on centrosome and chromosome number distribution in daughter cells, and their probabilities of long-term survival. We then discuss the mechanisms that may alter centrosome and chromosome numbers in tetraploid cells and their relevance to cancer progression following WGD.
Collapse
Affiliation(s)
- Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Daniela Cimini
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
13
|
Saito Y, Yin D, Kubota N, Wang X, Filliol A, Remotti H, Nair A, Fazlollahi L, Hoshida Y, Tabas I, Wangensteen KJ, Schwabe RF. A Therapeutically Targetable TAZ-TEAD2 Pathway Drives the Growth of Hepatocellular Carcinoma via ANLN and KIF23. Gastroenterology 2023; 164:1279-1292. [PMID: 36894036 PMCID: PMC10335360 DOI: 10.1053/j.gastro.2023.02.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/24/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND & AIMS Despite recent progress, long-term survival remains low for hepatocellular carcinoma (HCC). The most effective HCC therapies target the tumor immune microenvironment (TIME), and there are almost no therapies that directly target tumor cells. Here, we investigated the regulation and function of tumor cell-expressed Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in HCC. METHODS HCC was induced in mice by Sleeping Beauty-mediated expression of MET, CTNNB1-S45Y, or TAZ-S89A, or by diethylnitrosamine plus CCl4. Hepatocellular TAZ and YAP were deleted in floxed mice via adeno-associated virus serotype 8-mediated expression of Cre. TAZ target genes were identified from RNA sequencing, confirmed by chromatin immunoprecipitation, and evaluated in a clustered regularly interspaced short palindromic repeats interference (CRISPRi) screen. TEA domain transcription factors (TEADs), anillin (ANLN), Kif23, and programmed cell death protein ligand 1 were knocked down by guide RNAs in dead clustered regularly interspaced short palindromic repeats-associated protein 9 (dCas9) knock-in mice. RESULTS YAP and TAZ were up-regulated in murine and human HCC, but only deletion of TAZ consistently decreased HCC growth and mortality. Conversely, overexpression of activated TAZ was sufficient to trigger HCC. TAZ expression in HCC was regulated by cholesterol synthesis, as demonstrated by pharmacologic or genetic inhibition of 3-hydroxy-3-methylglutaryl- coenzyme A reductase (HMGCR), farnesyl pyrophosphate synthase, farnesyl-diphosphate farnesyltransferase 1 (FDFT1), or sterol regulatory element-binding protein 2 (SREBP2). TAZ- and MET/CTNNB1-S45Y-driven HCC required the expression of TEAD2 and, to a lesser extent, TEAD4. Accordingly, TEAD2 displayed the most profound effect on survival in patients with HCC. TAZ and TEAD2 promoted HCC via increased tumor cell proliferation, mediated by TAZ target genes ANLN and kinesin family member 23 (KIF23). Therapeutic targeting of HCC, using pan-TEAD inhibitors or the combination of a statin with sorafenib or anti-programmed cell death protein 1, decreased tumor growth. CONCLUSIONS Our results suggest the cholesterol-TAZ-TEAD2-ANLN/KIF23 pathway as a mediator of HCC proliferation and tumor cell-intrinsic therapeutic target that could be synergistically combined with TIME-targeted therapies.
Collapse
Affiliation(s)
- Yoshinobu Saito
- Department of Medicine, Columbia University, New York, New York.
| | - Dingzi Yin
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Mayo Clinic, Rochester, Minnesota
| | - Naoto Kubota
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiaobo Wang
- Department of Medicine, Columbia University, New York, New York
| | - Aveline Filliol
- Department of Medicine, Columbia University, New York, New York
| | - Helen Remotti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Ajay Nair
- Department of Medicine, Columbia University, New York, New York
| | - Ladan Fazlollahi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ira Tabas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York; Institute of Human Nutrition, New York, New York
| | - Kirk J Wangensteen
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Mayo Clinic, Rochester, Minnesota.
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, New York; Institute of Human Nutrition, New York, New York.
| |
Collapse
|
14
|
Gong L, Zhong M, Gong K, Wang Z, Zhong Y, Jin Y, Chen H, Tai P, Chen X, Chen A, Cao K. Multi-Omics Analysis and Verification of the Oncogenic Value of CCT8 in Pan-Cancers. J Inflamm Res 2023; 16:2297-2315. [PMID: 37273485 PMCID: PMC10238552 DOI: 10.2147/jir.s403499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/06/2023] [Indexed: 06/06/2023] Open
Abstract
Background Chaperonin-containing TCP1 subunit 8 (CCT8) has been proved to be involved in the occurrence and development of some cancers. However, no study has reported the potential role of CCT8 in a pan-cancer manner. Methods TIMER2.0, GEPIA2, UALCAN and Sangerbox were used to explore the expression, prognosis and methylation of CCT8. We used cBioPortal, TISIDB, SangerBox, TIMER2.0 and TISMO to investigate the genetic alteration of CCT8 and the relationship of CCT8 with molecular subtype, immune subtype, immune infiltration and immunotherapy response. CCT8-related genes were screened out through GEPIA and STRING for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. CCK-8, the colony formation assay, the wound healing assay and the Transwell assay were performed to explore the influence of CCT8 on proliferation and migration. Results CCT8 was highly expressed in most cancers with a poor prognosis. The expression level of CCT8, which was affected by the promoter region methylation and genetic alteration, was related to the molecular and immune subtype of cancers. Interestingly, CCT8 was positively associated with the activated CD4 T cells and type 2 T-helper cells. CCT8 played a vital role in the cell cycle and RNA transport of cancers, and it significantly inhibited the proliferation and migration of lung adenocarcinoma cells when it was knocked down. Conclusion CCT8 plays an indispensable role in promoting the proliferation and migration of many cancers. CCT8 might be a biomarker of T-helper type 2 (Th2) cell infiltration and a promising therapeutic target for T-helper type 1(Th1)/Th2 imbalance.
Collapse
Affiliation(s)
- Lian Gong
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Ming Zhong
- Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Kai Gong
- Department of Clinical Medicine, Xiangnan University, Chenzhou, People’s Republic of China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Yong Zhong
- Department of Clinical Medicine, Hubei Enshi College, Enshi, People’s Republic of China
| | - Yi Jin
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Haotian Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Panpan Tai
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Xinyu Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Aiyan Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, People’s Republic of China
| |
Collapse
|
15
|
Yang T, Chi Z, Liu G, Hong X, Cao S, Cheng K, Zhang Y. Screening ANLN and ASPM as bladder urothelial carcinoma-related biomarkers based on weighted gene co-expression network analysis. Front Genet 2023; 14:1107625. [PMID: 37051591 PMCID: PMC10083327 DOI: 10.3389/fgene.2023.1107625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Introduction: Bladder cancer (BLCA) is one of the most common malignancies in the urinary system with a poor prognosis and high treatment costs. Identifying potential prognostic biomarkers is significant for exploring new therapeutic and predictive targets of BLCA.Methods: In this study, we screened differentially expressed genes using the GSE37815 dataset. We then performed a weighted gene co‐expression network analysis (WGCNA) to identify the genes correlated with the histologic grade and T stage of BLCA using the GSE32548 dataset. Subsequently, Kaplan Meier survival analysis and Cox regression were used to further identify prognosis‐related hub genes using the datasets GSE13507 and TCGA‐BLCA. Moreover, we detected the expression of the hub genes in 35 paired samples, including BLCA and paracancerous tissue, from the Shantou Central Hospital by qRT‐polymerase chain reaction.Results: This study showed that Anillin (ANLN) and Abnormal spindle-like microcephaly-associated gene (ASPM) were prognostic biomarkers for BLCA. High expression of ANLN and ASPM was associated with poor overall survival.The qRT‐PCR results revealed that ANLN and ASPM genes were upregulated in BLCA, and there was a correlation between the expression of ANLN and ASPM in cancer tissues and paracancerous tissue. Additionally, the increasing multiples in the ANLN gene was obvious in high-grade BLCA.Discussion: In summary, this preliminary exploration indicated a correlation between ANLN and ASPM expression. These two genes, serving as the risk factors for BLCA progression, might be promising targets to improve the occurrence and progression of BLCA.
Collapse
|
16
|
Bernhard SV, Gemble S, Basto R, Storchova Z. Experimental Approaches to Generate and Isolate Human Tetraploid Cells. Methods Mol Biol 2023; 2545:391-399. [PMID: 36720824 DOI: 10.1007/978-1-0716-2561-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer cells are frequently affected by large-scale chromosome copy number changes, such as polyploidy or whole chromosome aneuploidy, and thus understanding the consequences of these changes is important for cancer research. In the past, it has been difficult to study the consequences of large-scale genomic changes, especially in pure isogenic populations. Here, we describe two methods to generate tetraploid cells induced either by cytokinesis failure or mitotic slippage. These treatments result in mixed population of diploids and tetraploids that can be analyzed directly. Alternatively, tetraploid populations can be established by single cell clone selection or by fluorescence activated cell sorting. These methods enable to analyze and compare the consequences of whole-genome doubling between the parental cell line, freshly arising tetraploid cells, and post-tetraploid aneuploid clones.
Collapse
Affiliation(s)
| | - Simon Gemble
- Institut Curie, PSL Research University, CNRS, UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Renata Basto
- Institut Curie, PSL Research University, CNRS, UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Zuzana Storchova
- Department of Molecular Genetics, Paul Ehrlich Strasse 24, Kaiserslautern, Germany.
| |
Collapse
|
17
|
Samant RS, Batista S, Larance M, Ozer B, Milton CI, Bludau I, Wu E, Biggins L, Andrews S, Hervieu A, Johnston HE, Al-Lazikhani B, Lamond AI, Clarke PA, Workman P. Native Size-Exclusion Chromatography-Based Mass Spectrometry Reveals New Components of the Early Heat Shock Protein 90 Inhibition Response Among Limited Global Changes. Mol Cell Proteomics 2023; 22:100485. [PMID: 36549590 PMCID: PMC9898794 DOI: 10.1016/j.mcpro.2022.100485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
The molecular chaperone heat shock protein 90 (HSP90) works in concert with co-chaperones to stabilize its client proteins, which include multiple drivers of oncogenesis and malignant progression. Pharmacologic inhibitors of HSP90 have been observed to exert a wide range of effects on the proteome, including depletion of client proteins, induction of heat shock proteins, dissociation of co-chaperones from HSP90, disruption of client protein signaling networks, and recruitment of the protein ubiquitylation and degradation machinery-suggesting widespread remodeling of cellular protein complexes. However, proteomics studies to date have focused on inhibitor-induced changes in total protein levels, often overlooking protein complex alterations. Here, we use size-exclusion chromatography in combination with mass spectrometry (SEC-MS) to characterize the early changes in native protein complexes following treatment with the HSP90 inhibitor tanespimycin (17-AAG) for 8 h in the HT29 colon adenocarcinoma cell line. After confirming the signature cellular response to HSP90 inhibition (e.g., induction of heat shock proteins, decreased total levels of client proteins), we were surprised to find only modest perturbations to the global distribution of protein elution profiles in inhibitor-treated HT29 cells at this relatively early time-point. Similarly, co-chaperones that co-eluted with HSP90 displayed no clear difference between control and treated conditions. However, two distinct analysis strategies identified multiple inhibitor-induced changes, including known and unknown components of the HSP90-dependent proteome. We validate two of these-the actin-binding protein Anillin and the mitochondrial isocitrate dehydrogenase 3 complex-as novel HSP90 inhibitor-modulated proteins. We present this dataset as a resource for the HSP90, proteostasis, and cancer communities (https://www.bioinformatics.babraham.ac.uk/shiny/HSP90/SEC-MS/), laying the groundwork for future mechanistic and therapeutic studies related to HSP90 pharmacology. Data are available via ProteomeXchange with identifier PXD033459.
Collapse
Affiliation(s)
- Rahul S Samant
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom; Signalling Programme, The Babraham Institute, Cambridge, United Kingdom.
| | - Silvia Batista
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Mark Larance
- Centre for Gene Regulation & Expression, University of Dundee, Dundee, United Kingdom
| | - Bugra Ozer
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Christopher I Milton
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Isabell Bludau
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Estelle Wu
- Signalling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Laura Biggins
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Alexia Hervieu
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Harvey E Johnston
- Signalling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Bissan Al-Lazikhani
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Angus I Lamond
- Centre for Gene Regulation & Expression, University of Dundee, Dundee, United Kingdom
| | - Paul A Clarke
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Paul Workman
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
18
|
Cao YF, Xie L, Tong BB, Chu MY, Shi WQ, Li X, He JZ, Wang SH, Wu ZY, Deng DX, Zheng YQ, Li ZM, Xu XE, Liao LD, Cheng YW, Li LY, Xu LY, Li EM. Targeting USP10 induces degradation of oncogenic ANLN in esophageal squamous cell carcinoma. Cell Death Differ 2023; 30:527-543. [PMID: 36526897 PMCID: PMC9950447 DOI: 10.1038/s41418-022-01104-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Anillin (ANLN) is a mitosis-related protein that promotes contractile ring formation and cytokinesis, but its cell cycle-dependent degradation mechanisms in cancer cells remain unclear. Here, we show that high expression of ANLN promotes cytokinesis and proliferation in esophageal squamous cell carcinoma (ESCC) cells and is associated with poor prognosis in ESCC patients. Furthermore, the findings of the study showed that the deubiquitinating enzyme USP10 interacts with ANLN and positively regulates ANLN protein levels. USP10 removes the K11- and K63-linked ubiquitin chains of ANLN through its deubiquitinase activity and prevents ANLN ubiquitin-mediated degradation. Importantly, USP10 promotes contractile ring assembly at the cytokinetic furrow as well as cytokinesis by stabilizing ANLN. Interestingly, USP10 and the E3 ubiquitin ligase APC/C co-activator Cdh1 formed a functional complex with ANLN in a non-competitive manner to balance ANLN protein levels. In addition, the macrolide compound FW-04-806 (F806), a natural compound with potential for treating ESCC, inhibited the mitosis of ESCC cells by targeting USP10 and promoting ANLN degradation. F806 selectively targeted USP10 and inhibited its catalytic activity but did not affect the binding of Cdh1 to ANLN and alters the balance of the USP10-Cdh1-ANLN complex. Additionally, USP10 expression was positively correlated with ANLN level and poor prognosis of ESCC patients. Overall, targeting the USP10-ANLN axis can effectively inhibit ESCC cell-cycle progression.
Collapse
Affiliation(s)
- Yu-Fei Cao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Lei Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Bei-Bei Tong
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Man-Yu Chu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Wen-Qi Shi
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong, PR China
| | - Xiang Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Jian-Zhong He
- Department of Pathology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, PR China
| | - Shao-Hong Wang
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong, PR China
| | - Zhi-Yong Wu
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong, PR China
| | - Dan-Xia Deng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Ya-Qi Zheng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Zhi-Mao Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Xiu-E Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Yin-Wei Cheng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Li-Yan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong, PR China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| |
Collapse
|
19
|
Zheng H, Cheng ZJ, Liang B, Wang ZG, Tao YP, Huang SY, Ni JS, Li HF, Yang L, Yuan SX, Wu J, Kawaguchi T, Samant H, Zhou WP, Xiang DM, Yang Y. N 6-Methyladenosine Modification of ANLN Enhances Hepatocellular Carcinoma Bone Metastasis. Int J Biol Sci 2023; 19:1009-1023. [PMID: 36923927 PMCID: PMC10008695 DOI: 10.7150/ijbs.73570] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 12/28/2022] [Indexed: 02/04/2023] Open
Abstract
Bones are categorized as the second most prevalent location of extra-hepatic metastasis in Hepatocellular Carcinoma (HCC), which is linked to an extremely poor prognosis due to limited therapeutic options. N6-methyladenosine (m6A) is a prominent modification involved in HCC, but the exact mechanisms on how m6A modifications induce HCC bone metastases (BM) remain unclear. The key modulators responsible for the abundant m6A RNA modification-induced HCC BM was found to be the METTL3 and YTHDF1. The expression of Anillin actin-binding protein (ANLN) was dramatically higher in HCC with BM tissues, and its messenger RNA (mRNA) stability was enhanced via m6A epitranscriptomic regulation by METTL3 and YTHDF1. High METTL3 and YTHDF1 expression along with nuclear ANLN protein was clinically correlated with BM in HCC patients. Furthermore, HCC BM was attributed to over-expression of nuclear ANLN forming a transcriptional complex with SP1 which enhanced KIF2C transcriptional activity to activate the mTORC1 pathway, therefore increased the expression of RANKL and disproportionated RANKL-OPG expression in bone microenvironment leading to malignant neoplasms invade bone tissue. In addition, inhibition of ANLN m6A modification by DZNeP attenuated HCC BM. This data provides meaningful understanding of the modulation and association of m6A epitranscriptomic-regulated BM in HCC, and moreover, defines potentially valuable therapeutic targets.
Collapse
Affiliation(s)
- Hao Zheng
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Zhang-Jun Cheng
- Department of Hepato-Pancreato-Biliary Centers, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bo Liang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Zhen-Guang Wang
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Yuan-Ping Tao
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Sheng-Yu Huang
- Department of Hepatobiliary and Pancreatic Surgery, The 10th People's Hospital, Tongji University, Shanghai 200433, China
| | - Jun-Sheng Ni
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Hui-Fen Li
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Le Yang
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Sheng-Xian Yuan
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Jennifer Wu
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hrishikesh Samant
- Hrishikesh Samant, Division of Gastroenterology and Hepatology, LSU Health Science Center, Shreveport, LA, USA
| | - Wei-Ping Zhou
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| | - Dai-Min Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yuan Yang
- Third Department of Hepatic Surgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China.,Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, China.,Department of Organization Sample Bank, Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, China
| |
Collapse
|
20
|
Hao F, Wang N, Zhang Y, Xu W, Chen Y, Fei X, Wang J. E2F7 enhances hepatocellular carcinoma growth by preserving the SP1/SOX4/Anillin axis via repressing miRNA-383-5p transcription. Mol Carcinog 2022; 61:975-988. [PMID: 35924788 PMCID: PMC9804269 DOI: 10.1002/mc.23454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/20/2022] [Accepted: 07/22/2022] [Indexed: 01/05/2023]
Abstract
E2F family participates in most human malignancies by activating the transcription of the cell cycle-related genes. Whereas, as a specifical atypical member of this family, E2F7 was described as a repressor against its downstream genes and exerted oscillatory and controversial functions in cancers. Our previous study identified a molecular interaction promoting hepatocellular carcinoma (HCC) growth induced by SOX4 and Anillin. Meanwhile, we preliminarily identified SP1 as the upstream activator of SOX4. Intriguingly, we observed that the repressive E2F7 presents a remarkable high expression in HCC, and is positively correlated and involved in the same pathway with the potentially SP1/SOX4/Anillin axis. However, their exact interaction or mechanism controlling tumor progress between these genes has not been illustrated. Thus, we focused on this point in this study and attempted to improve the potential regulating axis in HCC cell proliferation and tumor growth for promoting tumor prevention and control. The expression profile of E2F7 in HCC tissues and tumor cells was detected along with the related candidate genes, through real-time quantitative polymerase chain reaction assay, the Western blot analysis, and the immunohistochemistry assay, combined with bioinformatics analysis of the HCC information from the the Cancer Genome Altas and Gene Expression Omnibus data sets. The correlation between E2F7 and HCC patients' clinicopathologic features was explored. Gain-of and loss-of-function assays were conducted both in vitro and in vivo along with the rescue experiment, for revealing the relative genes' functions in HCC progress. The ChIP and the dual-luciferase reporter assays were performed to verify the transcriptional regulating profile between E2F7 and SP1/SOX4/Anillin axis. E2F7 was upregulated in HCC and significantly correlated with SP1/SOX4/Anillin axis. High E2F7 expression is associated with dismal clinicopathologic features and poor survival of the patients. E2F7 depletion potently impaired SP1/SOX4/Anillin expression and significantly inhibited HCC growth. Furthermore, intensive exploration demonstrated that E2F7 preserves high SP1 levels by abrogating miR-383-5p in a transcriptional way. Atypical E2F7 is an important repressive transcription factor commonly upregulated in the HCC environment. E2F7 facilitates HCC growth by repressing miR-383-5p transcription and sequentially promoting SP1/SOX4/Anillin axis. Our findings provide us with probable targets for HCC prevention and therapeutic treatment.
Collapse
Affiliation(s)
- Fengjie Hao
- Department of General SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| | - Nan Wang
- Department of General SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| | - Yifan Zhang
- Department of General SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| | - Wen Xu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of PharmacyEast China University of Science and TechnologyShanghaiP.R. China
| | - Yongjun Chen
- Department of General SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| | - Xiaochun Fei
- Department of PathologyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| | - Junqing Wang
- Department of General SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP.R. China
| |
Collapse
|
21
|
Aslebagh R, Whitham D, Channaveerappa D, Mutsengi P, Pentecost BT, Arcaro KF, Darie CC. Mass Spectrometry-Based Proteomics of Human Milk to Identify Differentially Expressed Proteins in Women with Breast Cancer versus Controls. Proteomes 2022; 10:36. [PMID: 36412635 PMCID: PMC9680319 DOI: 10.3390/proteomes10040036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
It is thought that accurate risk assessment and early diagnosis of breast cancer (BC) can help reduce cancer-related mortality. Proteomics analysis of breast milk may provide biomarkers of risk and occult disease. Our group works on the analysis of human milk samples from women with BC and controls to investigate alterations in protein patterns of milk that could be related to BC. In the current study, we used mass spectrometry (MS)-based proteomics analysis of 12 milk samples from donors with BC and matched controls. Specifically, we used one-dimensional (1D)-polyacrylamide gel electrophoresis (PAGE) coupled with nanoliquid chromatography tandem MS (nanoLC-MS/MS), followed by bioinformatics analysis. We confirmed the dysregulation of several proteins identified previously in a different set of milk samples. We also identified additional dysregulations in milk proteins shown to play a role in cancer development, such as Lactadherin isoform A, O-linked N-acetylglucosamine (GlcNAc) transferase, galactosyltransferase, recoverin, perilipin-3 isoform 1, histone-lysine methyltransferase, or clathrin heavy chain. Our results expand our current understanding of using milk as a biological fluid for identification of BC-related dysregulated proteins. Overall, our results also indicate that milk has the potential to be used for BC biomarker discovery, early detection and risk assessment in young, reproductively active women.
Collapse
Affiliation(s)
- Roshanak Aslebagh
- Biochemistry and Proteomics Laboratories, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Danielle Whitham
- Biochemistry and Proteomics Laboratories, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Devika Channaveerappa
- Biochemistry and Proteomics Laboratories, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Panashe Mutsengi
- Biochemistry and Proteomics Laboratories, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Brian T. Pentecost
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA 01003-9298, USA
| | - Kathleen F. Arcaro
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA 01003-9298, USA
| | - Costel C. Darie
- Biochemistry and Proteomics Laboratories, Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| |
Collapse
|
22
|
Cui Z, Mo J, Song P, Wang L, Wang R, Cheng F, Wang L, Zou F, Guan X, Zheng N, Yang X, Wang W. Comprehensive bioinformatics analysis reveals the prognostic value, predictive value, and immunological roles of ANLN in human cancers. Front Genet 2022; 13:1000339. [PMID: 36199577 PMCID: PMC9527346 DOI: 10.3389/fgene.2022.1000339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022] Open
Abstract
Anillin (ANLN) is a unique scaffolding, actin-binding protein, which is essential for the integrity and ingression of the cleavage furrow. It is mainly involved in the cytokinesis process, while its role in various tumors has not been fully addressed and remains largely elusive. To provide a thorough perspective of ANLN’s roles among diverse malignancies, we conducted a comprehensive, pan-cancer analysis about ANLN, including but not limited to gene expression levels, prognostic value, biological functions, interacting proteins, immune-related analysis, and predictive value. As a result, when compared to normal tissues, ANLN expression is elevated in most cancers, and its expression also differs in different immune subtypes and molecular subtypes in diverse cancers. In addition, in 17 types of cancer, ANLN expression is increased in early tumor stages, and higher ANLN expression predicts worse survival outcomes in more than ten cancers. Furthermore, ANLN shows close correlations with the infiltration levels of most immune cells, and enrichment analysis using ANLN co-expressed genes reveals that ANLN plays essential roles in cell cycle, mitosis, cellular senescence, and p53 signaling pathways. In the final, ANLN exhibits high accuracy in predicting many cancers, and subsequent multivariate analysis suggests ANLN could be an independent prognostic factor in specific cancer types. Taken together, ANLN is proved to be a novel and promising biomarker for its excellent predictive utility, promising prognostic value, and potential immunological roles in pan-cancer. Targeting ANLN might be an attractive approach to tumor treatment.
Collapse
Affiliation(s)
- Zhiwei Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ping Song
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lijun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rongli Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Feiyan Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lihui Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fan Zou
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Nini Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xinyuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wei Wang, ; Xinyuan Yang,
| | - Wei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wei Wang, ; Xinyuan Yang,
| |
Collapse
|
23
|
Matsumoto T. Implications of Polyploidy and Ploidy Alterations in Hepatocytes in Liver Injuries and Cancers. Int J Mol Sci 2022; 23:ijms23169409. [PMID: 36012671 PMCID: PMC9409051 DOI: 10.3390/ijms23169409] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Polyploidy, a condition in which more than two sets of chromosomes are present in a cell, is a characteristic feature of hepatocytes. A significant number of hepatocytes physiologically undergo polyploidization at a young age. Polyploidization of hepatocytes is enhanced with age and in a diseased liver. It is worth noting that polyploid hepatocytes can proliferate, in marked contrast to other types of polyploid cells, such as megakaryocytes and cardiac myocytes. Polyploid hepatocytes divide to maintain normal liver homeostasis and play a role in the regeneration of the damaged liver. Furthermore, polyploid hepatocytes have been shown to dynamically reduce ploidy during liver regeneration. Although it is still unclear why hepatocytes undergo polyploidization, accumulating evidence has revealed that alterations in the ploidy in hepatocytes are involved in the pathophysiology of liver cirrhosis and carcinogenesis. This review discusses the significance of hepatocyte ploidy in physiological liver function, liver injury, and liver cancer.
Collapse
Affiliation(s)
- Tomonori Matsumoto
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
24
|
Sladky VC, Akbari H, Tapias-Gomez D, Evans LT, Drown CG, Strong MA, LoMastro GM, Larman T, Holland AJ. Centriole signaling restricts hepatocyte ploidy to maintain liver integrity. Genes Dev 2022; 36:gad.349727.122. [PMID: 35981754 PMCID: PMC9480857 DOI: 10.1101/gad.349727.122] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 01/03/2023]
Abstract
Hepatocyte polyploidization is a tightly controlled process that is initiated at weaning and increases with age. The proliferation of polyploid hepatocytes in vivo is restricted by the PIDDosome-P53 axis, but how this pathway is triggered remains unclear. Given that increased hepatocyte ploidy protects against malignant transformation, the evolutionary driver that sets the upper limit for hepatocyte ploidy remains unknown. Here we show that hepatocytes accumulate centrioles during cycles of polyploidization in vivo. The presence of excess mature centrioles containing ANKRD26 was required to activate the PIDDosome in polyploid cells. As a result, mice lacking centrioles in the liver or ANKRD26 exhibited increased hepatocyte ploidy. Under normal homeostatic conditions, this increase in liver ploidy did not impact organ function. However, in response to chronic liver injury, blocking centriole-mediated ploidy control leads to a massive increase in hepatocyte polyploidization, severe liver damage, and impaired liver function. These results show that hyperpolyploidization sensitizes the liver to injury, posing a trade-off for the cancer-protective effect of increased hepatocyte ploidy. Our results may have important implications for unscheduled polyploidization that frequently occurs in human patients with chronic liver disease.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Hanan Akbari
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Lauren T Evans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Chelsea G Drown
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Gina M LoMastro
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Tatianna Larman
- Divison of Gastrointestinal and Liver Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
25
|
Pan Y, Wei M, Gong T. Ultrasound microbubble-mediated delivery of ANLN silencing-repressed EZH2 expression alleviates cervical cancer progression. Tissue Cell 2022; 77:101843. [PMID: 35679682 DOI: 10.1016/j.tice.2022.101843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
Ultrasound-targeted microbubble destruction (UTMD) is a new gene therapy method that uses ultrasound and microbubbles carrying target genes to achieve gene transfection. However, whether UTMD-mediated ANLN silencing transfection helps to restrain the growth of cervical cancer (CC) is obscure. ANLN level in tumor tissues, adjacent tissues, and cells was tested using the database, qRT-PCR, and western blot. The optimal concentration of SF6 was determined by MTT assay. Mechanical index (MI) was selected by flow cytometry. After transfection with liposome or UTMD-mediated liposome, cell function experiments, qRT-PCR, and western blot were employed to assess CC cell biological behaviors and EZH2 level. Epithelial-mesenchymal transition (EMT)-related marker and apoptosis-related marker expressions were examined utilizing qRT-PCR and western blot. 10% SF6 and MI of 0.28 were selected for subsequent tests. ANLN was highly expressed in CC and cells. The transfection efficiency of the UTMD-siANLN group was higher than that of the L-siANLN group. Moreover, the repression of UTMD-siANLN on CC cell malignant phenotypes was stronger than L-siANLN. UTMD-siANLN attenuated EZH2 expression in CC cells. The modulatory role of UTMD-siANLN on EMT- and apoptosis-related markers was reversed by EZH2 overexpression. UTMD can improve the efficiency of siANLN transfection into CC cells to induce suppression of CC cell malignant phenotypes, which may become a new target of gene therapy for CC.
Collapse
Affiliation(s)
- Yanyan Pan
- Ultrasonic Diagnosis Center, Northwest Women's and Children's Hospital, Xi'an 710061, China
| | - Min Wei
- Department of Ultrasonic, Shanghai Rici Women's and Children's Hospital, Shanghai 200040, China
| | - Ting Gong
- Ultrasonic Diagnosis Center, Northwest Women's and Children's Hospital, Xi'an 710061, China.
| |
Collapse
|
26
|
Dong P, Cai Z, Li B, Zhu Y, Chan AKY, Chiang MWL, Au CH, Sung WK, Cheung TT, Lo CM, Man K, Lee NP. HFE promotes mitotic cell division through recruitment of cytokinetic abscission machinery in hepatocellular carcinoma. Oncogene 2022; 41:4185-4199. [PMID: 35882980 DOI: 10.1038/s41388-022-02419-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
HFE (Hemochromatosis) is a conventional iron level regulator and its loss of function due to gene mutations increases the risk of cancers including hepatocellular carcinoma (HCC). Likewise, studies focusing on HFE overexpression in cancers are all limited to linking up these events as a consequence of iron level deregulation. No study has explored any iron unrelated role of HFE in cancers. Here, we first reported HFE as an oncogene in HCC and its undescribed function on promoting abscission in cytokinesis during mitotic cell division, independent of its iron-regulating ability. Clinical analyses revealed HFE upregulation in tumors linking to large tumor size and poor prognosis. Functionally and mechanistically, HFE promoted cytokinetic abscission via facilitating ESCRT abscission machinery recruitment to the abscission site through signaling a novel HFE/ALK3/Smads/LIF/Hippo/YAP/YY1/KIF13A axis. Pharmacological blockage of HFE signaling axis impeded tumor phenotypes in vitro and in vivo. Our data on HFE-driven HCC unveiled a new mechanism utilized by cancer cells to propel rapid cell division. This study also laid the groundwork for tumor intolerable therapeutics development given the high cytokinetic dependency of cancer cells and their vulnerability to cytokinetic blockage.
Collapse
Affiliation(s)
- Pingping Dong
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Radiation Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziqing Cai
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Bingfeng Li
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Yueqin Zhu
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alice K Y Chan
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong.,Po Leung Kuk Tong Nai Kan Junior Secondary College, Hong Kong, Hong Kong
| | - Michael W L Chiang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Hang Au
- Hong Kong Genome Institute, Hong Kong, Hong Kong
| | - Wing Kin Sung
- Hong Kong Genome Institute, Hong Kong, Hong Kong.,School of Computing, National University of Singapore, Singapore, Singapore.,Computational and Systems Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong.
| | - Nikki P Lee
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong. .,Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong, Hong Kong.
| |
Collapse
|
27
|
Kim JY, Choi H, Kim HJ, Jee Y, Noh M, Lee MO. Polyploidization of Hepatocytes: Insights into the Pathogenesis of Liver Diseases. Biomol Ther (Seoul) 2022; 30:391-398. [PMID: 35790893 PMCID: PMC9424332 DOI: 10.4062/biomolther.2022.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 11/26/2022] Open
Abstract
Polyploidization is a process by which cells are induced to possess more than two sets of chromosomes. Although polyploidization is not frequent in mammals, it is closely associated with development and differentiation of specific tissues and organs. The liver is one of the mammalian organs that displays ploidy dynamics in physiological homeostasis during its development. The ratio of polyploid hepatocytes increases significantly in response to hepatic injury from aging, viral infection, iron overload, surgical resection, or metabolic overload, such as that from non-alcoholic fatty liver diseases (NAFLDs). One of the unique features of NAFLD is the marked heterogeneity of hepatocyte nuclear size, which is strongly associated with an adverse liver-related outcome, such as hepatocellular carcinoma, liver transplantation, and liver-related death. Thus, hepatic polyploidization has been suggested as a potential driver in the progression of NAFLDs that are involved in the control of the multiple pathogenicity of the diseases. However, the importance of polyploidy in diverse pathophysiological contexts remains elusive. Recently, several studies reported successful improvement of symptoms of NAFLDs by reducing pathological polyploidy or by controlling cell cycle progression in animal models, suggesting that better understanding the mechanisms of pathological hepatic polyploidy may provide insights into the treatment of hepatic disorders.
Collapse
Affiliation(s)
- Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
| | - Haena Choi
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 00826, Republic of Korea
| | - Yelin Jee
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 00826, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul 00826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 00826, Republic of Korea
- Bio-MAX institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
28
|
Zhang D, Wang G, Yu X, Wei T, Farbiak L, Johnson LT, Taylor AM, Xu J, Hong Y, Zhu H, Siegwart DJ. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. NATURE NANOTECHNOLOGY 2022; 17:777-787. [PMID: 35551240 PMCID: PMC9931497 DOI: 10.1038/s41565-022-01122-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 03/21/2022] [Indexed: 05/06/2023]
Abstract
Genome editing holds great potential for cancer treatment due to the ability to precisely inactivate or repair cancer-related genes. However, delivery of CRISPR/Cas to solid tumours for efficient cancer therapy remains challenging. Here we targeted tumour tissue mechanics via a multiplexed dendrimer lipid nanoparticle (LNP) approach involving co-delivery of focal adhesion kinase (FAK) siRNA, Cas9 mRNA and sgRNA (siFAK + CRISPR-LNPs) to enable tumour delivery and enhance gene-editing efficacy. We show that gene editing was enhanced >10-fold in tumour spheroids due to increased cellular uptake and tumour penetration of nanoparticles mediated by FAK-knockdown. siFAK + CRISPR-PD-L1-LNPs reduced extracellular matrix stiffness and efficiently disrupted PD-L1 expression by CRISPR/Cas gene editing, which significantly inhibited tumour growth and metastasis in four mouse models of cancer. Overall, we provide evidence that modulating the stiffness of tumour tissue can enhance gene editing in tumours, which offers a new strategy for synergistic LNPs and other nanoparticle systems to treat cancer using gene editing.
Collapse
Affiliation(s)
- Di Zhang
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guoxun Wang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xueliang Yu
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuo Wei
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lukas Farbiak
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lindsay T Johnson
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alan Mark Taylor
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Children's Research Institute Mouse Genome Engineering Core, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
29
|
Chen J, Li Z, Jia X, Song W, Wu H, Zhu H, Xuan Z, Du Y, Zhu X, Song G, Dong H, Bian S, Wang S, Zhao Y, Xie H, Zheng S, Song P. Targeting anillin inhibits tumorigenesis and tumor growth in hepatocellular carcinoma via impairing cytokinesis fidelity. Oncogene 2022; 41:3118-3130. [PMID: 35477750 DOI: 10.1038/s41388-022-02274-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Abstract
Targeting cytokinesis can suppress tumor growth by blocking cell division and promoting apoptosis. We aimed to characterize key cytokinesis regulator in hepatocellular carcinoma (HCC) progression, providing insights into identifying promising HCC therapeutic targets. The unbiased bioinformatic screening identified Anillin actin binding protein (ANLN) as a critical cytokinesis regulator involved in HCC development. Functional assay demonstrated that knockdown of ANLN inhibited HCC growth by inducing cytokinesis failure and DNA damage, leading to multinucleation and mitotic catastrophe. Mechanistically, ANLN acts as a scaffold to strengthen interaction between RACGAP1 and PLK1. ANLN promotes PLK1-mediated RACGAP1 phosphorylation and RhoA activation to ensure cytokinesis fidelity. To explore the function of ANLN in HCC tumorigenesis, we hydrodynamically transfected c-Myc and NRAS plasmids into Anln+/+, Anln+/-, and Anln-/- mice through tail vein injection. Hepatic Anln ablation significantly impaired c-Myc/NRAS-driven hepatocarcinogenesis. Moreover, enhanced hepatic polyploidization was observed in Anln ablation mice, manifesting as increasing proportion of cellular and nuclear polyploidy. Clinically, ANLN is upregulated in human HCC tissues and high level of ANLN is correlated with poor patients' prognosis. Additionally, the proportion of cellular polyploidy decreases during HCC progression and ANLN level is significantly correlated with cellular polyploidy proportion in human HCC samples. In conclusion, ANLN is identified as a key cytokinesis regulator contributing to HCC initiation and progression. Our findings revealed a novel mechanism of ANLN in the regulation of cytokinesis to promote HCC tumorigenesis and growth, suggesting targeting ANLN to inhibit cytokinesis may be a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Xing Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hai Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zefeng Xuan
- Division of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yehui Du
- Division of Thyroid Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingxin Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Guangyuan Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Haijiang Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Suchen Bian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shuo Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
30
|
Yang K, Shen Z, Yin N, Quan J, Wang M, Gao K. Development and Validation of a Novel Hypoxia Score for Predicting Prognosis and Immune Microenvironment in Rectal Cancer. Front Surg 2022; 9:881554. [PMID: 35548187 PMCID: PMC9081503 DOI: 10.3389/fsurg.2022.881554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/21/2022] [Indexed: 12/04/2022] Open
Abstract
Hypoxia plays a major role in various tumor types. However, few studies have concentrated on the prognostic model of hypoxia-related genes in rectal cancer and the effect of hypoxia on neutrophil-mediated immunosuppression. We performed Kaplan–Meier analysis, random survival forest analysis, and Cox regression analysis on 342 hypoxia-related genes, constructed hypoxia score in the Gene Expression Omnibus (GEO) cohort, and verified them in the Cancer Genome Atlas (TCGA) cohort. Then the patients were divided into two groups according to the risk level. The overall survival rate of the high-risk (HRisk) group was significantly higher than that of the low-risk (LRisk) group (GEO, p < 0.001; TCGA, p = 0.016). Through receiver operating characteristic and decision curve analysis, the nomogram based on hypoxia score has excellent prediction ability. Functional enrichment analysis showed that hypoxia, metastasis, inflammation, immunity, and other related pathways were enriched. The HRisk group was associated with lower tumor purity, higher immune and stromal score, higher neutrophils, and lower activated memory CD4 + T cells. More importantly, the checkpoint of neutrophil-mediated immunosuppression increased in the HRisk group. In conclusion, a hypoxia score based on 5 hypoxia-related genes can be used to predict the prognosis of rectal cancer and ANLN with a cancer-suppressing effect and SRPX (Sushi Repeat Containing Protein X-Linked) with a cancer-promoting effect may be potential therapeutic targets for rectal cancer.
Collapse
|
31
|
In vivo CRISPR screening identifies BAZ2 chromatin remodelers as druggable regulators of mammalian liver regeneration. Cell Stem Cell 2022; 29:372-385.e8. [PMID: 35090595 PMCID: PMC8897233 DOI: 10.1016/j.stem.2022.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 09/17/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022]
Abstract
Identifying new pathways that regulate mammalian regeneration is challenging due to the paucity of in vivo screening approaches. We employed pooled CRISPR knockout and activation screening in the regenerating liver to evaluate 165 chromatin regulatory proteins. Both screens identified the imitation-SWI chromatin remodeling components Baz2a and Baz2b, not previously implicated in regeneration. In vivo sgRNA, siRNA, and knockout strategies against either paralog confirmed increased regeneration. Distinct BAZ2-specific bromodomain inhibitors, GSK2801 and BAZ2-ICR, resulted in accelerated liver healing after diverse injuries. Inhibitor-treated mice also exhibited improved healing in an inflammatory bowel disease model, suggesting multi-tissue applicability. Transcriptomics on regenerating livers showed increases in ribosomal and cell cycle mRNAs. Surprisingly, CRISPRa screening to define mechanisms showed that overproducing Rpl10a or Rpl24 was sufficient to drive regeneration, whereas Rpl24 haploinsufficiency was rate limiting for BAZ2 inhibition-mediated regeneration. The discovery of regenerative roles for imitation-SWI components provides immediate strategies to enhance tissue repair.
Collapse
|
32
|
Zhang L, Yang Z, Zhang S, Zhou K, Zhang W, Ling S, Sun R, Tang H, Wen X, Feng X, Song P, Xu X, Xie H, Zheng S. Polyploidy Spectrum Correlates with Immunophenotype and Shapes Hepatocellular Carcinoma Recurrence Following Liver Transplantation. J Inflamm Res 2022; 15:217-233. [PMID: 35046696 PMCID: PMC8760994 DOI: 10.2147/jir.s345681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Liang Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Zhentao Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Shiyu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Ke Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Wu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang, 310004, People’s Republic of China
| | - Sunbin Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Ruiqi Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Hong Tang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Xue Wen
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Xiaowen Feng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
- Correspondence: Haiyang Xie; Shusen Zheng School of Medicine, Zhejiang University, 79# Qingchun Road, Hangzhou, Zhejiang, 310000, People’s Republic of ChinaTel/Fax +86 571 87236570; +86 571 87236466 Email ;
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, 310003, People’s Republic of China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang, 310004, People’s Republic of China
| |
Collapse
|
33
|
Wang N, Hao F, Shi Y, Wang J. The Controversial Role of Polyploidy in Hepatocellular Carcinoma. Onco Targets Ther 2021; 14:5335-5344. [PMID: 34866913 PMCID: PMC8636953 DOI: 10.2147/ott.s340435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022] Open
Abstract
Polyploidy, a physiological phenomenon in which cells contain more than two sets of homologous chromosomes, commonly exists in plants, fish, and amphibians but is rare in mammals. In humans, polyploid cells are detected commonly in specific organs or tissues including the heart, marrow, and liver. As the largest solid organ in the body, the liver is responsible for a myriad of functions, most of which are closely related to polyploid hepatocytes. It has been confirmed that polyploid hepatocytes are related to liver regeneration, homeostasis, terminal differentiation, and aging. Polyploid hepatocytes accumulate during the aging process as well as in chronically injured livers. The relationship between polyploid hepatocytes and hepatocellular carcinoma, the endpoint of most chronic liver diseases, is not yet fully understood. Recently, accumulated evidence has revealed that polyploid involves in the process of tumorigenesis and development. The study of the correlation and relationship between polyploidy hepatocytes and the development of hepatocellular carcinoma can potentially promote the prevention, early diagnosis, and treatment of hepatocellular carcinoma. In this review, we conclude the potential mechanisms of polyploid hepatocytes formation, focusing on the specific biological significance of polyploid hepatocytes. In addition, we examine recent discoveries that have begun to clarify the relevance between polyploid hepatocytes and hepatocellular carcinoma and discuss recent excellent findings that reveal the role of polyploid hepatocytes as resisters of hepatocellular carcinoma or as promoters of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Nan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
34
|
Sladky VC, Eichin F, Reiberger T, Villunger A. Polyploidy control in hepatic health and disease. J Hepatol 2021; 75:1177-1191. [PMID: 34228992 DOI: 10.1016/j.jhep.2021.06.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/25/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022]
Abstract
A balanced increase in DNA content (ploidy) is observed in some human cell types, including bone-resorbing osteoclasts, platelet-producing megakaryocytes, cardiomyocytes or hepatocytes. The impact of increased hepatocyte ploidy on normal physiology and diverse liver pathologies is still poorly understood. Recent findings suggest swift genetic adaptation to hepatotoxic stress and the protection from malignant transformation as beneficial effects. Herein, we discuss the molecular mechanisms regulating hepatocyte polyploidisation and its implication for different liver diseases and hepatocellular carcinoma. We report on centrosomes' role in limiting polyploidy by activating the p53 signalling network (via the PIDDosome multiprotein complex) and we discuss the role of this pathway in liver disease. Increased hepatocyte ploidy is a hallmark of hepatic inflammation and may play a protective role against liver cancer. Our evolving understanding of hepatocyte ploidy is discussed from the perspective of its potential clinical application for risk stratification, prognosis, and novel therapeutic strategies in liver disease and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Felix Eichin
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria.
| |
Collapse
|
35
|
Li Y, Wu D, Wei C, Yang X, Zhou S. [CDK1, CCNB1 and NDC80 are associated with prognosis and progression of hepatitis B virus-associated hepatocellular carcinoma: a bioinformatic analysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1509-1518. [PMID: 34755666 DOI: 10.12122/j.issn.1673-4254.2021.10.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To identify the key genes involved in the transformation of hepatitis B virus (HBV) into hepatocellular carcinoma (HCC) and explore the underlying molecular mechanisms. METHODS We analyzed the mRNA microarray data of 119 HBV-related HCC tissues and 252 HBV-related non-tumor tissues in GSE55092, GSE84044 and GSE121248 from the GEO database, and the "sva" R package was used to remove the batch effects. Integration analysis was performed to identify the differentially expressed genes (DEGs) in HBV-related liver cancer and liver tissues with HBV infection. The significant DEGs were functionally annotated using GO and KEGG analyses, and the most important modules and hub genes were explored with STRING analysis. Kaplan-Meier and Oncomine databases were used to verify the HCC gene expression data in the TCGA database to explore the correlations of the hub genes with the occurrence, progression and prognosis of HCC. We also examined the expressions of the hub genes in 17 pairs of surgical specimens of HCC and adjacent tissues using RT-qPCR. RESULTS We identified a total of 121 DEGs and 3 genetic markers in HCC (P < 0.01). These DEGs included cyclin1 (CDK1), cyclin B1 (CCNB1), and nuclear division cycle 80 (NDC80), which participated in cell cycle, pyrimidine metabolism and DNA replication and were highly correlated (P < 0.05). Analysis of the UALCAN database confirmed high expressions of these 3 genes in HCC tissues, which were correlated with a low survival rate of the patients, as shown by Kaplan-Meier analysis of the prognostic data from the UALCAN database. CDK1, CCNB1 and NDC80 were all correlated with the clinical grading of HCC (P < 0.05). The results of RT-qPCR on the surgical specimens verified significantly higher expressions of CDK1, CCNB1 and NDC80 mRNA in HCC tissues than in the adjacent tissues. CONCLUSION CDK1, CCNB1 and NDC80 genes can be used as prognostic markers of HBV-related HCC and may serve as potential targets in preclinical studies and clinical treatment of HCC.
Collapse
Affiliation(s)
- Y Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Nanning 530021, China.,The Key Laboratory of Longevity and Geriatric-related Diseases of the Ministry of Education, Nanning 530021, China
| | - D Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Nanning 530021, China.,The Key Laboratory of Biomolecular Medicine Research in Guangxi Universities, Nanning 530021, China
| | - C Wei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Nanning 530021, China.,The Key Laboratory of Biomolecular Medicine Research in Guangxi Universities, Nanning 530021, China
| | - X Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Nanning 530021, China.,The Key Laboratory of Biomolecular Medicine Research in Guangxi Universities, Nanning 530021, China
| | - S Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Nanning 530021, China.,The Key Laboratory of the Ministry of Education for Early Prevention and Treatment of Regional High-incidence Tumors, Nanning 530021, China
| |
Collapse
|
36
|
Donne R, Sangouard F, Celton-Morizur S, Desdouets C. Hepatocyte Polyploidy: Driver or Gatekeeper of Chronic Liver Diseases. Cancers (Basel) 2021; 13:cancers13205151. [PMID: 34680300 PMCID: PMC8534039 DOI: 10.3390/cancers13205151] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 12/25/2022] Open
Abstract
Polyploidy, also known as whole-genome amplification, is a condition in which the organism has more than two basic sets of chromosomes. Polyploidy frequently arises during tissue development and repair, and in age-associated diseases, such as cancer. Its consequences are diverse and clearly different between systems. The liver is a particularly fascinating organ in that it can adapt its ploidy to the physiological and pathological context. Polyploid hepatocytes are characterized in terms of the number of nuclei per cell (cellular ploidy; mononucleate/binucleate hepatocytes) and the number of chromosome sets in each nucleus (nuclear ploidy; diploid, tetraploid, octoploid). The advantages and disadvantages of polyploidy in mammals are not fully understood. About 30% of the hepatocytes in the human liver are polyploid. In this review, we explore the mechanisms underlying the development of polyploid cells, our current understanding of the regulation of polyploidization during development and pathophysiology and its consequences for liver function. We will also provide data shedding light on the ways in which polyploid hepatocytes cope with centrosome amplification. Finally, we discuss recent discoveries highlighting the possible roles of liver polyploidy in protecting against tumor formation, or, conversely, contributing to liver tumorigenesis.
Collapse
Affiliation(s)
- Romain Donne
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
- Icahn School of Medicine at Mount Sinai, The Precision Immunology Institute, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Flora Sangouard
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, F-75006 Paris, France;
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Séverine Celton-Morizur
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, F-75006 Paris, France;
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
- Correspondence: (S.C.-M.); (C.D.)
| | - Chantal Desdouets
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, F-75006 Paris, France;
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
- Correspondence: (S.C.-M.); (C.D.)
| |
Collapse
|
37
|
Xu W, Lu X, Liu J, Chen Q, Huang X, Huang K, Liu H, Zhu W, Zhang X. Identification of PAFAH1B3 as Candidate Prognosis Marker and Potential Therapeutic Target for Hepatocellular Carcinoma. Front Oncol 2021; 11:700700. [PMID: 34490100 PMCID: PMC8418329 DOI: 10.3389/fonc.2021.700700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related deaths worldwide. PAFAH1B3 plays an important role on occurrence and development in a variety tumor. However, the function of PAFAH1B3 in HCC remains unclear. Methods The TIMER, ONCOMINE, Human Protein Atlas (HPA), GEPIA, The Cancer Genome Atlas (TCGA), HCCDB, UALCAN and LinkedOmics database were used to analyze the prognostic value, co-expression genes and regulator networks of PAFAH1B3 in HCC. siRNA transfections and inhibitor of PAFAH1B3 P11 were used to verify the anti-tumor effect on HCC cell lines. Gene expression was detected by qRT-PCR. The functions of PAFAH1B3 downregulation in HCC cell lines were investigated using cell cycle analysis, apoptosis detection, CCK8 assay and transwell assay. Western blot was used to evaluate the role of PAFAH1B3 on metabolic pathways in HCC cells. Results Based on the data from databases, the expression of PAFAH1B3 was remarkably increased in HCC patients. High expression of PAFAH1B3 was associated with poorer overall survival (OS) and disease-free survival (DFS). And PAFAH1B3 was notably linked to age, sex, grade, stage, race, and TP53 mutational status. Then, the functional network analysis showed PAFAH1B3 may be involved in HCC through cell cycle, cell metabolism, spliceosome, and RNA transport. Furthermore, the mRNA expression of PAFAH1B3 was also increased in HCC cell lines. Flow cytometry analysis showed that PAFAH1B3 manipulated apoptosis and cell cycle regulation. CCK8 assay showed that PAFAH1B3 silencing or pharmacologic inhibitor of PAFAH1B3 inhibited the proliferation of HepG2, Huh7 and MHCC-97H cells. Transwell assay results showed that PAFAH1B3 silencing also significantly impaired the invasion and migratory ability of HCC cells. In addition, PAFAH1B3 silencing significantly downregulated the expression of glycolysis and lipid synthesis signaling pathways. Conclusion Our findings suggested that PAFAH1B3 plays a critical role in progression of HCC. PAFAH1B3 as a prognosis marker and potential target for HCC has prospective clinical significance.
Collapse
Affiliation(s)
- Weikang Xu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Liu
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kuiyuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongyan Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Alternatively spliced ANLN isoforms synergistically contribute to the progression of head and neck squamous cell carcinoma. Cell Death Dis 2021; 12:764. [PMID: 34344861 PMCID: PMC8333361 DOI: 10.1038/s41419-021-04063-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common cancer with high mortality. Anilin actin-binding protein (ANLN) has been reported to be associated with carcinogenesis in multiple tumors. However, the expression pattern and functional effects of ANLN in HNSCC remain to be unclear. Clinical data and online databases were used to analyze the expression of ANLN and its relationship with HNSCC patient survival. Expression of two major splice variants of ANLN was assessed in HNSCC tissues and cell lines. The functional effects and related mechanisms of ANLN isoforms were investigated in HNSCC in vitro and in vivo. Our study showed that patients with high expression of ANLN had a poor prognosis. The two primary isoforms of ANLN transcripts ANLN-201 and ANLN-210 were highly expressed in HNSCC tissues and cell lines. Knockout of ANLN restrained cell proliferation, migration, and invasion of SCC-9 cells. Mechanically, ANLN-201 could interact with c-Myc to keep its protein stability, thereby playing a oncogenic role in HNSCC. ANLN-210 could be transferred to macrophages via exosomes by binding to RNA-binding protein hnRNPC. Exosomal ANLN-210 promoted macrophage polarization via PTEN/PI3K/Akt signaling pathway, thus stimulating tumor growth of HNSCC. ANLN was an independent prognostic factor in patients with HNSCC. Alternatively spliced ANLN isoforms collaboratively promote HNSCC tumorigenesis in vitro and in vivo, which might provide the in-depth role and mechanism of ANLN in HNSCC development.
Collapse
|
39
|
Zhang LH, Wang D, Li Z, Wang G, Chen DB, Cheng Q, Hu SH, Zhu JY. Overexpression of anillin is related to poor prognosis in patients with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2021; 20:337-344. [PMID: 32933876 DOI: 10.1016/j.hbpd.2020.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/18/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Anillin (ANLN) is required for tumor growth. It has been proven that knockdown of ANLN effectively reduces the occurrence of hepatocellular carcinoma (HCC) in transgenic mice. However, the functional role of ANLN in HCC patients remains to be elucidated. METHODS Both microarray and TCGA project were used for the analyses of ANLN expression and regulation in HCC. The effect of ANLN on proliferation and cell cycle was detected by CCK-8, colony formation assay and flow cytometry. ANLN expression was measured by immunohistochemistry. Correlation between ANLN expression and clinicopathological features was assessed by Pearson Chi-square test and 5-year overall survival after liver resection was evaluated by Kaplan-Meier method. RESULTS Increased copy number, decreased methylation levels in the CpG island and upregulated histone hypermethylation of ANLN were found in HCC. Knockdown of ANLN inhibited proliferation and induced G2/M phase arrest in SMMC-7721 cells. ANLN was mainly expressed in the nucleus and showed significantly higher expression levels in cancerous tissues than those in paired adjacent tissues. Moreover, nuclear ANLN expression levels in HCC metastases were significantly higher than those in primary HCC. The results of Cox proportional hazards regression model suggested that ANLN nuclear expression in HCC was an independent risk factor for poor 5-year overall survival of patients after liver resection. CONCLUSIONS ANLN is a potential therapeutic target for HCC. Patients with nuclear ANLN overexpression in HCC tissue may need adjuvant therapy after liver resection.
Collapse
Affiliation(s)
- Long-Hui Zhang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Dong Wang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Gang Wang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Ding-Bao Chen
- Department of Pathology, Peking University People's Hospital, Beijing 100044, China
| | - Qian Cheng
- Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Shi-Hua Hu
- Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Ji-Ye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University Institute for Organ Transplantation, Beijing 100044, China; Beijing Key Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China.
| |
Collapse
|
40
|
Nawroth JC, Petropolis DB, Manatakis DV, Maulana TI, Burchett G, Schlünder K, Witt A, Shukla A, Kodella K, Ronxhi J, Kulkarni G, Hamilton G, Seki E, Lu S, Karalis KC. Modeling alcohol-associated liver disease in a human Liver-Chip. Cell Rep 2021; 36:109393. [PMID: 34289365 PMCID: PMC8342038 DOI: 10.1016/j.celrep.2021.109393] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/03/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol-associated liver disease (ALD) is a global health issue and leads to progressive liver injury, comorbidities, and increased mortality. Human-relevant preclinical models of ALD are urgently needed. Here, we leverage a triculture human Liver-Chip with biomimetic hepatic sinusoids and bile canaliculi to model ALD employing human-relevant blood alcohol concentrations (BACs) and multimodal profiling of clinically relevant endpoints. Our Liver-Chip recapitulates established ALD markers in response to 48 h of exposure to ethanol, including lipid accumulation and oxidative stress, in a concentration-dependent manner and supports the study of secondary insults, such as high blood endotoxin levels. We show that remodeling of the bile canalicular network can provide an in vitro quantitative readout of alcoholic liver toxicity. In summary, we report the development of a human ALD Liver-Chip as a powerful platform for modeling alcohol-induced liver injury with the potential for direct translation to clinical research and evaluation of patient-specific responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anke Witt
- Emulate, Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | | | | | - Janey Ronxhi
- Emulate, Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | | | | | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shelly Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | |
Collapse
|
41
|
Richter ML, Deligiannis IK, Yin K, Danese A, Lleshi E, Coupland P, Vallejos CA, Matchett KP, Henderson NC, Colome-Tatche M, Martinez-Jimenez CP. Single-nucleus RNA-seq2 reveals functional crosstalk between liver zonation and ploidy. Nat Commun 2021; 12:4264. [PMID: 34253736 PMCID: PMC8275628 DOI: 10.1038/s41467-021-24543-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 06/24/2021] [Indexed: 12/19/2022] Open
Abstract
Single-cell RNA-seq reveals the role of pathogenic cell populations in development and progression of chronic diseases. In order to expand our knowledge on cellular heterogeneity, we have developed a single-nucleus RNA-seq2 method tailored for the comprehensive analysis of the nuclear transcriptome from frozen tissues, allowing the dissection of all cell types present in the liver, regardless of cell size or cellular fragility. We use this approach to characterize the transcriptional profile of individual hepatocytes with different levels of ploidy, and have discovered that ploidy states are associated with different metabolic potential, and gene expression in tetraploid mononucleated hepatocytes is conditioned by their position within the hepatic lobule. Our work reveals a remarkable crosstalk between gene dosage and spatial distribution of hepatocytes.
Collapse
Affiliation(s)
- M L Richter
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
| | - I K Deligiannis
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
| | - K Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, United Kingdom
| | - A Danese
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - E Lleshi
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, United Kingdom
| | - P Coupland
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, United Kingdom
| | - C A Vallejos
- MRC Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - K P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Little France Crescent, Edinburgh, United Kingdom
| | - N C Henderson
- MRC Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Little France Crescent, Edinburgh, United Kingdom
| | - M Colome-Tatche
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Biomedical Center (BMC), Physiological Chemistry, Faculty of Medicine, LMU Munich, Munich, Germany.
| | - C P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany.
- TUM School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
42
|
Farbiak L, Cheng Q, Wei T, Álvarez-Benedicto E, Johnson LT, Lee S, Siegwart DJ. All-In-One Dendrimer-Based Lipid Nanoparticles Enable Precise HDR-Mediated Gene Editing In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006619. [PMID: 34137093 PMCID: PMC10041668 DOI: 10.1002/adma.202006619] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/19/2021] [Indexed: 05/08/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) protein gene editing is poised to transform the treatment of genetic diseases. However, limited progress has been made toward precise editing of DNA via homology-directed repair (HDR) that requires careful orchestration of complex steps. Herein, dendrimer-based lipid nanoparticles (dLNPs) are engineered to co-encapsulate and deliver multiple components for in vivo HDR correction. BFP/GFP switchable HEK293 cells with a single Y66H amino acid mutation are employed to assess HDR-mediated gene editing following simultaneous, one-pot delivery of Cas9 mRNA, single-guide RNA, and donor DNA. Molar ratios of individual LNP components and weight ratios of the three nucleic acids are systematically optimized to increase HDR efficiency. Using flow cytometry, fluorescence imaging, and DNA sequencing to quantify editing, optimized 4A3-SC8 dLNPs edit >91% of all cells with 56% HDR efficiency in vitro and >20% HDR efficiency in xenograft tumors in vivo. Due to the all-in-one simplicity and high efficacy, the developed dLNPs offer a promising route toward the gene correction of disease-causing mutations.
Collapse
Affiliation(s)
- Lukas Farbiak
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qiang Cheng
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tuo Wei
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ester Álvarez-Benedicto
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lindsay T Johnson
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sang Lee
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
43
|
Actin-Binding Proteins as Potential Biomarkers for Chronic Inflammation-Induced Cancer Diagnosis and Therapy. ACTA ACUST UNITED AC 2021; 2021:6692811. [PMID: 34194957 PMCID: PMC8203385 DOI: 10.1155/2021/6692811] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/13/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022]
Abstract
Actin-binding proteins (ABPs), by interacting with actin, regulate the polymerization, depolymerization, bundling, and cross-linking of actin filaments, directly or indirectly, thereby mediating the maintenance of cell morphology, cell movement, and many other biological functions. Consequently, these functions of ABPs help regulate cancer cell invasion and metastasis when cancer occurs. In recent years, a variety of ABPs have been found to be abnormally expressed in various cancers, indicating that the detection and interventions of unusual ABP expression to alter this are available for the treatment of cancer. The early stages of most cancer development involve long-term chronic inflammation or repeated stimulation. This is the case for breast cancer, gastric cancer, lung cancer, prostate cancer, liver cancer, esophageal cancer, pancreatic cancer, melanoma, and colorectal cancer. This article discusses the relationship between chronic inflammation and the above-mentioned cancers, emphatically introduces relevant research on the abnormal expression of ABPs in chronic inflammatory diseases, and reviews research on the expression of different ABPs in the above-mentioned cancers. Furthermore, there is a close relationship between ABP-induced inflammation and cancer. In simple terms, abnormal expression of ABPs contributes to the chronic inflammation developing into cancer. Finally, we provide our viewpoint regarding these unusual ABPs serving as potential biomarkers for chronic inflammation-induced cancer diagnosis and therapy, and interventions to reverse the abnormal expression of ABPs represent a potential approach to preventing or treating the corresponding cancers.
Collapse
|
44
|
Hachim MY, Elemam NM, Ramakrishnan RK, Salameh L, Olivenstein R, Hachim IY, Venkatachalam T, Mahboub B, Al Heialy S, Hamid Q, Hamoudi R. Derangement of cell cycle markers in peripheral blood mononuclear cells of asthmatic patients as a reliable biomarker for asthma control. Sci Rep 2021; 11:11873. [PMID: 34088958 PMCID: PMC8178351 DOI: 10.1038/s41598-021-91087-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
In asthma, most of the identified biomarkers pertain to the Th2 phenotype and no known biomarkers have been verified for severe asthmatics. Therefore, identifying biomarkers using the integrative phenotype-genotype approach in severe asthma is needed. The study aims to identify novel biomarkers as genes or pathways representing the core drivers in asthma development, progression to the severe form, resistance to therapy, and tissue remodeling regardless of the sample cells or tissues examined. Comprehensive reanalysis of publicly available transcriptomic data that later was validated in vitro, and locally recruited patients were used to decipher the molecular basis of asthma. Our in-silicoanalysis revealed a total of 10 genes (GPRC5A, SFN, ABCA1, KRT8, TOP2A, SERPINE1, ANLN, MKI67, NEK2, and RRM2) related to cell cycle and proliferation to be deranged in the severe asthmatic bronchial epithelium and fibroblasts compared to their healthy counterparts. In vitro, RT qPCR results showed that (SERPINE1 and RRM2) were upregulated in severe asthmatic bronchial epithelium and fibroblasts, (SFN, ABCA1, TOP2A, SERPINE1, MKI67, and NEK2) were upregulated in asthmatic bronchial epithelium while (GPRC5A and KRT8) were upregulated only in asthmatic bronchial fibroblasts. Furthermore, MKI76, RRM2, and TOP2A were upregulated in Th2 high epithelium while GPRC5A, SFN, ABCA1 were upregulated in the blood of asthmatic patients. SFN, ABCA1 were higher, while MKI67 was lower in severe asthmatic with wheeze compared to nonasthmatics with wheezes. SERPINE1 and GPRC5A were downregulated in the blood of eosinophilic asthmatics, while RRM2 was upregulated in an acute attack of asthma. Validation of the gene expression in PBMC of locally recruited asthma patients showed that SERPINE1, GPRC5A, SFN, ABCA1, MKI67, and RRM2 were downregulated in severe uncontrolled asthma. We have identified a set of biologically crucial genes to the homeostasis of the lung and in asthma development and progression. This study can help us further understand the complex interplay between the transcriptomic data and the external factors which may deviate our understanding of asthma heterogeneity.
Collapse
Affiliation(s)
- Mahmood Yaseen Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
- Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
| | - Noha Mousaad Elemam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rakhee K Ramakrishnan
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Laila Salameh
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Ibrahim Yaseen Hachim
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Thenmozhi Venkatachalam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bassam Mahboub
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Saba Al Heialy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, UCL, London, UK
| |
Collapse
|
45
|
Ithal D, Sukumaran SK, Bhattacharjee D, Vemula A, Nadella R, Mahadevan J, Sud R, Viswanath B, Purushottam M, Jain S. Exome hits demystified: The next frontier. Asian J Psychiatr 2021; 59:102640. [PMID: 33892377 DOI: 10.1016/j.ajp.2021.102640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Severe mental illnesses such as schizophrenia and bipolar disorder have complex inheritance patterns, involving both common and rare variants. Whole exome sequencing is a promising approach to find out the rare genetic variants. We had previously reported several rare variants in multiplex families with severe mental illnesses. The current article tries to summarise the biological processes and pattern of expression of genes harbouring the aforementioned variants, linking them to known clinical manifestations through a methodical narrative review. Of the 28 genes considered for this review from 7 families with multiple affected individuals, 6 genes are implicated in various neuropsychiatric manifestations including some variations in the brain morphology assessed by magnetic resonance imaging. Another 15 genes, though associated with neuropsychiatric manifestations, did not have established brain morphological changes whereas the remaining 7 genes did not have any previously recorded neuropsychiatric manifestations at all. Wnt/b-catenin signaling pathway was associated with 6 of these genes and PI3K/AKT, calcium signaling, ERK, RhoA and notch signaling pathways had at least 2 gene associations. We present a comprehensive review of biological and clinical knowledge about the genes previously reported in multiplex families with severe mental illness. A 'disease in dish approach' can be helpful to further explore the fundamental mechanisms.
Collapse
Affiliation(s)
- Dhruva Ithal
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Salil K Sukumaran
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Debanjan Bhattacharjee
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Alekhya Vemula
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Ravi Nadella
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Jayant Mahadevan
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Reeteka Sud
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Biju Viswanath
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Meera Purushottam
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India.
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| |
Collapse
|
46
|
Jiang N, Zhang X, Qin D, Yang J, Wu A, Wang L, Sun Y, Li H, Shen X, Lin J, Kantawong F, Wu J. Identification of Core Genes Related to Progression and Prognosis of Hepatocellular Carcinoma and Small-Molecule Drug Predication. Front Genet 2021; 12:608017. [PMID: 33708237 PMCID: PMC7940693 DOI: 10.3389/fgene.2021.608017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most leading causes of cancer death with a poor prognosis. However, the underlying molecular mechanisms are largely unclear, and effective treatment for it is limited. Using an integrated bioinformatics method, the present study aimed to identify the key candidate prognostic genes that are involved in HCC development and identify small-molecule drugs with treatment potential. Methods and Results In this study, by using three expression profile datasets from Gene Expression Omnibus database, 1,704 differentially expressed genes were identified, including 671 upregulated and 1,033 downregulated genes. Then, weighted co-expression network analysis revealed nine modules are related with pathological stage; turquoise module was the most associated module. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway analyses (KEGG) indicated that these genes were enriched in cell division, cell cycle, and metabolic related pathways. Furthermore, by analyzing the turquoise module, 22 genes were identified as hub genes. Based on HCC data from gene expression profiling interactive analysis (GEPIA) database, nine genes associated with progression and prognosis of HCC were screened, including ANLN, BIRC5, BUB1B, CDC20, CDCA5, CDK1, NCAPG, NEK2, and TOP2A. According to the Human Protein Atlas and the Oncomine database, these genes were highly upregulated in HCC tumor samples. Moreover, multivariate Cox regression analysis showed that the risk score based on the gene expression signature of these nine genes was an independent prognostic factor for overall survival and disease-free survival in HCC patients. In addition, the candidate small-molecule drugs for HCC were identified by the CMap database. Conclusion In conclusion, the nine key gene signatures related to HCC progression and prognosis were identified and validated. The cell cycle pathway was the core pathway enriched with these key genes. Moreover, several candidate molecule drugs were identified, providing insights into novel therapeutic approaches for HCC.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,School of Pharmacy, Southwest Medical University, Luzhou, China.,International Education School, Southwest Medical University, Luzhou, China
| | - Xinzhuo Zhang
- International Education School, Southwest Medical University, Luzhou, China
| | - Dalian Qin
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Jing Yang
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Long Wang
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Yueshan Sun
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Hong Li
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Xin Shen
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Jing Lin
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
47
|
Naydenov NG, Koblinski JE, Ivanov AI. Anillin is an emerging regulator of tumorigenesis, acting as a cortical cytoskeletal scaffold and a nuclear modulator of cancer cell differentiation. Cell Mol Life Sci 2021; 78:621-633. [PMID: 32880660 PMCID: PMC11072349 DOI: 10.1007/s00018-020-03605-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/29/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022]
Abstract
Remodeling of the intracellular cytoskeleton plays a key role in accelerating tumor growth and metastasis. Targeting different cytoskeletal elements is important for existing and future anticancer therapies. Anillin is a unique scaffolding protein that interacts with major cytoskeletal structures, e.g., actin filaments, microtubules and septin polymers. A well-studied function of this scaffolding protein is the regulation of cytokinesis at the completion of cell division. Emerging evidence suggest that anillin has other important activities in non-dividing cells, including control of intercellular adhesions and cell motility. Anillin is markedly overexpressed in different solid cancers and its high expression is commonly associated with poor prognosis of patient survival. This review article summarizes rapidly accumulating evidence that implicates anillin in the regulation of tumor growth and metastasis. We focus on molecular and cellular mechanisms of anillin-dependent tumorigenesis that include both canonical control of cytokinesis and novel poorly understood functions as a nuclear regulator of the transcriptional reprogramming and phenotypic plasticity of cancer cells.
Collapse
Affiliation(s)
- Nayden G Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, 9500 Euclid Avenue, NC22, Cleveland, OH, 44195, USA
| | - Jennifer E Koblinski
- Department of Pathology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, 9500 Euclid Avenue, NC22, Cleveland, OH, 44195, USA.
| |
Collapse
|
48
|
Sladky VC, Knapp K, Szabo TG, Braun VZ, Bongiovanni L, van den Bos H, Spierings DCJ, Westendorp B, Curinha A, Stojakovic T, Scharnagl H, Timelthaler G, Tsuchia K, Pinter M, Semmler G, Foijer F, de Bruin A, Reiberger T, Rohr‐Udilova N, Villunger A. PIDDosome-induced p53-dependent ploidy restriction facilitates hepatocarcinogenesis. EMBO Rep 2020; 21:e50893. [PMID: 33225610 PMCID: PMC7726793 DOI: 10.15252/embr.202050893] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Polyploidization frequently precedes tumorigenesis but also occurs during normal development in several tissues. Hepatocyte ploidy is controlled by the PIDDosome during development and regeneration. This multi-protein complex is activated by supernumerary centrosomes to induce p53 and restrict proliferation of polyploid cells, otherwise prone for chromosomal instability. PIDDosome deficiency in the liver results in drastically increased polyploidy. To investigate PIDDosome-induced p53-activation in the pathogenesis of liver cancer, we chemically induced hepatocellular carcinoma (HCC) in mice. Strikingly, PIDDosome deficiency reduced tumor number and burden, despite the inability to activate p53 in polyploid cells. Liver tumors arise primarily from cells with low ploidy, indicating an intrinsic pro-tumorigenic effect of PIDDosome-mediated ploidy restriction. These data suggest that hyperpolyploidization caused by PIDDosome deficiency protects from HCC. Moreover, high tumor cell density, as a surrogate marker of low ploidy, predicts poor survival of HCC patients receiving liver transplantation. Together, we show that the PIDDosome is a potential therapeutic target to manipulate hepatocyte polyploidization for HCC prevention and that tumor cell density may serve as a novel prognostic marker for recurrence-free survival in HCC patients.
Collapse
Affiliation(s)
- Valentina C Sladky
- Institute of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Katja Knapp
- Institute of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Tamas G Szabo
- Institute of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Vincent Z Braun
- Institute of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Laura Bongiovanni
- Department of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Hilda van den Bos
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Diana CJ Spierings
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Bart Westendorp
- Department of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Ana Curinha
- Institute of PathophysiologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsUniversity Hospital GrazGrazAustria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Gerald Timelthaler
- Institute for Cancer ResearchInternal Medicine IMedical University of ViennaViennaAustria
| | - Kaoru Tsuchia
- Department of Gastroenterology & HepatologyMusashino Red Cross HospitalTokyoJapan
| | - Matthias Pinter
- Division of Gastroenterology and HepatologyDepartment of Medicine IIIMedical University of ViennaViennaAustria
| | - Georg Semmler
- Division of Gastroenterology and HepatologyDepartment of Medicine IIIMedical University of ViennaViennaAustria
| | - Floris Foijer
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Alain de Bruin
- Department of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
- Department PediatricsUniversity Medical Center GroningenUniversity GroningenGroningenThe Netherlands
| | - Thomas Reiberger
- Division of Gastroenterology and HepatologyDepartment of Medicine IIIMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI‐RUD)ViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Nataliya Rohr‐Udilova
- Division of Gastroenterology and HepatologyDepartment of Medicine IIIMedical University of ViennaViennaAustria
| | - Andreas Villunger
- Institute of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI‐RUD)ViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| |
Collapse
|
49
|
A genomic-clinicopathologic nomogram for predicting overall survival of hepatocellular carcinoma. BMC Cancer 2020; 20:1176. [PMID: 33261584 PMCID: PMC7709450 DOI: 10.1186/s12885-020-07688-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common digestive tumor with great heterogeneity and different overall survival (OS) time, causing stern problems for selecting optimal treatment. Here we aim to establish a nomogram to predict the OS in HCC patients. Methods International Cancer Genome Consortium (ICGC) database was searched for the target information in our study. Lasso regression, univariate and multivariate cox analysis were applied during the analysis process. And a nomogram integrating model scoring and clinical characteristic was drawn. Results Six mRNAs were screened out by Lasso regression to make a model for predicting the OS of HCC patients. And this model was proved to be an independent prognostic model predicting OS in HCC patients. The area under the ROC curve (AUC) of this model was 0.803. TCGA database validated the significant value of this 6-mRNA model. Eventually a nomogram including 6-mRNA risk score, gender, age, tumor stage and prior malignancy was set up to predict the OS in HCC patients. Conclusions We established an independent prognostic model of predicting OS for 1–3 years in HCC patients, which is available to all populations. And we developed a nomogram on the basis of this model, which could be of great help to precisely individual treatment measures.
Collapse
|
50
|
Xiong H, Liu S, Wei T, Cheng Q, Siegwart DJ. Theranostic dendrimer-based lipid nanoparticles containing PEGylated BODIPY dyes for tumor imaging and systemic mRNA delivery in vivo. J Control Release 2020; 325:198-205. [PMID: 32629133 DOI: 10.1016/j.jconrel.2020.06.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/26/2022]
Abstract
mRNA-based therapeutics have emerged as a promising approach to treat cancer. However, creation of theranostic strategies to both deliver mRNA and simultaneously detect cancer is an important unmet goal. Here, we report the development of theranostic dendrimer-based lipid nanoparticle (DLNP) system containing PEGylated BODIPY dyes (PBD) for mRNA delivery and near-infrared (NIR) imaging in vitro and in vivo. DLNPs formulated with a pH-responsive PBD-lipid produced 5- to 35-fold more functional protein than control DLNPs formulated with traditional PEG-lipid in vitro and enabled higher mRNA delivery potency in vivo at a low dose of 0.1 mg kg-1 when formulated with a PBD-lipid containing a BODIPY core, indole linker, and PEG length between 1000 and 5000 g/mol. Moreover, we found the intensity of mRNA expression in the liver correlated with the pKa of DLNPs, indicating that DLNPs with a pKa close to 6.3 could generally produce more protein in livers. Notably, 4A3-SC8&PEG2k5d formulated DLNPs successfully mediated mRNA expression in tumors and simultaneously illuminated tumors via pH-responsive NIR imaging. The described theranostic lipid nanoparticles that combine mRNA delivery and NIR imaging hold promise as an applicable future approach to simultaneously detect and treat cancer.
Collapse
Affiliation(s)
- Hu Xiong
- The University of Texas Southwestern Medical Center, Department of Biochemistry, Simmons Comprehensive Cancer Center, Dallas, TX 75390, United States; Nankai University, Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Tianjin 300071, China
| | - Shuai Liu
- The University of Texas Southwestern Medical Center, Department of Biochemistry, Simmons Comprehensive Cancer Center, Dallas, TX 75390, United States
| | - Tuo Wei
- The University of Texas Southwestern Medical Center, Department of Biochemistry, Simmons Comprehensive Cancer Center, Dallas, TX 75390, United States
| | - Qiang Cheng
- The University of Texas Southwestern Medical Center, Department of Biochemistry, Simmons Comprehensive Cancer Center, Dallas, TX 75390, United States
| | - Daniel J Siegwart
- The University of Texas Southwestern Medical Center, Department of Biochemistry, Simmons Comprehensive Cancer Center, Dallas, TX 75390, United States.
| |
Collapse
|