1
|
Ji M, Ganesan S, Xia B, Huo Y. Targeting c-MET Alterations in Cancer: A Review of Genetic Drivers and Therapeutic Implications. Cancers (Basel) 2025; 17:1493. [PMID: 40361420 PMCID: PMC12070916 DOI: 10.3390/cancers17091493] [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: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Recent research has increasingly highlighted alterations in the proto-oncogene MET, whose abnormal activation has been implicated in multiple cancers. MET encodes c-MET, a receptor tyrosine kinase critical for cellular growth, survival, and migration. Aberrant c-MET signaling, driven by mutations or gene amplification, promotes proliferation and invasion, contributing to tumorigenesis. SCOPE OF THE REVIEW While MET mutations are most often observed in non-small cell lung cancer (NSCLC), they also occur in other malignancies, including breast and gastric cancers. This review highlights key MET alterations, such as gene amplification, gene fusions, and exon 14 skipping deletions, and examines their prevalence across various tumor types. MAJOR CONCLUSIONS We discuss the clinical significance of c-MET as a therapeutic target and identify gaps in knowledge that could inform the development of alternative treatment strategies.
Collapse
Affiliation(s)
- Michelle Ji
- Department of Radiation Oncology, Rutgers Cancer Institute, 195 Little Albany Street, New Brunswick, NJ 08903, USA; (M.J.); (B.X.)
- University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Shridar Ganesan
- NYU Langone Perlmutter Cancer Center, 160 East 34th Street, New York, NY 10016, USA
| | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute, 195 Little Albany Street, New Brunswick, NJ 08903, USA; (M.J.); (B.X.)
| | - Yanying Huo
- Department of Radiation Oncology, Rutgers Cancer Institute, 195 Little Albany Street, New Brunswick, NJ 08903, USA; (M.J.); (B.X.)
| |
Collapse
|
2
|
Luo R, Xu L, Chen J, Zhang W, Feng S, Qiu Z, Hong Y, Feng G. Near-Infrared Fluorescent Probe for Simultaneously Imaging Ferrous Ions and Viscosity in a Mouse Model of Hepatocellular Carcinoma. Anal Chem 2025; 97:1719-1728. [PMID: 39815399 DOI: 10.1021/acs.analchem.4c05120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Abnormal ferrous ion (Fe2+) levels lead to an increase in reactive oxygen species (ROS) in cells, disrupting intracellular viscosity and the occurrence of hepatocellular carcinoma (HCC). Simultaneously visualizing Fe2+ and intracellular viscosity is essential for understanding the detailed pathophysiological processes of HCC. Herein, we report the first dual-responsive probe, QM-FV, capable of simultaneously monitoring Fe2+ and viscosity. QM-FV shows highly selective turn-on near-infrared fluorescence (∼30-fold enhancement at 740 nm) for Fe2+ with high sensitivity (LOD = 25 nM) and a significant Stokes shift (290 nm). Moreover, QM-FV shows a distinct orange-red fluorescence enhancement at 587 nm as the viscosity increases. Due to its lower cytotoxicity and high sensitivity, QM-FV can distinguish cancer cells from normal cells by detecting Fe2+ and viscosity in dual channels. More importantly, using QM-FV, we found that the levels of Fe2+ and viscosity elevated in the precancerous stage of HCC and gradually increased as the disease progressed. Overall, this work provides a new potential tool for investigating viscosity and Fe2+-related pathological processes underlying HCC.
Collapse
Affiliation(s)
- Rongqing Luo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Li Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Wenxuan Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Shumin Feng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guoqiang Feng
- College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, China
| |
Collapse
|
3
|
Mo Y, Han Y, Chen Y, Fu C, Li Q, Liu Z, Xiao M, Xu B. ZDHHC20 mediated S-palmitoylation of fatty acid synthase (FASN) promotes hepatocarcinogenesis. Mol Cancer 2024; 23:274. [PMID: 39696259 DOI: 10.1186/s12943-024-02195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Protein palmitoylation is a reversible fatty acyl modification that undertakes important functions in multiple physiological processes. Dysregulated palmitoylations are frequently associated with the formation of cancer. How palmitoyltransferases for S-palmitoylation are involved in the occurrence and development of hepatocellular carcinoma (HCC) is largely unknown. METHODS Chemical carcinogen diethylnitrosamine (DEN)-induced and DEN combined CCl4 HCC models were used in the zinc finger DHHC-type palmitoyltransferase 20 (ZDHHC20) knockout mice to investigate the role of ZDHHC20 in HCC tumourigenesis. Palmitoylation liquid chromatography-mass spectrometry analysis, acyl-biotin exchange assay, co-immunoprecipitation, ubiquitination assays, protein half-life assays and immunofluorescence microscopy were conducted to explore the downstream regulators and corresponding mechanisms of ZDHHC20 in HCC. RESULTS Knocking out of ZDHHC20 significantly reduced hepatocarcinogenesis induced by chemical agents in the two HCC mouse models in vivo. 97 proteins with 123 cysteine sites were found to be palmitoylated in a ZDHHC20-dependent manner. Among these, fatty acid synthase (FASN) was palmitoylated at cysteines 1471 and 1881 by ZDHHC20. The genetic knockout or pharmacological inhibition of ZDHHC20, as well as the mutation of the critical cysteine sites of FASN (C1471S/C1881S) accelerated the degradation of FASN. Furthermore, ZDHHC20-mediated FASN palmitoylation competed against the ubiquitin-proteasome pathway via the E3 ubiquitin ligase complex SNX8-TRIM28. CONCLUSIONS Our findings demonstrate the critical role of ZDHHC20 in promoting hepatocarcinogenesis, and a mechanism underlying a mutual restricting mode for protein palmitoylation and ubiquitination modifications.
Collapse
Affiliation(s)
- Yaqi Mo
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Yamei Han
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yang Chen
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Radiation Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Chunling Fu
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qing Li
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Zhuang Liu
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Mingming Xiao
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Bo Xu
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China.
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
4
|
Chiu V, Yee C, Main N, Stevanovski I, Watt M, Wilson T, Angus P, Roberts T, Shackel N, Herath C. Oncogenic plasmid DNA and liver injury agent dictates liver cancer development in a mouse model. Clin Sci (Lond) 2024; 138:1227-1248. [PMID: 39254423 PMCID: PMC11427747 DOI: 10.1042/cs20240560] [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: 04/07/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/11/2024]
Abstract
Primary liver cancer is an increasing problem worldwide and is associated with significant mortality. A popular method of modeling liver cancer in mice is plasmid hydrodynamic tail vein injection (HTVI). However, plasmid-HTVI models rarely recapitulate the chronic liver injury which precedes the development of most human liver cancer. We sought to investigate how liver injury using thioacetamide contributes to the pathogenesis and progression of liver cancer in two oncogenic plasmid-HTVI-induced mouse liver cancer models. Fourteen-week-old male mice received double-oncogene plasmid-HTVI (SB/AKT/c-Met and SB/AKT/NRas) and then twice-weekly intraperitoneal injections of thioacetamide for 6 weeks. Liver tissue was examined for histopathological changes, including fibrosis and steatosis. Further characterization of fibrosis and inflammation was performed with immunostaining and real-time quantitative PCR. RNA sequencing with pathway analysis was used to explore novel pathways altered in the cancer models. Hepatocellular and cholangiocellular tumors were observed in mice injected with double-oncogene plasmid-HTVI models (SB/AKT/c-Met and SB/AKT/NRas). Thioacetamide induced mild fibrosis and increased alpha smooth muscle actin-expressing cells. However, the combination of plasmids and thioacetamide did not significantly increase tumor size, but increased multiplicity of small neoplastic lesions. Cancer and/or liver injury up-regulated profibrotic and proinflammatory genes while metabolic pathway genes were mostly down-regulated. We conclude that the liver injury microenvironment can interact with liver cancer and alter its presentation. However, the effects on cancer development vary depending on the genetic drivers with differing active oncogenic pathways. Therefore, the choice of plasmid-HTVI model and injury agent may influence the extent to which injury promotes liver cancer development.
Collapse
Affiliation(s)
- Vincent Chiu
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Christine Yee
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Nathan Main
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Igor Stevanovski
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Matthew Watt
- School of Biomedical Sciences, University of Melbourne, Victoria, Australia
| | - Trevor Wilson
- Hudson Institute of Medical Research, Monash University, Victoria, Australia
| | - Peter Angus
- Department of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Tara Roberts
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Nicholas Shackel
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Chandana Herath
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
- Department of Medicine, Austin Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Cigliano A, Liao W, Deiana GA, Rizzo D, Chen X, Calvisi DF. Preclinical Models of Hepatocellular Carcinoma: Current Utility, Limitations, and Challenges. Biomedicines 2024; 12:1624. [PMID: 39062197 PMCID: PMC11274649 DOI: 10.3390/biomedicines12071624] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the predominant primary liver tumor, remains one of the most lethal cancers worldwide, despite the advances in therapy in recent years. In addition to the traditional chemically and dietary-induced HCC models, a broad spectrum of novel preclinical tools have been generated following the advent of transgenic, transposon, organoid, and in silico technologies to overcome this gloomy scenario. These models have become rapidly robust preclinical instruments to unravel the molecular pathogenesis of liver cancer and establish new therapeutic approaches against this deadly disease. The present review article aims to summarize and discuss the commonly used preclinical models for HCC, evaluating their strengths and weaknesses.
Collapse
Affiliation(s)
- Antonio Cigliano
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.A.D.); (D.R.)
| | - Weiting Liao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (W.L.); (X.C.)
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Giovanni A. Deiana
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.A.D.); (D.R.)
| | - Davide Rizzo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.A.D.); (D.R.)
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (W.L.); (X.C.)
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Diego F. Calvisi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.A.D.); (D.R.)
| |
Collapse
|
6
|
Cigliano A, Simile MM, Vidili G, Pes GM, Dore MP, Urigo F, Cossu E, Che L, Feo C, Steinmann SM, Ribback S, Pascale RM, Evert M, Chen X, Calvisi DF. Fatty Acid Synthase Promotes Hepatocellular Carcinoma Growth via S-Phase Kinase-Associated Protein 2/p27 KIP1 Regulation. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1160. [PMID: 39064589 PMCID: PMC11278665 DOI: 10.3390/medicina60071160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Aberrant upregulation of fatty acid synthase (FASN), catalyzing de novo synthesis of fatty acids, occurs in various tumor types, including human hepatocellular carcinoma (HCC). Although FASN oncogenic activity seems to reside in its pro-lipogenic function, cumulating evidence suggests that FASN's tumor-supporting role might also be metabolic-independent. Materials and Methods: In the present study, we show that FASN inactivation by specific small interfering RNA (siRNA) promoted the downregulation of the S-phase kinase associated-protein kinase 2 (SKP2) and the consequent induction of p27KIP1 in HCC cell lines. Results: Expression levels of FASN and SKP2 directly correlated in human HCC specimens and predicted a dismal outcome. In addition, forced overexpression of SKP2 rendered HCC cells resistant to the treatment with the FASN inhibitor C75. Furthermore, FASN deletion was paralleled by SKP2 downregulation and p27KIP1 induction in the AKT-driven HCC preclinical mouse model. Moreover, forced overexpression of an SKP2 dominant negative form or a p27KIP1 non-phosphorylatable (p27KIP1-T187A) construct completely abolished AKT-dependent hepatocarcinogenesis in vitro and in vivo. Conclusions: In conclusion, the present data indicate that SKP2 is a critical downstream effector of FASN and AKT-dependent hepatocarcinogenesis in liver cancer, envisaging the possibility of effectively targeting FASN-positive liver tumors with SKP2 inhibitors or p27KIP1 activators.
Collapse
Affiliation(s)
- Antonio Cigliano
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Maria M. Simile
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Gianpaolo Vidili
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Giovanni M. Pes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Maria P. Dore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Francesco Urigo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Eleonora Cossu
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
| | - Claudio Feo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Sara M. Steinmann
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, 17489 Greifswald, Germany;
| | - Rosa M. Pascale
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Diego F. Calvisi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| |
Collapse
|
7
|
Zhu B, Chen X, Zhang T, Zhang Q, Fu K, Hua J, Zhang M, Qi Q, Zhao B, Zhao M, Yang L, Zhou B. Interactions between intestinal microbiota and metabolites in zebrafish larvae exposed to polystyrene nanoplastics: Implications for intestinal health and glycolipid metabolism. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134478. [PMID: 38696962 DOI: 10.1016/j.jhazmat.2024.134478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/09/2024] [Accepted: 04/27/2024] [Indexed: 05/04/2024]
Abstract
Previous studies have shown the harmful effects of nanoscale particles on the intestinal tracts of organisms. However, the specific mechanisms remain unclear. Our present study focused on examining the uptake and distribution of polystyrene nanoplastics (PS-NPs) in zebrafish larvae, as well as its toxic effects on the intestine. It was found that PS-NPs, marked with red fluorescence, primarily accumulated in the intestine section. Subsequently, zebrafish larvae were exposed to normal PS-NPs (0.2-25 mg/L) over a critical 10-day period for intestinal development. Histopathological analysis demonstrated that PS-NPs caused structural changes in the intestine, resulting in inflammation and oxidative stress. Additionally, PS-NPs disrupted the composition of the intestinal microbiota, leading to alterations in the abundance of bacterial genera such as Pseudomonas and Aeromonas, which are associated with intestinal inflammation. Metabolomics analysis showed alterations in metabolites that are primarily involved in glycolipid metabolism. Furthermore, MetOrigin analysis showed a significant correlation between bacterial flora (Pedobacter and Bacillus) and metabolites (D-Glycerate 2-phosphate and D-Glyceraldehyde 3-phosphate), which are related to the glycolysis/gluconeogenesis pathways. These findings were further validated through alterations in multiple biomarkers at various levels. Collectively, our data suggest that PS-NPs may impair the intestinal health, disrupt the intestinal microbiota, and subsequently cause metabolic disorders.
Collapse
Affiliation(s)
- Biran Zhu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Xianglin Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Taotao Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Qianqian Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Kaiyu Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jianghuan Hua
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Mengyuan Zhang
- Key Laboratory of Fermentation Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Qing Qi
- Wuhan Business University, Wuhan 430056, China
| | - Binbin Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
8
|
Cao R, Guo S, Min L, Li P. Roles of Rictor alterations in gastrointestinal tumors (Review). Oncol Rep 2024; 51:37. [PMID: 38186315 PMCID: PMC10807360 DOI: 10.3892/or.2024.8696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
Gastrointestinal tumors account for five of the top 10 causes of mortality from all cancers (colorectal, liver, stomach, esophageal and pancreatic cancer). Mammalian target of rapamycin (mTOR) signaling is commonly dysregulated in various human cancers. As a core component of the mTOR complex 2 (mTORC2), Rictor is a key effector molecule of the PI3K/Akt pathway. A high alteration rate of Rictor has been observed in gastrointestinal tumors, and such Rictor alterations are often associated with resistance to chemotherapy and related adverse clinical outcomes. However, the exact roles of Rictor in gastrointestinal tumors remain elusive. The aim of the present study was to critically discuss the following: i) Mutation and biological characteristics of Rictor in tumors with a detailed overview of Rictor in cell proliferation, angiogenesis, apoptosis, autophagy and drug resistance; ii) the role of Rictor in tumors of the digestive system, particularly colorectal, hepatobiliary, gastric, esophageal and pancreatic cancer and cholangiocarcinoma; and iii) the current status and prospects of targeted therapy for Rictor by inhibiting Akt activation. Despite the growing realization of the importance of Rictor/mTORC2 in cancer, the underlying mechanistic details remain poorly understood; this needs to change in order for the development of efficient targeted therapies and re‑sensitization of therapy‑resistant cancers to be made possible.
Collapse
Affiliation(s)
- Ruizhen Cao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, P.R. China
| | - Shuilong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, P.R. China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, P.R. China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, P.R. China
| |
Collapse
|
9
|
Bang J, Jun M, Lee S, Moon H, Ro SW. Targeting EGFR/PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma. Pharmaceutics 2023; 15:2130. [PMID: 37631344 PMCID: PMC10458925 DOI: 10.3390/pharmaceutics15082130] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a significant global health concern, with its incidence steadily increasing. The development of HCC is a multifaceted, multi-step process involving alterations in various signaling cascades. In recent years, significant progress has been made in understanding the molecular signaling pathways that play central roles in hepatocarcinogenesis. In particular, the EGFR/PI3K/AKT/mTOR signaling pathway in HCC has garnered renewed attention from both basic and clinical researchers. Preclinical studies in vitro and in vivo have shown the effectiveness of targeting the key components of this signaling pathway in human HCC cells. Thus, targeting these signaling pathways with small molecule inhibitors holds promise as a potential therapeutic option for patients with HCC. In this review, we explore recent advancements in understanding the role of the EGFR/PI3K/AKT/mTOR signaling pathway in HCC and assess the effectiveness of targeting this signaling cascade as a potential strategy for HCC therapy based on preclinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Simon Weonsang Ro
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea; (J.B.); (M.J.); (S.L.); (H.M.)
| |
Collapse
|
10
|
Zhou Y, Cui G, Xu H, Chun J, Yang D, Zhang Z, Yang L, Wang J, Wan M, Calvisi DF, Lin S, Chen X, Wang H. Loss of TP53 cooperates with c-MET overexpression to drive hepatocarcinogenesis. Cell Death Dis 2023; 14:476. [PMID: 37500626 PMCID: PMC10374654 DOI: 10.1038/s41419-023-05958-y] [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: 10/17/2022] [Revised: 06/13/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is a deadly malignancy with high genetic heterogeneity. TP53 mutation and c-MET activation are frequent events in human HCCs. Here, we discovered that the simultaneous mutations in TP53 and activation of c-MET occur in ~20% of human HCCs, and these patients show a poor prognosis. Importantly, we found that concomitant deletion of Trp53 and overexpression of c-MET (c-MET/sgp53) in the mouse liver led to HCC formation in vivo. Consistent with human HCCs, RNAseq showed that c-MET/sgp53 mouse HCCs were characterized by activated c-MET and Ras/MAPK cascades and increased tumor cell proliferation. Subsequently, a stably passaged cell line derived from a c-MET/sgp53 HCC and corresponding subcutaneous xenografts were generated. Also, in silico analysis suggested that the MEK inhibitor trametinib has a higher inhibition score in TP53 null human HCC cell lines, which was validated experimentally. We consistently found that trametinib effectively inhibited the growth of c-MET/sgp53 HCC cells and xenografts, supporting the possible usefulness of this drug for treating human HCCs with TP53-null mutations. Altogether, our study demonstrates that loss of TP53 cooperates with c-MET to drive hepatocarcinogenesis in vivo. The c-MET/sgp53 mouse model and derived HCC cell lines represent novel and useful preclinical tools to study hepatocarcinogenesis in the TP53 null background.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
| | - Guofei Cui
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Hongwei Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Joanne Chun
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Doris Yang
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Zheng Zhang
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Yang
- Beijing University of Chinese Medicine, Beijing, China
| | - Jingxiao Wang
- School of Life Sciences, Beijing, University of Chinese Medicine, Beijing, China
| | - Meijuan Wan
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, 93053, Germany
| | - Shumei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA.
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA.
| | - Haichuan Wang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Zhang C, Song Y, Yuan M, Chen L, Zhang Q, Hu J, Meng Y, Li S, Zheng G, Qiu Z. Ellagitannins-Derived Intestinal Microbial Metabolite Urolithin A Ameliorates Fructose-Driven Hepatosteatosis by Suppressing Hepatic Lipid Metabolic Reprogramming and Inducing Lipophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3967-3980. [PMID: 36825491 DOI: 10.1021/acs.jafc.2c05776] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Excessive fructose consumption exacerbates the progression of nonalcoholic fatty liver disease (NAFLD) by disrupting hepatic lipid homeostasis. This study sought to evaluate the efficacy of urolithin A (UroA) in a fructose-induced NAFLD mouse model. UroA was administered in the high-fructose-fed mice to investigate the antisteatotic effects in vivo. Fructose-stimulated HepG2 cells and primary hepatocytes were established for in vitro mechanistic assessment. The results suggested that UroA ameliorated fructose-induced hepatic steatosis in mice. Mechanistically, UroA impaired lipogenesis and enhanced β-oxidation in the livers of fructose-fed mice. Notably, UroA facilitated hepatic lipophagy through the AMPK/ULK1 pathway both in vivo and in vitro, degrading lipid droplets for fueling β-oxidation. This study indicates that UroA alleviates excessive lipid accumulation and restores lipid homeostasis in the livers of fructose-fed mice by suppressing lipid metabolic reprogramming and triggering lipophagy. Therefore, dietary supplementation of UroA or ellagitannins-rich foods may be beneficial for NAFLD individuals with high fructose intake.
Collapse
Affiliation(s)
- Cong Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yingying Song
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Ming Yuan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Liang Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Qianyu Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Junjie Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yan Meng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Shan Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China
- Department of Biochemistry, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| |
Collapse
|
12
|
The Role of PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma Metabolism. Int J Mol Sci 2023; 24:ijms24032652. [PMID: 36768977 PMCID: PMC9916527 DOI: 10.3390/ijms24032652] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths in the world. Metabolic reprogramming is considered a new hallmark of cancer, but it remains unclearly described in HCC. The dysregulation of the PI3K/AKT/mTOR signaling pathway is common in HCC and is, therefore, a topic of further research and the concern of developing a novel target for liver cancer therapy. In this review, we illustrate mechanisms by which this signaling network is accountable for regulating HCC cellular metabolism, including glucose metabolism, lipid metabolism, amino acid metabolism, pyrimidine metabolism, and oxidative metabolism, and summarize the ongoing clinical trials based on the inhibition of the PI3K/AKT/mTOR pathway in HCC.
Collapse
|
13
|
Tang B, Zhu J, Wang Y, Chen W, Fang S, Mao W, Xu Z, Yang Y, Weng Q, Zhao Z, Chen M, Ji J. Targeted xCT-mediated Ferroptosis and Protumoral Polarization of Macrophages Is Effective against HCC and Enhances the Efficacy of the Anti-PD-1/L1 Response. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203973. [PMID: 36442849 PMCID: PMC9839855 DOI: 10.1002/advs.202203973] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/26/2022] [Indexed: 05/14/2023]
Abstract
Tumor-associated macrophages (TAMs) play an essential role in tumor progression, metastasis, and antitumor immunity. Ferroptosis has attracted extensive attention for its lethal effect on tumor cells, but the role of ferroptosis in TAMs and its impact on tumor progression have not been clearly defined. Using transgenic mouse models, this study determines that xCT-specific knockout in macrophages is sufficient to limit tumorigenicity and metastasis in the mouse HCC models, achieved by reducing TAM recruitment and infiltration, inhibiting M2-type polarization, and activating and enhancing ferroptosis activity within TAMs. The SOCS3-STAT6-PPAR-γ signaling may be a crucial pathway in macrophage phenotypic shifting, and activation of intracellular ferroptosis is associated with GPX4/RRM2 signaling regulation. Furthermore, that xCT-mediated macrophage ferroptosis significantly increases PD-L1 expression in macrophages and improves the antitumor efficacy of anti-PD-L1 therapy is unveiled. The constructed Man@pSiNPs-erastin specifically targets macrophage ferroptosis and protumoral polarization and combining this treatment with anti-PD-L1 exerts substantial antitumor efficacy. xCT expression in tumor tissues, especially in CD68+ macrophages, can serve as a reliable factor to predict the prognosis of HCC patients. These findings provide further insight into targeting ferroptosis activation in TAMs and regulating TAM infiltration and functional expression to achieve precise tumor prevention and improve therapeutic efficacy.
Collapse
Affiliation(s)
- Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
| | - Jinyu Zhu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
| | - Yajie Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Weiyang Mao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Ziwei Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
| | - Yang Yang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Qiaoyou Weng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention ResearchInstitute of Imaging Diagnosis and Minimally Invasive Intervention ResearchThe Fifth Affiliated Hospital of Wenzhou Medical UniversityLishui Hospital of Zhejiang UniversityLishui323000China
- Clinical College of The Affiliated Central HospitalSchool of MedicineLishui UniversityLishui323000China
| |
Collapse
|
14
|
Fornari F, Giovannini C, Piscaglia F, Gramantieri L. Animal Models of Hepatocellular Carcinoma: Current Applications in Clinical Research. J Hepatocell Carcinoma 2022; 9:1263-1278. [PMID: 36523954 PMCID: PMC9744868 DOI: 10.2147/jhc.s347946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/23/2022] [Indexed: 01/11/2025] Open
Abstract
In the last decade, relevant advances have occurred in the treatment of hepatocellular carcinoma (HCC), with novel drugs entering the clinical practice, among which tyrosine kinase inhibitors (TKIs) such as lenvatinib, cabozantinib and regorafenib, and immune checkpoint inhibitors (ICPIs) either alone or in combination with VEGF inhibitors. Clinical trials have driven the introduction of such novel molecules into the clinics but, at present, no biomarker drives the choice of first-line options, which relies only upon clinical and imaging assessment. Remarkably, clinical and imaging-based evaluations do not consider the huge heterogeneity of HCC and do not allow to realize the potential of personalized treatments. Preclinical research still does not inform the design of clinical trials, even though many animal models mimicking specific subgroups of HCC are available and might provide relevant information. Although animal models directly informing the clinical practice, such as patients-derived xenografts, are not used to help the choice of treatment in advanced HCC, however, the preclinical research can count on a wide range of valuable models. Here we will review some HCC models which might turn informative for specific questions in defined patient subgroups, and we will describe recent preclinical studies for the mechanistic evaluation of immunotherapy-based treatment approaches. To this aim, we will mainly focus on two issues: (i) HCC models informative on NAFLD-NASH HCC and (ii) HCC models helping to elucidate mechanisms underneath immunotherapy. We have chosen these two settings since they represent, respectively, the most rapidly arising cause of chronic liver disease (CLD) and HCC in western countries and the most promising therapeutic option for advanced HCC.
Collapse
Affiliation(s)
- Francesca Fornari
- Centre for Applied Biomedical Research - CRBA, University of Bologna, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Catia Giovannini
- Centre for Applied Biomedical Research - CRBA, University of Bologna, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
15
|
Ji X, Chen X, Sheng L, Deng D, Wang Q, Meng Y, Qiu Z, Zhang B, Zheng G, Hu J. Metabolomics profiling of AKT/c-Met-induced hepatocellular carcinogenesis and the inhibitory effect of Cucurbitacin B in mice. Front Pharmacol 2022; 13:1009767. [PMID: 36506561 PMCID: PMC9728611 DOI: 10.3389/fphar.2022.1009767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC), the most common kind of liver cancer, accounts for the majority of liver cancer diagnoses and fatalities. Clinical aggressiveness, resistance to traditional therapy, and a high mortality rate are all features of this disease. Our previous studies have shown that co-activation of AKT and c-Met induces HCC development, which is the malignant biological feature of human HCC. Cucurbitacin B (CuB), a naturally occurring tetracyclic triterpenoid compound with potential antitumor activity. However, the metabolic mechanism of AKT/c-Met-induced Hepatocellular Carcinogenesis and CuB in HCC remains unclear. In this study, we established an HCC mouse model by hydrodynamically transfecting active AKT and c-Met proto-oncogenes. Based on the results of hematoxylin-eosin (H&E), oil red O (ORO) staining, and immunohistochemistry (IHC), HCC progression was divided into two stages: the early stage of HCC (3 weeks after AKT/c-Met injection) and the formative stage of HCC (6 weeks after AKT/c-Met injection), and the therapeutic effect of CuB was evaluated. Through UPLC-Q-TOF-MS/MS metabolomics, a total of 26 distinct metabolites were found in the early stage of HCC for serum samples, while in the formative stage of HCC, 36 distinct metabolites were found in serum samples, and 13 different metabolites were detected in liver samples. 33 metabolites in serum samples and 11 in live samples were affected by CuB administration. Additionally, metabolic pathways and western blotting analysis revealed that CuB influences lipid metabolism, amino acid metabolism, and glucose metabolism by altering the AKT/mTORC1 signaling pathway, hence decreasing tumor progression. This study provides a metabolic basis for the early diagnosis, therapy, and prognosis of HCC and the clinical application of CuB in HCC.
Collapse
Affiliation(s)
- Xiangyu Ji
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Xin Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Lei Sheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Dongjie Deng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Qi Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Yan Meng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Baohui Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Guohua Zheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China,Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, Hubei, China,*Correspondence: Guohua Zheng, ; Junjie Hu,
| | - Junjie Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China,*Correspondence: Guohua Zheng, ; Junjie Hu,
| |
Collapse
|
16
|
Wang H, Zhou Y, Xu H, Wang X, Zhang Y, Shang R, O'Farrell M, Roessler S, Sticht C, Stahl A, Evert M, Calvisi DF, Zeng Y, Chen X. Therapeutic efficacy of FASN inhibition in preclinical models of HCC. Hepatology 2022; 76:951-966. [PMID: 35076948 PMCID: PMC9309180 DOI: 10.1002/hep.32359] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Aberrant activation of fatty acid synthase (FASN) is a major metabolic event during the development of HCC. We evaluated the therapeutic efficacy of TVB3664, a FASN inhibitor, either alone or in combination, for HCC treatment. APPROACH AND RESULTS The therapeutic efficacy and the molecular pathways targeted by TVB3664, either alone or with tyrosine kinase inhibitors or the checkpoint inhibitor anti-programmed death ligand 1 antibody, were assessed in human HCC cell lines and multiple oncogene-driven HCC mouse models. RNA sequencing was performed to elucidate the effects of TVB3664 on global gene expression and tumor metabolism. TVB3664 significantly ameliorated the fatty liver phenotype in the aged mice and AKT-induced hepatic steatosis. TVB3664 monotherapy showed moderate efficacy in NASH-related murine HCCs, induced by loss of phosphatase and tensin homolog and MET proto-oncogene, receptor tyrosine kinase (c-MET) overexpression. TVB3664, in combination with cabozantinib, triggered tumor regression in this murine model but did not improve the responsiveness to immunotherapy. Global gene expression revealed that TVB3664 predominantly modulated metabolic processes, whereas TVB3664 synergized with cabozantinib to down-regulate multiple cancer-related pathways, especially the AKT/mammalian target of rapamycin pathway and cell proliferation genes. TVB3664 also improved the therapeutic efficacy of sorafenib and cabozantinib in the FASN-dependent c-MYC-driven HCC model. However, TVB3664 had no efficacy nor synergistic effects in FASN-independent murine HCC models. CONCLUSIONS This preclinical study suggests the limited efficacy of targeting FASN as monotherapy for HCC treatment. However, FASN inhibitors could be combined with other drugs for improved effectiveness. These combination therapies could be developed based on the driver oncogenes, supporting precision medicine approaches for HCC treatment.
Collapse
Affiliation(s)
- Haichuan Wang
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Yi Zhou
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Hongwei Xu
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Xue Wang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Yi Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Runze Shang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | | | | | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Yong Zeng
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| |
Collapse
|
17
|
Barzaman K, Vafaei R, Samadi M, Kazemi MH, Hosseinzadeh A, Merikhian P, Moradi-Kalbolandi S, Eisavand MR, Dinvari H, Farahmand L. Anti-cancer therapeutic strategies based on HGF/MET, EpCAM, and tumor-stromal cross talk. Cancer Cell Int 2022; 22:259. [PMID: 35986321 PMCID: PMC9389806 DOI: 10.1186/s12935-022-02658-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
As an intelligent disease, tumors apply several pathways to evade the immune system. It can use alternative routes to bypass intracellular signaling pathways, such as nuclear factor-κB (NF-κB), Wnt, and mitogen-activated protein (MAP)/phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR). Therefore, these mechanisms lead to therapeutic resistance in cancer. Also, these pathways play important roles in the proliferation, survival, migration, and invasion of cells. In most cancers, these signaling pathways are overactivated, caused by mutation, overexpression, etc. Since numerous molecules share these signaling pathways, the identification of key molecules is crucial to achieve favorable consequences in cancer therapy. One of the key molecules is the mesenchymal-epithelial transition factor (MET; c-Met) and its ligand hepatocyte growth factor (HGF). Another molecule is the epithelial cell adhesion molecule (EpCAM), which its binding is hemophilic. Although both of them are involved in many physiologic processes (especially in embryonic stages), in some cancers, they are overexpressed on epithelial cells. Since they share intracellular pathways, targeting them simultaneously may inhibit substitute pathways that tumor uses to evade the immune system and resistant to therapeutic agents.
Collapse
|
18
|
Leslie J, Geh D, Elsharkawy AM, Mann DA, Vacca M. Metabolic dysfunction and cancer in HCV: Shared pathways and mutual interactions. J Hepatol 2022; 77:219-236. [PMID: 35157957 DOI: 10.1016/j.jhep.2022.01.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
HCV hijacks many host metabolic processes in an effort to aid viral replication. The resulting hepatic metabolic dysfunction underpins many of the hepatic and extrahepatic manifestations of chronic hepatitis C (CHC). However, the natural history of CHC is also substantially influenced by the host metabolic status: obesity, insulin resistance and hepatic steatosis are major determinants of CHC progression toward hepatocellular carcinoma (HCC). Direct-acting antivirals (DAAs) have transformed the treatment and natural history of CHC. While DAA therapy effectively eradicates the virus, the long-lasting overlapping metabolic disease can persist, especially in the presence of obesity, increasing the risk of liver disease progression. This review covers the mechanisms by which HCV tunes hepatic and systemic metabolism, highlighting how systemic metabolic disturbance, lipotoxicity and chronic inflammation favour disease progression and a precancerous niche. We also highlight the therapeutic implications of sustained metabolic dysfunction following sustained virologic response as well as considerations for patients who develop HCC on the background of metabolic dysfunction.
Collapse
Affiliation(s)
- Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Geh
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ahmed M Elsharkawy
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Birmingham, B15 2TH UK; National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey.
| | - Michele Vacca
- Interdisciplinary Department of Medicine, Università degli Studi di Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
19
|
Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. BIOLOGY 2022; 11:biology11040585. [PMID: 35453784 PMCID: PMC9027240 DOI: 10.3390/biology11040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and its incidence is steadily increasing. The development of HCC is a complex, multi-step process that is accompanied by alterations in multiple signaling cascades. Recent years have seen advancement in understanding molecular signaling pathways that play central roles in hepatocarcinogenesis. Aberrant activation of YAP/TAZ, Hedgehog, or Wnt/β-catenin signaling is frequently found in a subset of HCC patients. Targeting the signaling pathway via small molecule inhibitors could be a promising therapeutic option for the subset of patients. In this review, we will introduce the signaling pathways, discuss their roles in the development of HCC, and propose a therapeutic approach targeting the signaling pathways in the context of HCC. Abstract Hepatocellular carcinoma (HCC) is a major health concern worldwide, and its incidence is increasing steadily. To date, receptor tyrosine kinases (RTKs) are the most favored molecular targets for the treatment of HCC, followed by immune checkpoint regulators such as PD-1, PD-L1, and CTLA-4. With less than desirable clinical outcomes from RTK inhibitors as well as immune checkpoint inhibitors (ICI) so far, novel molecular target therapies have been proposed for HCC. In this review, we will introduce diverse molecular signaling pathways that are aberrantly activated in HCC, focusing on YAP/TAZ, Hedgehog, and Wnt/β-catenin signaling pathways, and discuss potential therapeutic strategies targeting the signaling pathways in HCC.
Collapse
|
20
|
Modulation of the tumour microenvironment in hepatocellular carcinoma by tyrosine kinase inhibitors: from modulation to combination therapy targeting the microenvironment. Cancer Cell Int 2022; 22:73. [PMID: 35148789 PMCID: PMC8840552 DOI: 10.1186/s12935-021-02435-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide. Tyrosine kinase inhibitors (TKIs) remain the backbone of systematic therapy for advanced hepatocellular carcinoma. Sorafenib and lenvatinib are currently approved as first-line therapeutic drugs, and regorafenib and cabozantinib are applied as second-line treatments. With inhibition of angiogenesis as the main target, TKIs exert a profound effect on the tumour microenvironment (TME). The TME is a complex mixture of cellular and noncellular components surrounding the tumour mass, and is associated with tumour progression partially through the epithelial-mesenchymal transition. Specifically, the TME of HCC is characterized by profound extracellular matrix remodelling and an immunosuppressive microenvironment. The purpose of this review is to provide a summary of TME remodelling mediated by four Food and Drug Administration approved TKIs in HCC and thus summarize the rationale and potential targets for combination therapy. The modulatory effect of TKIs on the TME of HCC was reported to enhance the antitumour effect of TKIs through pyroptosis of macrophages and subsequent natural killer cell activation, T cell activation, regulatory T cell reduction in HCC. Meanwhile, TKIs also induce drug resistance via M2 polarization and accumulation, recruitment of tumour-associated neutrophils, and induction of the epithelial-mesenchymal transition. In conclusion, the effect of TKIs on TME can enhance its antitumour effect, but might also partially contribute to the drug resistance that hinders the progression of TKIs as treatment for HCC. Additionally, the effect of TKIs also provides the rationale for combination therapy, including combining TKIs with immune checkpoint inhibitors, to facilitate increased drug efficacy of TKIs.
Collapse
|
21
|
Abstract
Metabolic rewiring is one of the hallmarks of cancer. Altered de novo lipogenesis is one of the pivotal metabolic events deregulated in cancers. Sterol regulatory element-binding transcription factor 1 (SREBP1) controls the transcription of major enzymes involved in de novo lipogenesis, including ACLY, ACACA, FASN, and SCD. Studies have shown the increased de novo lipogenesis in human hepatocellular carcinoma (HCC) samples. Multiple mechanisms, such as activation of the AKT/mechanistic target of rapamycin (mTOR) pathway, lead to high SREBP1 induction and the coordinated enhanced expression of ACLY, ACACA, FASN, and SCD genes. Subsequent functional analyses have unraveled these enzymes' critical role(s) and the related de novo lipogenesis in hepatocarcinogenesis. Importantly, targeting these molecules might be a promising strategy for HCC treatment. This paper comprehensively summarizes de novo lipogenesis rewiring in HCC and how this pathway might be therapeutically targeted.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| |
Collapse
|
22
|
Downregulation of CYP39A1 Serves as a Novel Biomarker in Hepatocellular Carcinoma with Worse Clinical Outcome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:5175581. [PMID: 35003516 PMCID: PMC8741352 DOI: 10.1155/2021/5175581] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/19/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Background CYP39A1 is a poorly characterized metabolic enzyme that has been investigated in a few tumors. However, the role of CYP39A1 in hepatocellular carcinoma (HCC) has not yet been clarified. In this study, the expression and clinical significance of CYP39A1 in HCC were explored. Methods CYP39A1 protein expression was detected in Akt/c-Met-induced HCC mice and 14 paired fresh HCC samples as well as another 159 HCC and matched noncancerous tissues. Meanwhile, the mRNA expression was analyzed by GEO and TCGA analysis and validated in 14 paired fresh HCC tissues. Furthermore, the relationships between CYP39A1 expression and clinicopathologic features as well as prognosis were analyzed. HCC cell growth changes were analyzed by cell viability assays after CYP39A1 overexpression and then validated after CYP39A1 knockout by DepMap database analysis. Results CYP39A1 protein expression was lower expressed in HCC mouse models, and its mRNA and protein expression were also downregulated in HCC compared with noncancerous liver tissues. Higher CYP39A1 expression was associated with well differentiation. Moreover, survival analysis indicated that lower CYP39A1 expression was associated with poorer overall survival. In addition, HepG2 and SMMC-7721 cell viability were inhibited after CYP39A1 overexpression. Genome-wide CRISPR/Cas9 proliferation screening indicated that knockout of CYP39A1 could promote HCC cell growth. Likewise, p-NF-κB and Nrf2 were suppressed after CYP39A1 overexpression. It is worth mentioning that total bile acid, total bilirubin, and direct bilirubin were significantly increased in the patients with low CYP39A1 expression. Conclusions Downregulation of CYP39A1 is associated with HCC carcinogenesis, tumor differentiation, and poor overall survival, suggesting that CYP39A1 may serve as a tumor suppressor gene and novel biomarker for HCC patients.
Collapse
|
23
|
Gu CY, Lee TKW. Preclinical mouse models of hepatocellular carcinoma: An overview and update. Exp Cell Res 2022; 412:113042. [PMID: 35101391 DOI: 10.1016/j.yexcr.2022.113042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022]
|
24
|
Liang W, Shi C, Hong W, Li P, Zhou X, Fu W, Lin L, Zhang J. Super-enhancer-driven lncRNA-DAW promotes liver cancer cell proliferation through activation of Wnt/β-catenin pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1351-1363. [PMID: 34853732 PMCID: PMC8608597 DOI: 10.1016/j.omtn.2021.10.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/06/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022]
Abstract
Aberrant expression of long non-coding RNAs (lncRNAs) has been reported in multiple cancers. However, the underlying mechanisms mediated by super-enhancers remain elusive. Here we sought to define the role of a novel lncRNA termed lncRNA-DAW in tumorigenesis. Our results revealed that lncRNA-DAW was driven by a liver-specific super-enhancer and transcriptionally activated by HNF4G, leading to frequent elevation in hepatocellular carcinoma (HCC) specimens. Ectopic expression of lncRNA-DAW promoted both in vivo and in vitro tumor growth. By using RNA sequencing, Wnt2 was screened out as a downstream effector of lncRNA-DAW. We next found that lncRNA-DAW physically interacted with EZH2, a negative regulator of Wnt2. This interplay subsequently potentiated CDK1-EZH2 interaction, leading to the phosphorylation and ubiquitination of EZH2. The lncRNA-DAW-mediated EZH2 degradation facilitated the de-repression of Wnt2 transcription, which eventually activated the Wnt/β-catenin pathway. Furthermore, we verified that Wnt2 potentiated in vitro and in vivo cancer cell growth by activating the Wnt/β-catenin pathway. Finally, Wnt2 amplification was confirmed as a common event in liver cancer, and the expression of lncRNA-DAW was positively correlated with Wnt2 in HCC specimens. Collectively, we are the first to identify lncRNA-DAW as a novel candidate oncogene in liver cancer, and this lncRNA may serve as a novel clinical diagnosis biomarker for liver cancer.
Collapse
Affiliation(s)
- Weicheng Liang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China.,Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Chuanjian Shi
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Weilong Hong
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Panlong Li
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Xue Zhou
- Department of Ultrasonic Medicine, Guangzhou Women and Children's Medical Center, Guangzhou, P.R. China
| | - Weiming Fu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Lizhu Lin
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Jinfang Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China.,Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
25
|
Joechle K, Guenzle J, Hellerbrand C, Strnad P, Cramer T, Neumann UP, Lang SA. Role of mammalian target of rapamycin complex 2 in primary and secondary liver cancer. World J Gastrointest Oncol 2021; 13:1632-1647. [PMID: 34853640 PMCID: PMC8603445 DOI: 10.4251/wjgo.v13.i11.1632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) acts in two structurally and functionally distinct protein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Upon deregulation, activated mTOR signaling is associated with multiple processes involved in tumor growth and metastasis. Compared with mTORC1, much less is known about mTORC2 in cancer, mainly because of the unavailability of a selective inhibitor. However, existing data suggest that mTORC2 with its two distinct subunits Rictor and mSin1 might play a more important role than assumed so far. It is one of the key effectors of the PI3K/AKT/mTOR pathway and stimulates cell growth, cell survival, metabolism, and cytoskeletal organization. It is not only implicated in tumor progression, metastasis, and the tumor microenvironment but also in resistance to therapy. Rictor, the central subunit of mTORC2, was found to be upregulated in different kinds of cancers and is associated with advanced tumor stages and a bad prognosis. Moreover, AKT, the main downstream regulator of mTORC2/Rictor, is one of the most highly activated proteins in cancer. Primary and secondary liver cancer are major problems for current cancer therapy due to the lack of specific medical treatment, emphasizing the need for further therapeutic options. This review, therefore, summarizes the role of mTORC2/Rictor in cancer, with special focus on primary liver cancer but also on liver metastases.
Collapse
Affiliation(s)
- Katharina Joechle
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Sven Arke Lang
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| |
Collapse
|
26
|
Romualdo GR, Leroy K, Costa CJS, Prata GB, Vanderborght B, da Silva TC, Barbisan LF, Andraus W, Devisscher L, Câmara NOS, Vinken M, Cogliati B. In Vivo and In Vitro Models of Hepatocellular Carcinoma: Current Strategies for Translational Modeling. Cancers (Basel) 2021; 13:5583. [PMID: 34771745 PMCID: PMC8582701 DOI: 10.3390/cancers13215583] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer-related death globally. HCC is a complex multistep disease and usually emerges in the setting of chronic liver diseases. The molecular pathogenesis of HCC varies according to the etiology, mainly caused by chronic hepatitis B and C virus infections, chronic alcohol consumption, aflatoxin-contaminated food, and non-alcoholic fatty liver disease associated with metabolic syndrome or diabetes mellitus. The establishment of HCC models has become essential for both basic and translational research to improve our understanding of the pathophysiology and unravel new molecular drivers of this disease. The ideal model should recapitulate key events observed during hepatocarcinogenesis and HCC progression in view of establishing effective diagnostic and therapeutic strategies to be translated into clinical practice. Despite considerable efforts currently devoted to liver cancer research, only a few anti-HCC drugs are available, and patient prognosis and survival are still poor. The present paper provides a state-of-the-art overview of in vivo and in vitro models used for translational modeling of HCC with a specific focus on their key molecular hallmarks.
Collapse
Affiliation(s)
- Guilherme Ribeiro Romualdo
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Kaat Leroy
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Cícero Júlio Silva Costa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Gabriel Bacil Prata
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
| | - Wellington Andraus
- Department of Gastroenterology, Clinics Hospital, School of Medicine, University of São Paulo (HC-FMUSP), São Paulo 05403-000, Brazil;
| | - Lindsey Devisscher
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| |
Collapse
|
27
|
Deng S, Solinas A, Calvisi DF. Cabozantinib for HCC Treatment, From Clinical Back to Experimental Models. Front Oncol 2021; 11:756672. [PMID: 34722310 PMCID: PMC8548824 DOI: 10.3389/fonc.2021.756672] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Patients with early-stage HCC can be treated successfully with surgical resection or liver transplantation. However, the usual late diagnosis of HCC precludes curative treatments, and systemic therapies are the only viable option for inoperable patients. Sorafenib, an orally available multikinase inhibitor, is a systemic therapy approved for treating patients with advanced HCC yet providing limited benefits. Consequently, new drugs have been developed to overcome sorafenib resistance and improve patients' prognoses. A new promising strategy is using c-MET inhibitors, such as cabozantinib, as activation of c-MET occurs in up to 40% of HCC patients. In particular, cabozantinib, in combination with the checkpoint inhibitor atezolizumab, is currently in phase 3 clinical trial for HCC, and the results are eagerly awaited. Herein, we summarize and review the drugs approved for the treatment of advanced HCC, mainly focusing on the clinical and preclinical efficacy evaluation of cabozantinib. Also, we report the available preclinical data on cabozantinib-based combination therapies for HCC, current obstacles for cabozantinib therapy, and the future directions for cabozantinib-based treatment for HCC.
Collapse
Affiliation(s)
- Shanshan Deng
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, United States
| | - Antonio Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
28
|
Haberl EM, Pohl R, Rein-Fischboeck L, Höring M, Krautbauer S, Liebisch G, Buechler C. Accumulation of cholesterol, triglycerides and ceramides in hepatocellular carcinomas of diethylnitrosamine injected mice. Lipids Health Dis 2021; 20:135. [PMID: 34629057 PMCID: PMC8502393 DOI: 10.1186/s12944-021-01567-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/21/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Dysregulated lipid metabolism is critically involved in the development of hepatocellular carcinoma (HCC). The respective metabolic pathways affected in HCC can be identified using suitable experimental models. Mice injected with diethylnitrosamine (DEN) and fed a normal chow develop HCC. For the analysis of the pathophysiology of HCC in this model a comprehensive lipidomic analysis was performed. METHODS Lipids were measured in tumor and non-tumorous tissues by direct flow injection analysis. Proteins with a role in lipid metabolism were analysed by immunoblot. Mann-Whitney U-test or paired Student´s t-test were used for data analysis. RESULTS Intra-tumor lipid deposition is a characteristic of HCCs, and di- and triglycerides accumulated in the tumor tissues of the mice. Peroxisome proliferator-activated receptor gamma coactivator 1 alpha, lipoprotein lipase and hepatic lipase protein were low in the tumors whereas proteins involved in de novo lipogenesis were not changed. Higher rates of de novo lipogenesis cause a shift towards saturated acyl chains, which did not occur in the murine HCC model. Besides, LDL-receptor protein and cholesteryl ester levels were higher in the murine HCC tissues. Ceramides are cytotoxic lipids and are low in human HCCs. Notably, ceramide levels increased in the murine tumors, and the simultaneous decline of sphingomyelins suggests that sphingomyelinases were involved herein. DEN is well described to induce the tumor suppressor protein p53 in the liver, and p53 was additionally upregulated in the tumors. CONCLUSIONS Ceramides mediate the anti-cancer effects of different chemotherapeutic drugs and restoration of ceramide levels was effective against HCC. High ceramide levels in the tumors makes the DEN injected mice an unsuitable model to study therapies targeting ceramide metabolism. This model is useful for investigating how tumors evade the cytotoxic effects of ceramides.
Collapse
Affiliation(s)
- Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| |
Collapse
|
29
|
Fan GH, Wei RL, Wei XY, Zhang CZ, Qi ZT, Xie HY, Zheng SS, Xu X. Key factors and potential drug combinations of nonalcoholic steatohepatitis: Bioinformatic analysis and experimental validation-based study. Hepatobiliary Pancreat Dis Int 2021; 20:433-451. [PMID: 34233850 DOI: 10.1016/j.hbpd.2021.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/11/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease and its advanced stage, nonalcoholic steatohepatitis (NASH), are the major cause of hepatocellular carcinoma (HCC) and other end-stage liver disease. However, the potential mechanism and therapeutic strategies have not been clarified. This study aimed to identify potential roles of miRNA/mRNA axis in the pathogenesis and drug combinations in the treatment of NASH. METHODS Microarray GSE59045 and GSE48452 were downloaded from the Gene Expression Omnibus and analyzed using R. Then we obtained differentially expressed genes (DE-genes). DAVID database was used for Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway analysis. Protein-protein interaction (PPI) networks were used for the identification of hub genes. We found upstream regulators of hub genes using miRTarBase. The expression and correlation of key miRNA and its targets were detected by qPCR. Drug Pair Seeker was employed to predict drug combinations against NASH. The expression of miRNA and hub genes in HCC was identified in the Cancer Genome Atlas database and Human Protein Atlas database. RESULTS Ninety-four DE-genes were accessed. GO and KEGG analysis showed that these predicted genes were linked to lipid metabolism. Eleven genes were identified as hub genes in PPI networks, and they were highly expressed in cells with vigorous lipid metabolism. hsa-miR-335-5p was the upstream regulator of 9 genes in the 11 hub genes, and it was identified as a key miRNA. The hub genes were highly expressed in NASH models, while hsa-miR-335-5p was lowly expressed. The correlation of miRNA-mRNA was established by qPCR. Functional verification indicated that hsa-miR-335-5p had inhibitory effect on the development of NASH. Finally, drug combinations were predicted and the expression of miRNA and hub genes in HCC was identified. CONCLUSIONS In the study, potential miRNA-mRNA pathways related to NASH were identified. Targeting these pathways may be novel strategies against NASH.
Collapse
Affiliation(s)
- Guang-Han Fan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Rong-Li Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xu-Yong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Chen-Zhi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhe-Tuo Qi
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hai-Yang Xie
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Shu-Sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou 310000, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
30
|
Wang H, Chen X, Calvisi DF. Hepatocellular carcinoma (HCC): the most promising therapeutic targets in the preclinical arena based on tumor biology characteristics. Expert Opin Ther Targets 2021; 25:645-658. [PMID: 34477018 DOI: 10.1080/14728222.2021.1976142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION : Hepatocellular carcinoma (HCC) is a malignant liver tumor characterized by high molecular heterogeneity, which has hampered the development of effective targeted therapies severely. Recent experimental data have unraveled novel promising targets for HCC treatment. AREAS COVERED : Eligible articles were retrieved from PubMed and Web of Science databases up to July 2021. This review summarizes the established targeted therapies for advanced HCC, focusing on the strategies to overcome drug resistance and the search for combinational treatments. In addition, conventional biomarkers holding the promises for HCC treatments and novel therapeutic targets from the research field are discussed. EXPERT OPINION : HCC is a molecularly complex disease, with several and distinct pathways playing critical roles in different tumor subtypes. Experimental models recapitulating the features of each tumor subset would be highly beneficial to design novel and more effective therapies against this disease. Furthermore, a deeper understanding of combinatorial drug synergism and the role of the tumor microenvironment in HCC will lead to improved therapeutic outcomes.
Collapse
Affiliation(s)
- Haichuan Wang
- Liver Transplantation Division, Department of Liver Surgery and Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
31
|
Shang R, Song X, Wang P, Zhou Y, Lu X, Wang J, Xu M, Chen X, Utpatel K, Che L, Liang B, Cigliano A, Evert M, Calvisi DF, Chen X. Cabozantinib-based combination therapy for the treatment of hepatocellular carcinoma. Gut 2021; 70:1746-1757. [PMID: 33144318 PMCID: PMC8089119 DOI: 10.1136/gutjnl-2020-320716] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with limited treatment options. Cabozantinib, an orally bioavailable multikinase inhibitor is now approved by Food and Drug Administration (FDA) for HCC patients. We evaluated the therapeutic efficacy of cabozantinib, either alone or in combination, in vitro and in vivo. DESIGN Human HCC cell lines and HCC mouse models were used to assess the therapeutic efficacy and targeted molecular pathways of cabozantinib, either alone or in combination with the pan-mTOR inhibitor MLN0128 or the checkpoint inhibitor anti-PD-L1 antibody. RESULTS Cabozantinib treatment led to stable disease in c-Met/β-catenin and Akt/c-Met mouse HCC while possessing limited efficacy on Akt/Ras and c-Myc liver tumours. Importantly, cabozantinib effectively inhibited c-MET and ERK activity, leading to decreased PKM2 and increased p21 expression in HCC cells and in c-Met/β-catenin and Akt/c-Met HCC. However, cabozantinib was ineffective in inhibiting the Akt/mTOR cascade. Intriguingly, a strong inhibition of angiogenesis by cabozantinib occurred regardless of the oncogenic drivers. However, cabozantinib had limited impact on other tumour microenvironment parameters, including tumour infiltrating T cells, and did not induce programmed death-ligand 1 (PD-L1) expression. Combining cabozantinib with MLN0128 led to tumour regression in c-Met/β-catenin mice. In contrast, combined treatment with cabozantinib and the checkpoint inhibitor anti-PD-L1 antibody did not provide any additional therapeutic benefit in the four mouse HCC models tested. CONCLUSION c-MET/ERK/p21/PKM2 cascade and VEGFR2-induced angiogenesis are the primary targets of cabozantinib in HCC treatment. Combination therapies with cabozantinib and mTOR inhibitors may be effective against human HCC.
Collapse
Affiliation(s)
- Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, Xian, Shaanxi, China,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of General Surgery, The 910 Hospital, Quanzhou, Fujian, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Pan Wang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Collaborative Innovation Center for Agricultural Product Processing and Nutrition & Health, Beijing Vegetable Research Center, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yi Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinjun Lu
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Hepatic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jingxiao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, Beijing, China
| | - Meng Xu
- Department of General Surgery, The Second Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyan Chen
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,R&D Center, Legend Biotech USA Inc, Piscataway, New Jersey, USA
| | - Binyong Liang
- Hepatic Surgery Center, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Antonio Cigliano
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
32
|
Reducing FASN expression sensitizes acute myeloid leukemia cells to differentiation therapy. Cell Death Differ 2021; 28:2465-2481. [PMID: 33742137 PMCID: PMC8329134 DOI: 10.1038/s41418-021-00768-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 02/14/2021] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Fatty acid synthase (FASN) is the only human lipogenic enzyme available for de novo fatty acid synthesis and is often highly expressed in cancer cells. We found that FASN mRNA levels were significantly higher in acute myeloid leukemia (AML) patients than in healthy granulocytes or CD34+ hematopoietic progenitors. Accordingly, FASN levels decreased during all-trans retinoic acid (ATRA)-mediated granulocytic differentiation of acute promyelocytic leukemia (APL) cells, partially via autophagic degradation. Furthermore, our data suggest that inhibition of FASN expression levels using RNAi or (-)-epigallocatechin-3-gallate (EGCG) accelerated the differentiation of APL cell lines and significantly re-sensitized ATRA refractory non-APL AML cells. FASN reduction promoted translocation of transcription factor EB (TFEB) to the nucleus, paralleled by activation of CLEAR network genes and lysosomal biogenesis. Together, our data demonstrate that inhibition of FASN expression in combination with ATRA treatment facilitates granulocytic differentiation of APL cells and may extend differentiation therapy to non-APL AML cells.
Collapse
|
33
|
Goodwin JS, Tsai LL, Mwin D, Coutinho de Souza P, Dialani S, Moon JT, Zhang Z, Grant AK, Ahmed M. In vivo detection of distal tumor glycolytic flux stimulated by hepatic ablation in a breast cancer model using hyperpolarized 13C MRI. Magn Reson Imaging 2021; 80:90-97. [PMID: 33901585 DOI: 10.1016/j.mri.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Hepatic thermal ablation therapy can result in c-Met-mediated off-target stimulation of distal tumor growth. The purpose of this study was to determine if a similar effect on tumor metabolism could be detected in vivo with hyperpolarized 13C MRI. MATERIALS AND METHODS In this prospective study, female Fisher rats (n = 28, 120-150 g) were implanted with R3230 rat breast adenocarcinoma cells and assigned to either: sham surgery, hepatic radiofrequency ablation (RFA), or hepatic RFA + adjuvant c-Met inhibition with PHA-665752 (RFA + PHA). PHA-665752 was administered at 0.83 mg/kg at 24 h post-RFA. Tumor growth was measured daily. MRI was performed 24 h before and 72 h after treatment on 14 rats, and the conversion of 13C-pyruvate into 13C-lactate within each tumor was quantified as lactate:pyruvate ratio (LPR). Comparisons of tumor growth and LPR were performed using paired and unpaired t-tests. RESULTS Hepatic RFA alone resulted in increased growth of the distant tumor compared to sham treatment (0.50 ± 0.13 mm/day versus 0.11 ± 0.07 mm/day; p < 0.001), whereas RFA + PHA (0.06 ± 0.13 mm/day) resulted in no significant change from sham treatment (p = 0.28). A significant increase in LPR was seen following hepatic RFA (+0.016 ± 0.010, p = 0.02), while LPR was unchanged for sham treatment (-0.048 ± 0.051, p = 0.10) or RFA + PHA (0.003 ± 0.041, p = 0.90). CONCLUSION In vivo hyperpolarized 13C MRI can detect hepatic RFA-induced increase in lactate flux within a distant R3230 tumor, which correlates with increased tumor growth. Adjuvant inhibition of c-Met suppresses these off-target effects, supporting a role for the HGF/c-Met signaling axis in these tumorigenic responses.
Collapse
Affiliation(s)
- J Scott Goodwin
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; UT Austin Dell Medical School Transitional Program, 1400 IH-35, CEC 2.404, Austin, TX 78701, USA
| | - Leo L Tsai
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA.
| | - David Mwin
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Patricia Coutinho de Souza
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Genmab, 777 Scudders Mill Rd, Plainsboro, NJ 08536, USA
| | - Svayam Dialani
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Northwestern University, 2145 Ridge Ave, Evanston, IL 60201, USA
| | - John T Moon
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Division of Interventional Radiology, Emory University School of Medicine, Emory University Hospital, 1364 Clifton Road NE, Atlanta, GA 30322, USA
| | - Zheng Zhang
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Aaron K Grant
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Muneeb Ahmed
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
34
|
Shojaei-Zarghani S, Rafraf M, Yari-Khosroushahi A. Theanine and cancer: A systematic review of the literature. Phytother Res 2021; 35:4782-4794. [PMID: 33891786 DOI: 10.1002/ptr.7110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/23/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
A growing literature indicates several health benefits of theanine, a major nonprotein derivative amino acid special to tea, and a nonedible mushroom. This study aimed to systematically review the scientific evidence regarding the anticarcinogen and anticancer effects of natural theanine. A systematic search for the relevant articles published until January 2021 on MEDLINE, Scopus, and Web of Knowledge was conducted. Out of 377 initial records, 14 in vitro, ex vivo, and in vivo studies met our inclusion criteria. Most of the included in vitro and ex vivo studies reported beneficial effects of theanine on the proliferation, apoptosis, metastasis, migration, and invasion in various cancer cell lines. The in vivo studies also supported the potential impacts of theanine on cancer incidence or progression. Theanine exerted its anticancer function by inhibiting EGFR, VEGFR, Met, and Akt/mTOR, JAK2/STAT3, and ERK/NFκB pathways, as well as activating the intrinsic apoptosis pathway and caspase-independent programmed cell death. In conclusion, the results indicated moderate apoptotic, antimetastatic, antimigration, and anti-invasion effects, along with the mild antiproliferative influence of theanine on cancer. Further studies are necessary to ascertain the effectiveness of theanine on the prevention and suppression of cancer and shed light upon the attributable mechanisms in the in vivo condition.
Collapse
Affiliation(s)
- Sara Shojaei-Zarghani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.,Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Rafraf
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari-Khosroushahi
- Drug Applied Research Center, Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Overexpression of Human Syndecan-1 Protects against the Diethylnitrosamine-Induced Hepatocarcinogenesis in Mice. Cancers (Basel) 2021; 13:cancers13071548. [PMID: 33801718 PMCID: PMC8037268 DOI: 10.3390/cancers13071548] [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: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Syndecan-1 is a Janus-faced proteoglycan: depending on the type of cancer, it can promote or inhibit the development of tumors. Our previous in vitro experiments revealed that transfection of human syndecan-1 (hSDC1) into hepatoma cells, initiating hepatocyte-like differentiation. To further confirm the antitumor action of hSDC1 in the context of liver carcinogenesis, mice transgenic for albumin promoter-driven hSDC1 were created with exclusive expression of hSDC1 in the liver. Indeed, hSDC1 interfered with the development of liver cancer in diethylnitrosamine (DEN)-induced hepatocarcinogenesis experiments. The mechanism was found to be related to lipid metabolism that plays an important role in the induction of nonalcoholic liver cirrhosis. Nonalcoholic fatty liver disease is known to promote the development of cancer; therefore, the oncoprotective effect of hSDC1 may be mediated by a beneficial modulation of lipid metabolism. Abstract Although syndecan-1 (SDC1) is known to be dysregulated in various cancer types, its implication in tumorigenesis is poorly understood. Its effect may be detrimental or protective depending on the type of cancer. Our previous data suggest that SDC1 is protective against hepatocarcinogenesis. To further verify this notion, human SDC1 transgenic (hSDC1+/+) mice were generated that expressed hSDC1 specifically in the liver under the control of the albumin promoter. Hepatocarcinogenesis was induced by a single dose of diethylnitrosamine (DEN) at an age of 15 days after birth, which resulted in tumors without cirrhosis in wild-type and hSDC1+/+ mice. At the experimental endpoint, livers were examined macroscopically and histologically, as well as by immunohistochemistry, Western blot, receptor tyrosine kinase array, phosphoprotein array, and proteomic analysis. Liver-specific overexpression of hSDC1 resulted in an approximately six month delay in tumor formation via the promotion of SDC1 shedding, downregulation of lipid metabolism, inhibition of the mTOR and the β-catenin pathways, and activation of the Foxo1 and p53 transcription factors that lead to the upregulation of the cell cycle inhibitors p21 and p27. Furthermore, both of them are implicated in the regulation of intermediary metabolism. Proteomic analysis showed enhanced lipid metabolism, activation of motor proteins, and loss of mitochondrial electron transport proteins as promoters of cancer in wild-type tumors, inhibited in the hSDC1+/+ livers. These complex mechanisms mimic the characteristics of nonalcoholic steatohepatitis (NASH) induced human liver cancer successfully delayed by syndecan-1.
Collapse
|
36
|
Luo X, Zheng E, Wei L, Zeng H, Qin H, Zhang X, Liao M, Chen L, Zhao L, Ruan XZ, Yang P, Chen Y. The fatty acid receptor CD36 promotes HCC progression through activating Src/PI3K/AKT axis-dependent aerobic glycolysis. Cell Death Dis 2021; 12:328. [PMID: 33771982 PMCID: PMC7997878 DOI: 10.1038/s41419-021-03596-w] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/25/2022]
Abstract
Metabolic reprogramming is a new hallmark of cancer but it remains poorly defined in hepatocellular carcinogenesis (HCC). The fatty acid receptor CD36 is associated with both lipid and glucose metabolism in the liver. However, the role of CD36 in metabolic reprogramming in the progression of HCC still remains to be elucidated. In the present study, we found that CD36 is highly expressed in human HCC as compared with non-tumor hepatic tissue. CD36 overexpression promoted the proliferation, migration, invasion, and in vivo tumor growth of HCC cells, whereas silencing CD36 had the opposite effects. By analysis of cell metabolic phenotype, CD36 expression showed a positive association with extracellular acidification rate, a measure of glycolysis, instead of oxygen consumption rate. Further experiments verified that overexpression of CD36 resulted in increased glycolysis flux and lactic acid production. Mechanistically, CD36 induced mTOR-mediated oncogenic glycolysis via activation of Src/PI3K/AKT signaling axis. Pretreatment of HCC cells with PI3K/AKT/mTOR inhibitors largely blocked the tumor-promoting effect of CD36. Our findings suggest that CD36 exerts a stimulatory effect on HCC growth and metastasis, through mediating aerobic glycolysis by the Src/PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaoqing Luo
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Enze Zheng
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Li Wei
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Han Zeng
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Qin
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoyu Zhang
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Meng Liao
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Lin Chen
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Lei Zhao
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Xiong Z Ruan
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.,John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, University College London, Royal Free Campus, London, NW3 2PF, UK
| | - Ping Yang
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Yaxi Chen
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
37
|
Hu X, Zhang J, Deng L, Hu H, Hu J, Zheng G. Galactose-Modified PH-Sensitive Niosomes for Controlled Release and Hepatocellular Carcinoma Target Delivery of Tanshinone IIA. AAPS PharmSciTech 2021; 22:96. [PMID: 33694067 PMCID: PMC7946689 DOI: 10.1208/s12249-021-01973-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/21/2021] [Indexed: 12/11/2022] Open
Abstract
Increasing the drug tumor-specific accumulation and controlling their release is considered one of the most effective ways to increase the efficacy of drugs. Here, we developed a vesicle system that can target hepatoma and release drugs rapidly within tumor cells. This non-ionic surfactant vesicle is biodegradable. Galactosylated stearate has been used to glycosylate the vesicles to achieve liver targeting; replacement of a portion (Chol:CHEMS = 1:1) of cholesterol by cholesteryl hemisuccinate (CHEMS) allows for a rapid release of drugs in an acidic environment. In vitro release experiments confirmed that galactose-modified pH-sensitive niosomes loaded with tanshinone IIA had excellent drug release performance in acid medium. In vitro experiments using ovarian cancer cells (A2780), colon cancer cells (HCT8), and hepatoma cell (Huh7, HepG2) confirmed that the preparation had specific targeting ability to hepatoma cells compared with free drugs, and this ability was dependent on the galactose content. Furthermore, the preparation also had a more substantial inhibitory effect on tumor cells, and subsequent apoptosis assays and cell cycle analyses further confirmed its enhanced anti-tumor effect. Results of pharmacokinetic experiments confirmed that the vesicle system could significantly extend the blood circulation time of tanshinone IIA, and the larger area under the curve indicated that the preparation had a better drug effect. Thus, the results of biodistribution experiments confirmed the in vivo liver targeting ability of this preparation. Niosomes designed in this manner are expected to be a safe and effective drug delivery system for liver cancer therapy.
Collapse
|
38
|
Tang X, Zheng Y, Jiao D, Chen J, Liu X, Xiong S, Chen Q. Anlotinib Inhibits Cell Proliferation, Migration and Invasion via Suppression of c-Met Pathway and Activation of ERK1/2 Pathway in H446 Cells. Anticancer Agents Med Chem 2021; 21:747-755. [PMID: 32682383 DOI: 10.2174/1871520620666200718235748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Small Cell Lung Cancer (SCLC) represents the most aggressive pulmonary neoplasm and is often diagnosed at late stage with limited survival, despite combined chemotherapies. The purpose of this study was to investigate the effect of anlotinib on SCLC and the potential molecular mechanisms. METHODS Cell viability was assessed by CCK-8 assay to determine the adequate concentration of anlotinib. Then, effects of anlotinib on cell apoptosis, cell cycle distribution, migration and invasion were analyzed by flow cytometry, PI staining, wound healing assay and transwell assay, respectively. The protein expression of c-met and ERK1/2 pathways in H446 cells were assessed by western blot analysis. RESULTS In this study, we found that anlotinib significantly reduced the cell viability of H446 cells, induced G2/M cell cycle arrest and decreased invasion and migration of H446 cells. Futhermore, we also found that anlotinib could suppress c-met signal transduction and activate the ERK1/2 pathway in H446 cells. More importantly, c-met was involved in the effects of anlotinib on migration and invasion in H446 cells. CONCLUSION Taken together, our results demonstrated that anlotinib was a potential anticancer agent that inhibited cell proliferation, migration and invasion via suppression of the c-met pathway and activation of the ERK1/2 pathway in H446 cells.
Collapse
Affiliation(s)
- Xiali Tang
- Department of Pulmonary and Critical Care Medicine, The 903rd Hospital of PLA, Hangzhou, Zhejiang, China
| | - Ying Zheng
- Department of Pulmonary and Critical Care Medicine, The 903rd Hospital of PLA, Hangzhou, Zhejiang, China
| | - Demin Jiao
- Department of Pulmonary and Critical Care Medicine, The 903rd Hospital of PLA, Hangzhou, Zhejiang, China
| | - Jun Chen
- Department of Pulmonary and Critical Care Medicine, The 903rd Hospital of PLA, Hangzhou, Zhejiang, China
| | - Xibang Liu
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shan Xiong
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong 250062, China
| | - Qingyong Chen
- Department of Pulmonary and Critical Care Medicine, The 903rd Hospital of PLA, Hangzhou, Zhejiang, China
| |
Collapse
|
39
|
Wang Q, Liu MJ, Bu J, Deng JL, Jiang BY, Jiang LD, He XJ. miR-485-3p regulated by MALAT1 inhibits osteosarcoma glycolysis and metastasis by directly suppressing c-MET and AKT3/mTOR signalling. Life Sci 2021; 268:118925. [PMID: 33358903 DOI: 10.1016/j.lfs.2020.118925] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022]
Abstract
AIMS Osteosarcoma (OS) is an extremely malignant bone cancer with high incidence and rapid progression. This study aims to investigate the role and underlying mechanisms of MALAT1 and miR-485-3p in OS. MATERIALS AND METHODS qRT-PCR and Western blotting were utilized to measure the levels of miR-485-3p, MALAT1, c-MET, AKT3, p-mTOR, mTOR, glycolysis-related proteins or migration-related proteins. Colony formation and transwell assay were used to test the roles of miR-485-3p, MALAT1, c-MET and AKT3 in cancer cell proliferation, migration and invasion. Dual luciferase assay was used to validate the interactions of miR-485-3p/c-MET, miR-485-3p/AKT3, and MALAT1/miR-485-3p. Glucose uptake assay and measurement of lactate production were employed to determine the glycolysis process. Mouse tumour xenograft model was used to determine the effect of shMALAT1 and miR-485-3p mimics on tumour growth and metastasis in vivo. KEY FINDINGS miR-485-3p was decreased while c-MET, AKT3, and MALAT1 were increased in human OS tissues and cells. miR-485-3p bound directly to c-MET and AKT3 mRNAs and repressed OS cell glycolysis, proliferation, migration, and invasion through decreasing glycolysis-related proteins and migration-related proteins via inhibiting c-MET and AKT3/mTOR pathway. In addition, MALAT1 interacted with miR-485-3p and disinhibited c-MET and AKT3/mTOR signalling. Knockdown MALAT1 or overexpression of miR-485-3p restrained OS tumour growth and lung metastasis in vivo. SIGNIFICANCE miR-485-3p suppresses OS glycolysis, proliferation, and metastasis via inhibiting c-MET and AKT3/mTOR signalling and MALAT1 acts as a sponge of miR-485-3p. MALAT1 and miR-485-3p may be the key regulators in OS progression, and potential molecular targets for future OS therapy.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital (University of South China Affiliated Changsha Central Hospital), Changsha 410004, Hunan Province, PR China
| | - Ming-Jiang Liu
- Department of Orthopaedics, Changsha Central Hospital (University of South China Affiliated Changsha Central Hospital), Changsha 410004, Hunan Province, PR China
| | - Jie Bu
- Orthopedics & Soft Tissue Department, Hunan Cancer Hospital, Changsha 410013, Hunan Province, PR China
| | - Jian-Liang Deng
- Department of Orthopaedics, Changsha Central Hospital (University of South China Affiliated Changsha Central Hospital), Changsha 410004, Hunan Province, PR China
| | - Bin-Yuan Jiang
- Central Laboratory, Changsha Central Hospital (University of South China Affiliated Changsha Central Hospital), Changsha 410004, Hunan Province, PR China
| | - Liang-Dong Jiang
- Department of Orthopaedics, Changsha Central Hospital (University of South China Affiliated Changsha Central Hospital), Changsha 410004, Hunan Province, PR China.
| | - Xiao-Jie He
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, Central South University, Changsha 410011, Hunan Province, PR China.
| |
Collapse
|
40
|
Wang H, Wang P, Xu M, Song X, Wu H, Evert M, Calvisi DF, Zeng Y, Chen X. Distinct functions of transforming growth factor-β signaling in c-MYC driven hepatocellular carcinoma initiation and progression. Cell Death Dis 2021; 12:200. [PMID: 33608500 PMCID: PMC7895828 DOI: 10.1038/s41419-021-03488-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/05/2023]
Abstract
Dysregulation of transforming growth factor-beta (TGFβ) signaling has been implicated in liver carcinogenesis with both tumor promoting and inhibiting activities. Activation of the c-MYC protooncogene is another critical genetic event in hepatocellular carcinoma (HCC). However, the precise functional crosstalk between c-MYC and TGFβ signaling pathways remains unclear. In the present investigation, we investigated the expression of TGFβ signaling in c-MYC amplified human HCC samples as well as the mechanisms whereby TGFβ modulates c-Myc driven hepatocarcinogenesis during initiation and progression. We found that several TGFβ target genes are overexpressed in human HCCs with c-MYC amplification. In vivo, activation of TGFβ1 impaired c-Myc murine HCC initiation, whereas inhibition of TGFβ pathway accelerated this process. In contrast, overexpression of TGFβ1 enhanced c-Myc HCC progression by promoting tumor cell metastasis. Mechanistically, activation of TGFβ promoted tumor microenvironment reprogramming rather than inducing epithelial-to-mesenchymal transition during HCC progression. Moreover, we identified PMEPA1 as a potential TGFβ1 target. Altogether, our data underline the divergent roles of TGFβ signaling during c-MYC induced HCC initiation and progression.
Collapse
Affiliation(s)
- Haichuan Wang
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Pan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
| | - Meng Xu
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
| | - Hong Wu
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Yong Zeng
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
41
|
Zhang Y, Liang B, Song X, Wang H, Evert M, Zhou Y, Calvisi DF, Tang L, Chen X. Loss of Apc Cooperates with Activated Oncogenes to Induce Liver Tumor Formation in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:930-946. [PMID: 33545120 DOI: 10.1016/j.ajpath.2021.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/03/2021] [Accepted: 01/14/2021] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) and hepatoblastoma are the major types of primary liver cancer in adulthood and childhood, respectively. Wnt/β-catenin signaling deregulation is one of the most frequent genetic events in hepatocarcinogenesis. APC regulator of WNT signaling pathway (APC) encodes an inhibitor of the Wnt cascade and acts as a tumor suppressor. Germline defects of the APC gene lead to familial adenomatous polyposis, and its somatic mutations occur in multiple tumor types. However, the contribution of APC in hepatocarcinogenesis remains unclear. Therefore, APC mutations and expression patterns were examined in human HCC and hepatoblastoma samples. Whether loss of Apc alone or in cooperation with other oncogenes triggers liver tumor development in vivo was also investigated. sgApc alone could not drive liver tumor formation, but synergized with activated oncogenes (YapS127A, TazS89A, and c-Met) to induce hepatocarcinogenesis. Mechanistically, Apc deletion induced the activation of β-catenin and its downstream targets in mouse liver tumors. Furthermore, Ctnnb1 ablation or TCF4-mediated transcription blockade completely prevented liver tumor formation, indicating the requirement of a functional β-catenin pathway for loss of Apc-driven hepatocarcinogenesis. This study shows that a subset of HCC patients with loss-of-function APC mutations might benefit from therapeutic strategies targeting the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China; Department of Bioengineering University of California, San Francisco, California
| | - Binyong Liang
- Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinhua Song
- Department of Bioengineering University of California, San Francisco, California
| | - Haichuan Wang
- Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Yi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Xin Chen
- Department of Bioengineering University of California, San Francisco, California; Department of Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
42
|
Lu X, Paliogiannis P, Calvisi DF, Chen X. Role of the Mammalian Target of Rapamycin Pathway in Liver Cancer: From Molecular Genetics to Targeted Therapies. Hepatology 2021; 73 Suppl 1:49-61. [PMID: 32394479 PMCID: PMC7655627 DOI: 10.1002/hep.31310] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022]
Abstract
Primary liver cancers, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), are highly lethal tumors, with high worldwide frequency and few effective treatment options. The mammalian target of rapamycin (mTOR) complex is a central regulator of cell growth and metabolism that integrates inputs from amino acids, nutrients, and extracellular signals. The mTOR protein is incorporated into two distinct complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Specifically, mTORC1 regulates protein synthesis, glucose and lipid metabolism, and autophagy, whereas mTORC2 promotes liver tumorigenesis through modulating the adenine/cytosine/guanine family of serine/threonine kinases, especially the protein kinase B proteins. In human HCC and iCCA samples, genomics analyses have revealed the frequent deregulation of the mTOR complexes. Both in vitro and in vivo studies have demonstrated the key role of mTORC1 and mTORC2 in liver-tumor development and progression. The first-generation mTOR inhibitors have been evaluated for effectiveness in liver-tumor treatment and have provided unsatisfactory results. Current research efforts are devoted to generating more efficacious mTOR inhibitors and identifying biomarkers for patient selection as well as for combination therapies. Here, we provide a comprehensive review of the mechanisms leading to a deregulated mTOR signaling cascade in liver cancers, the mechanisms whereby the mTOR pathway contributes to HCC and iCCA molecular pathogenesis, the therapeutic strategies, and the challenges to effectively inhibit mTOR in liver-cancer treatment. Conclusion: Deregulated mTOR signaling significantly contributes to HCC and iCCA molecular pathogenesis. mTOR inhibitors, presumably administered in association with other drugs, might be effective against subsets of human liver tumors.
Collapse
Affiliation(s)
- Xinjun Lu
- Department of Hepatic Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, United States
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, United States
| |
Collapse
|
43
|
Zhu M, Shi W, Chen K, Hu H, Ye X, Jiang Y. Pulsatilla saponin E suppresses viability, migration, invasion and promotes apoptosis of NSCLC cells through negatively regulating Akt/FASN pathway via inhibition of flotillin-2 in lipid raft. J Recept Signal Transduct Res 2020; 42:23-33. [PMID: 33243063 DOI: 10.1080/10799893.2020.1839764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Pulsatilla saponins from pulsatilla chinensis (Bunge) Regel have potential anti-tumor activities to certain human cancers. However, the roles of pulsatilla saponin E separated from pulsatilla saponins in non-small cell lung cancer (NSCLC) have not been reported. MATERIALS AND METHODS After treating NSCLC cells by pulsatilla saponin E at different concentrations, cell viability was measured by MTT and CCK-8 assays, and cell migration, invasion and apoptosis were detected by scratch wound-healing, transwell and flow cytometry assays. The contents of free cholesterol (FC) and total cholesterol (TC) were measured by high performance liquid chromatography (HPLC). The expression levels of flotillin-1, flotillin-2, Akt, fatty acid synthase (FASN) were detected by qRT-PCR and Western blot assays. RESULTS Pulsatilla saponin E suppressed viability, migration, invasion and promoted apoptosis of NSCLC cells followed by regulation of apoptosis-related proteins, reduced contents of FC and TC, and the expression levels of flotillin-1, flotillin-2, Akt, and FASN in a concentration-dependent manner. However, the inhibitory effects of pulsatilla saponin E on viability, migration, invasion of A549 cells and the expression levels of flotillin-1, flotillin-2, Akt, and FASN were reversed by flotillin-2 overexpression. CONCLUSIONS Our study revealed that pulsatilla saponin E suppressed migration, invasion and promoted apoptosis of NSCLC cells through negatively regulating Akt/FASN signaling pathway via the inhibition of flotillin-2 in lipid raft (LR). The current findings could be explored for developing a novel therapeutic drug for NSCLC treatment.
Collapse
Affiliation(s)
- Minghua Zhu
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, PR China
| | - Wei Shi
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, PR China
| | - Ke Chen
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, PR China
| | - Huiqun Hu
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, PR China
| | - Xiangqing Ye
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, PR China
| | - Yinfang Jiang
- Department of Cardiovascular Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| |
Collapse
|
44
|
Jia J, Che L, Cigliano A, Wang X, Peitta G, Tao J, Zhong S, Ribback S, Evert M, Chen X, Calvisi DF. Pivotal Role of Fatty Acid Synthase in c-MYC Driven Hepatocarcinogenesis. Int J Mol Sci 2020; 21:ijms21228467. [PMID: 33187130 PMCID: PMC7696085 DOI: 10.3390/ijms21228467] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/08/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly form of liver malignancy with limited treatment options. Amplification and/or overexpression of c-MYC is one of the most frequent genetic events in human HCC. The mammalian target of Rapamycin Complex 1 (mTORC1) is a major functional axis regulating various aspects of cellular growth and metabolism. Recently, we demonstrated that mTORC1 is necessary for c-Myc driven hepatocarcinogenesis as well as for HCC cell growth in vitro. Among the pivotal downstream effectors of mTORC1, upregulation of Fatty Acid Synthase (FASN) and its mediated de novo lipogenesis is a hallmark of human HCC. Here, we investigated the importance of FASN on c-Myc-dependent hepatocarcinogenesis using in vitro and in vivo approaches. In mouse and human HCC cells, we found that FASN suppression by either gene silencing or soluble inhibitors more effectively suppressed proliferation and induced apoptosis in the presence of high c-MYC expression. In c-Myc/Myeloid cell leukemia 1 (MCL1) mouse liver tumor lesions, FASN expression was markedly upregulated. Most importantly, genetic ablation of Fasn profoundly delayed (without abolishing) c-Myc/MCL1 induced HCC formation. Liver tumors developing in c-Myc/MCL1 mice depleted of Fasn showed a reduction in proliferation and an increase in apoptosis when compared with corresponding lesions from c-Myc/MCL1 mice with an intact Fasn gene. In human HCC samples, a significant correlation between the levels of c-MYC transcriptional activity and the expression of FASN mRNA was detected. Altogether, our study indicates that FASN is an important effector downstream of mTORC1 in c-MYC induced HCC. Targeting FASN may be helpful for the treatment of human HCC, at least in the tumor subset displaying c-MYC amplification or activation.
Collapse
Affiliation(s)
- Jiaoyuan Jia
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (J.J.); (L.C.); (J.T.); (S.Z.)
- Department of Oncology and Hematology, the Second Hospital, Jilin University, Changchun 130041, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (J.J.); (L.C.); (J.T.); (S.Z.)
- Legend Biotech USA R&D Center, Piscataway, NJ 08854, USA
| | - Antonio Cigliano
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (A.C.); (G.P.); (M.E.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Xue Wang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA;
| | - Graziella Peitta
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (A.C.); (G.P.); (M.E.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Junyan Tao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (J.J.); (L.C.); (J.T.); (S.Z.)
| | - Sheng Zhong
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (J.J.); (L.C.); (J.T.); (S.Z.)
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, 17475 Greifswald, Germany;
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (A.C.); (G.P.); (M.E.)
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143, USA; (J.J.); (L.C.); (J.T.); (S.Z.)
- Correspondence: (X.C.); (D.F.C.); Tel.: +1-415-502-6526 (X.C.); +39-079-228306 (D.F.C.)
| | - Diego F. Calvisi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
- Correspondence: (X.C.); (D.F.C.); Tel.: +1-415-502-6526 (X.C.); +39-079-228306 (D.F.C.)
| |
Collapse
|
45
|
Animal Models: A Useful Tool to Unveil Metabolic Changes in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12113318. [PMID: 33182674 PMCID: PMC7696782 DOI: 10.3390/cancers12113318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) represents an important health problem. At the moment, systemic therapies offered only modest clinical benefits. Thus, HCC represents a cancer extremely difficult to treat, and therapeutic breakthroughs are urgently needed. Metabolic reprogramming of neoplastic cells has been recognized as one of the core hallmarks of cancer. Experimental animal models represent an important tool that allows to investigate metabolic changes underlying HCC development and progression. In the present review, we characterize available rodent models of hepatocarcinogenesis. Moreover, we discuss the possibility that pharmacological targeting of Warburg metabolism may represent an additional tool to improve already available therapeutic approaches for HCC. Abstract Hepatocellular carcinoma (HCC) is one the most frequent and lethal human cancers. At present, no effective treatment for advanced HCC exist; therefore, the overall prognosis for HCC patients remains dismal. In recent years, a better knowledge of the signaling pathways involved in the regulation of HCC development and progression, has led to the identification of novel potential targets for therapeutic strategies. However, the obtained benefits from current therapeutic options are disappointing. Altered cancer metabolism has become a topic of renewed interest in the last decades, and it has been included among the core hallmarks of cancer. In the light of growing evidence for metabolic reprogramming in cancer, a wide number of experimental animal models have been exploited to study metabolic changes characterizing HCC development and progression and to further expand our knowledge of this tumor. In the present review, we discuss several rodent models of hepatocarcinogenesis, that contributed to elucidate the metabolic profile of HCC and the implications of these changes in modulating the aggressiveness of neoplastic cells. We also highlight the apparently contrasting results stemming from different animal models. Finally, we analyze whether these observations could be exploited to improve current therapeutic strategies for HCC.
Collapse
|
46
|
NASH, Fibrosis and Hepatocellular Carcinoma: Lipid Synthesis and Glutamine/Acetate Signaling. Int J Mol Sci 2020; 21:ijms21186799. [PMID: 32947972 PMCID: PMC7555727 DOI: 10.3390/ijms21186799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
Primary liver cancer is predicted to be the sixth most common cancer and the fourth leading cause of cancer mortality worldwide. Recent studies identified nonalcoholic fatty liver disease (NAFLD) as the underlying cause in 13-38.2% of patients with hepatocellular carcinoma unrelated to viral hepatitis and alcohol abuse. NAFLD progresses to nonalcoholic steatohepatitis (NASH), which increases the risk for the development of liver fibrosis, cirrhosis, and hepatocellular carcinoma. NAFLD is characterized by dysregulation of lipid metabolism. In addition, lipid metabolism is effected not only in NAFLD, but also in a broad range of chronic liver diseases and tumor development. Cancer cells manipulate a variety of metabolic pathways, including lipid metabolism, in order to build up their own cellular components. Identifying tumor dependencies on lipid metabolism would provide options for novel targeting strategies. This review article summarizes the research evidence on metabolic reprogramming and focuses on lipid metabolism in NAFLD, NASH, fibrosis, and cancer. As alternative routes of acetyl-CoA production for fatty acid synthesis, topics on glutamine and acetate metabolism are included. Further, studies on small compound inhibitors targeting lipid metabolism are discussed. Understanding reprogramming strategies in liver diseases, as well as the visualization of the metabolism reprogramming networks, could uncover novel therapeutic options.
Collapse
|
47
|
Lv S, Wang X, Bai X, Ning H, Li Y, Wen H, Lu W, Wang J. Mesenchymal epithelial transition factor regulates tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells through down-regulation of cyclin B1. Int J Biochem Cell Biol 2020; 128:105844. [PMID: 32882404 DOI: 10.1016/j.biocel.2020.105844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/25/2022]
Abstract
Tumor necrosis factor-related apoptotic induction ligand can induce cell apoptosis in various tumor cells. However, many cancer cells are resistant to tumor necrosis factor-related apoptotic induction ligand. Therefore, overcoming the tumor necrosis factor-related apoptotic induction ligand resistance makes it possible for tumor necrosis factor-related apoptotic induction ligand-based anti-cancer therapies. In this study, we took mesenchymal epithelial transition factor as the research target to study its role in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma. Mesenchymal epithelial transition factor gene has been proved to be an effective predictor of recurrence after hepatocellular carcinoma resection. The expression of mesenchymal epithelial transition factor and cyclin B1 were measured in tumor necrosis factor-related apoptotic induction ligand-resistant and non-resistant hepatocellular carcinoma tissues. Cyclin B1-knockdown and cyclin B1-overexpression hepatocellular carcinoma cells were treated with tumor necrosis factor-related apoptotic induction ligand; mesenchymal epithelial transition factor knockout, mesenchymal epithelial transition factor re-introduction and cyclin B1 restored in hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand were established. And MTT, bromodeoxyuridine, flow cytometry and western blotting were performed to evaluate the effect of mesenchymal epithelial transition factor and cyclin B1 on hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand. In addition, subcutaneous tumor transplantation in nude mice was conducted to access the effect of mesenchymal epithelial transition factor and cyclin B1 on tumor formation in vivo. In conclusion, cyclin B1 enhanced the cell growth and inhibited apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells. And mesenchymal epithelial transition factor promoted the cell growth and apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells by regulating cyclin B1. Therefore, mesenchymal epithelial transition factor regulates the cyclin B1 to regulate tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells. Our results suggest a novel molecular mechanism for regulating tumor necrosis factor-related apoptotic induction ligand resistance, which might be helpful to select drug targets in the treatment of liver cancer.
Collapse
Affiliation(s)
- Shuai Lv
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China.
| | - Xijuan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, China
| | - Xia Bai
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hanbing Ning
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Yingxia Li
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hongtao Wen
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Wenquan Lu
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Jingyun Wang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| |
Collapse
|
48
|
Kido T, Tabatabai ZL, Chen X, Lau YFC. Potential dual functional roles of the Y-linked RBMY in hepatocarcinogenesis. Cancer Sci 2020; 111:2987-2999. [PMID: 32473614 PMCID: PMC7419034 DOI: 10.1111/cas.14506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous liver cancer with significant male biases in incidence, disease progression, and outcomes. Previous studies have suggested that genes on the Y chromosome could be expressed and exert various male‐specific functions in the oncogenic processes. In particular, the RNA‐binding motif on the Y chromosome (RBMY) gene is frequently activated in HCC and postulated to promote hepatic oncogenesis in patients and animal models. In the present study, immunohistochemical analyses of HCC specimens and data mining of The Cancer Genome Atlas (TCGA) database revealed that high‐level RBMY expression is associated with poor prognosis and survival of the patients, suggesting that RBMY could possess oncogenic properties in HCC. To examine the immediate effect(s) of the RBMY overexpression in liver cancer cells, cell proliferation was analyzed on HuH‐7 and HepG2 cells. The results unexpectedly showed that RBMY overexpression inhibited cell proliferation in both cell lines as its immediate effect, which led to vast cell death in HuH‐7 cells. Transcriptome analysis showed that genes involved in various cell proliferative pathways, such as the RAS/RAF/MAP and PIP3/AKT signaling pathways, were downregulated by RBMY overexpression in HuH‐7 cells. Furthermore, in vivo analyses in a mouse liver cancer model using hydrodynamic tail vein injection of constitutively active AKT and RAS oncogenes showed that RBMY abolished HCC development. These findings support the notion that Y‐linked RBMY could serve dual tumor‐suppressing and tumor‐promoting functions, depending on the spatiotemporal and magnitude of its expression during oncogenic processes, thereby contributing to sexual dimorphisms in liver cancer.
Collapse
Affiliation(s)
- Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, San Francisco VA Health Care System, San Francisco, CA, USA.,Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Z Laura Tabatabai
- Department of Pathology, San Francisco VA Health Care System, San Francisco, CA, USA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.,Liver Center, University of California, San Francisco, San Francisco, CA, USA
| | - Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, San Francisco VA Health Care System, San Francisco, CA, USA.,Institute for Human Genetics, University of California, San Francisco, CA, USA.,Liver Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Xu D, Wang Z, Xia Y, Shao F, Xia W, Wei Y, Li X, Qian X, Lee JH, Du L, Zheng Y, Lv G, Leu JS, Wang H, Xing D, Liang T, Hung MC, Lu Z. The gluconeogenic enzyme PCK1 phosphorylates INSIG1/2 for lipogenesis. Nature 2020; 580:530-535. [PMID: 32322062 DOI: 10.1038/s41586-020-2183-2] [Citation(s) in RCA: 233] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/06/2020] [Indexed: 01/04/2023]
Abstract
Cancer cells increase lipogenesis for their proliferation and the activation of sterol regulatory element-binding proteins (SREBPs) has a central role in this process. SREBPs are inhibited by a complex composed of INSIG proteins, SREBP cleavage-activating protein (SCAP) and sterols in the endoplasmic reticulum. Regulation of the interaction between INSIG proteins and SCAP by sterol levels is critical for the dissociation of the SCAP-SREBP complex from the endoplasmic reticulum and the activation of SREBPs1,2. However, whether this protein interaction is regulated by a mechanism other than the abundance of sterol-and in particular, whether oncogenic signalling has a role-is unclear. Here we show that activated AKT in human hepatocellular carcinoma (HCC) cells phosphorylates cytosolic phosphoenolpyruvate carboxykinase 1 (PCK1), the rate-limiting enzyme in gluconeogenesis, at Ser90. Phosphorylated PCK1 translocates to the endoplasmic reticulum, where it uses GTP as a phosphate donor to phosphorylate INSIG1 at Ser207 and INSIG2 at Ser151. This phosphorylation reduces the binding of sterols to INSIG1 and INSIG2 and disrupts the interaction between INSIG proteins and SCAP, leading to the translocation of the SCAP-SREBP complex to the Golgi apparatus, the activation of SREBP proteins (SREBP1 or SREBP2) and the transcription of downstream lipogenesis-related genes, proliferation of tumour cells, and tumorigenesis in mice. In addition, phosphorylation of PCK1 at Ser90, INSIG1 at Ser207 and INSIG2 at Ser151 is not only positively correlated with the nuclear accumulation of SREBP1 in samples from patients with HCC, but also associated with poor HCC prognosis. Our findings highlight the importance of the protein kinase activity of PCK1 in the activation of SREBPs, lipogenesis and the development of HCC.
Collapse
Affiliation(s)
- Daqian Xu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Zheng Wang
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yan Xia
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fei Shao
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xinjian Li
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xu Qian
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jong-Ho Lee
- Department of Biological Sciences, Dong-A University, Busan, South Korea
| | - Linyong Du
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yanhua Zheng
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guishuai Lv
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Jia-Shiun Leu
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Hongyang Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, and Office of the President, China Medical University, Taichung, Taiwan.
| | - Zhimin Lu
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
50
|
Orlistat delays hepatocarcinogenesis in mice with hepatic co-activation of AKT and c-Met. Toxicol Appl Pharmacol 2020; 392:114918. [DOI: 10.1016/j.taap.2020.114918] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022]
|