1
|
Qi Q, Zhong R, Huang Y, Tang Y, Zhang XW, Liu C, Gao CF, Zhou L, Yu J, Wu LY. The RNA M5C methyltransferase NSUN2 promotes progression of hepatocellular carcinoma by enhancing PKM2-mediated glycolysis. Cell Death Dis 2025; 16:82. [PMID: 39924557 PMCID: PMC11808121 DOI: 10.1038/s41419-025-07414-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. The 5-methylcytosine (m5C) RNA methyltransferase NSUN2 is involved in cell proliferation and metastasis and is upregulated in a variety of cancers. However, the biological function and regulatory mechanism of NSUN2-mediated m5C modification have not been well studied in HCC. Our results showed that NSUN2 is upregulated and associated with poor prognosis in HCC patients after hepatectomy. NSUN2 overexpression significantly promoted HCC growth and metastasis, whereas NSUN2 knockdown had the opposite effect. m5C RNA immunoprecipitation sequencing (m5C-RIP-Seq) revealed that m5C hypermethylation correlates with mRNA overexpression and that NSUN2-mediated m5C hypermethylation promotes metabolism in HCC patients. Mechanistically, our data revealed that PKM2, a terminal enzyme in the glycolytic pathway, is a downstream target of NSUN2-mediated m5C modification. Specifically, NSUN2 could stabilize PKM2 mRNA by increasing the m5C level of the m5C site C773 in the 3'-UTR of PKM2 mRNA. In addition, rescue assays revealed that NSUN2 promotes HCC glycolysis and progression by upregulating PKM2. In conclusion, this study revealed that NSUN2-mediated m5C modification promotes glycolysis and the progression of hepatocellular carcinoma by stabilizing PKM2 mRNA, and provides a potential prognostic factor and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Qin Qi
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Zhong
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Wen Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang Liu
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chun-Fang Gao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Jian Yu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| | - Lu-Yi Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Abam F, Ghorbian S. The dual role of LncRNAs in hepatocellular carcinoma: Friend and foe. GASTROENTEROLOGY & ENDOSCOPY 2024; 2:186-195. [DOI: 10.1016/j.gande.2024.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
|
3
|
Kalaei Z, Shekarchi AA, Hojjat-Farsangi M, Jalali P, Jadidi-Niaragh F. The prognostic and therapeutic potential of vimentin in colorectal cancer. Mol Biol Rep 2024; 51:1027. [PMID: 39347868 DOI: 10.1007/s11033-024-09965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
Several cells and molecules in the tumor microenvironment have been introduced as effective factors in the prognosis and progression of colorectal cancer. As a key element of the intermediate filament family, vimentin is expressed by mesenchymal cells in a ubiquitous manner and contributes significantly to cellular integrity and stress resistance in colorectal cancer. Recent studies have shown that alterations in the expression patterns of intermediate filaments are significantly related to cancer progression, especially in phenotypes associated with cellular migration and invasion. In addition to its multiple biological roles, vimentin also has a substantial function in mediating the epithelial-mesenchymal transition. Therefore, evaluating vimentin as an effective factor involved in the prognosis of colorectal cancer and targeting it as a novel approach to cancer therapy have become one of the main goals of many researchers worldwide. In this article, we will review the various biological functions of vimentin, as well as its relationship with colorectal cancer with the aim of providing novel insights into its clinical importance in the prognosis and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Zahra Kalaei
- Department of Biology, Faculty of Natural Sciences, Tabriz University, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Di Capua DM, Shanahan W, Bourke M, Ramlaul N, Appel J, Canney A, Docherty NG, McGrath E, Ring E, Jones F, Boyle M, McCormack J, Gallagher T, Hoti E, Nolan N, Ryan JD, Houlihan DD, Fabre A. Tumour stemness and poor clinical outcomes in haemochromatosis patients with hepatocellular carcinoma. J Clin Pathol 2024; 77:669-675. [PMID: 37253536 PMCID: PMC11503110 DOI: 10.1136/jcp-2022-208679] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023]
Abstract
AIMS Patients with haemochromatosis (HFE) are known to have an increased risk of developing hepatocellular carcinoma (HCC). Available data are conflicting on whether such patients have poorer prognosis, and there is lack of data regarding the biology of HFE-HCC. We compared the course of HFE-HCC with a matched non-HFE-HCC control group and examined tumour characteristics using immunohistochemistry. METHODS In this tertiary care-based retrospective analysis, 12 patients with HFE and 34 patients with alcohol/non-alcoholic steatohepatitis who underwent initially successful curative HCC therapy with ablation or resection were identified from our registry. Time to tumour progression was compared. Resected liver tissue from a separate cohort of 11 matched patients with HFE-HCC and without HFE-HCC was assessed for the expression of progenitor and epithelial-mesenchymal transition markers using immunohistochemistry. RESULTS The median follow-up was 24.39 and 24.28 months for patients with HFE-HCC and those without HFE-HCC, respectively (p>0.05). The mean time to progression was shorter in the HFE group compared with the non-HFE group (12.87 months vs 17.78 months; HR 3.322, p<0.05). Patients with HFE-HCC also progressed to more advanced disease by the end of follow-up (p<0.05). Immunohistochemical analysis of matched HFE-HCC and non-HFE-HCC explants demonstrated increased expression of the cancer stem cell markers EpCAM (epithelial cell adhesion molecule) and EpCAM/SALL4 (spalt-like transcription factor 4) coexpression in HFE-HCC specimens (p<0.05). There was a high frequency of combined tumour subtypes within the HFE cohort. CONCLUSIONS This study demonstrates that the clinical course of patients with HFE-HCC is more aggressive and provides the first data indicating that their tumours have increased expression of progenitor markers. These findings suggest patients with HFE-HCC may need to be considered for transplant at an earlier stage.
Collapse
Affiliation(s)
| | | | - Michele Bourke
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Navneet Ramlaul
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Josh Appel
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Aoife Canney
- Histopathology, University Hospital Galway, Galway, Ireland
| | - Neil G Docherty
- University College Dublin School of Medicine, Dublin, Ireland
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland
| | - Erinn McGrath
- HIstopathology, St Vincent's University Hospital, Dublin, Ireland
| | - Eabha Ring
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Fiona Jones
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Marie Boyle
- Liver Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Janet McCormack
- Reseach Pathology Core, Conway Institute, University College Dublin, Dublin, Ireland
| | - Tom Gallagher
- University College Dublin School of Medicine, Dublin, Ireland
- Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland
| | - Emir Hoti
- University College Dublin School of Medicine, Dublin, Ireland
- Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland
| | - Niamh Nolan
- HIstopathology, St Vincent's University Hospital, Dublin, Ireland
| | - John D Ryan
- Hepatology Unit, Beaumont Hospital, Dublin, ireland
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Aurelie Fabre
- HIstopathology, St Vincent's University Hospital, Dublin, Ireland
- University College Dublin School of Medicine, Dublin, Ireland
- Reseach Pathology Core, Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Sato T, Shizu R, Baba R, Ooka A, Hosaka T, Kanno Y, Yoshinari K. Pregnane X receptor inhibits the transdifferentiation of hepatic stellate cells by down-regulating periostin expression. Biochem J 2024; 481:1173-1186. [PMID: 39171361 DOI: 10.1042/bcj20240172] [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: 04/14/2024] [Revised: 08/07/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor that plays a key role in drug metabolism. Recently, PXR was found to attenuate the development of liver cancer by suppressing epithelial-mesenchymal transition (EMT) in liver cancer cells in a mouse model of two-stage chemical carcinogenesis. To elucidate the role of PXR in the EMT of liver cancer cells, we focused on its role in hepatic stellate cells (HSCs), which are components of the tumor microenvironment in hepatocellular carcinoma (HCC). Human HSC-derived LX-2 cells stably expressed destabilization domain (DD)-fused human PXR (hPXR-LX2 cells). Human HCC-derived HepG2 cells were transfected with the EMT marker VIM promoter-regulated reporter plasmid and co-cultured with hPXR-LX2 cells or treated with hPXR-LX2-derived conditioned medium (CM). Co-culture or CM treatment increased reporter activity in HepG2 cells. This induction was attenuated upon PXR activation in hPXR-LX2 cells by treatment with the DD-stabilizing chemical Shield-1 and the human PXR ligand rifampicin. PXR activation in hPXR-LX2 cells exhibited inhibition of TGF-β1-induced transdifferentiation, supported by observations of morphological changes and protein or mRNA levels of the transdifferentiation markers COL1A1 and FN1. PXR activation in hPXR-LX2 cells also attenuated the mRNA levels of the key transdifferentiation factor, POSTN. Treatment of hPXR-LX2 cells with recombinant POSTN restored the PXR-mediated suppression of transdifferentiation. Reporter assays with the POSTN promoter showed that PXR inhibited the NF-κB-mediated transcription of POSTN. Consequently, PXR activation in HSCs is expected to inhibit transdifferentiation by down-regulating POSTN expression, thereby suppressing EMT of liver cancer cells.
Collapse
Affiliation(s)
- Takumi Sato
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Ryota Shizu
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Ryonosuke Baba
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Akira Ooka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Yuichiro Kanno
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| |
Collapse
|
6
|
Qin S, Gao K, Tian Z. Comprehensive characterization of differential glycation in hepatocellular carcinoma using tissue proteomics with stable isotopic labeling. Anal Bioanal Chem 2024; 416:4531-4541. [PMID: 38922433 DOI: 10.1007/s00216-024-05392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Glycation is a non-enzymatic posttranslational modification coming from the reaction between reducing sugars and free amino groups in proteins, where early glycation products (fructosyl-lysine, FL) and advanced glycation end products (AGEs) are formed. The occurrence of glycation and accumulation of AGEs have been closely associated with hepatocellular carcinoma (HCC). Here, we reported the characterization of differential glycation in HCC using tissue proteomics with stable isotopic labeling; early glycation-modified peptides were enriched with boronate affinity chromatography (BAC), and AGEs-modified peptides were fractionated with basic reversed-phase separation. By this integrated approach, 3717 and 1137 early and advanced glycated peptides corresponding to 4007 sites on 1484 proteins were identified with a false discovery rate (FDR) of no more than 1%. One hundred fifty-five sites were modified with both early and advanced end glycation products. Five early and 7 advanced glycated peptides were quantified to be differentially expressed in HCC tissues relative to paired adjacent tissues. Most (8 out of 10) of the proteins corresponding to the differential glycated peptides have previously been reported with dysregulation in HCC. The results together may deepen our knowledge of glycation as well as provide insights for therapeutics.
Collapse
Affiliation(s)
- Shanshan Qin
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China
| | - Ke Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhixin Tian
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
7
|
Li Y, Zhang P, Tang G, Zhong J, Wang Z, Zhu B. Lowering expression of Epsin-3 inhibits migration and invasion of lung adenocarcinoma cells by inhibiting the epithelial-mesenchymal transition. Sci Rep 2024; 14:17069. [PMID: 39048677 PMCID: PMC11269644 DOI: 10.1038/s41598-024-68193-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a genetic reprogramming that tumor cells utilize for metastasis. Epsin-3 (EPN3) is an endocytic adapter protein involved in clathrin-mediated endocytosis and had been previously linked to EMT in breast cancer and glioma metastasis. In this study, identified the role of epsin-3 in lung adenocarcinoma and metastasis and epsin-3 levels identified using an expression profile analysis of patient data indicated the protein was abnormally overexpressed in lung adenocarcinoma patients and this was directly linked to disease severity. Gene knockdowns of EPN3 in human adenocarcinoma cell line A549 and the non-small cell lung carcinoma cell line H1299 decreased the levels of mesenchymal markers, including vimentin (VIM), N-cadherin (NCAD) and embryonic transcription factors like zinc finger E-box binding homeobox 1(ZEB1), snail, and the key molecules of Wnt pathway such as β-catenin and resulted in increased expression of the epithelial marker E-cadherin (ECAD). Our data links EPN3 to the EMT process in lung cancer and inhibition of its expression reduced the metastatic and invasive ability of lung adenocarcinoma cells by inhibiting the EMT process.
Collapse
Affiliation(s)
- Yunhe Li
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pei Zhang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guoxu Tang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiahui Zhong
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenghong Wang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bing Zhu
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Vielmo A, Santos IR, Piva MM, Bandinelli MB, Pavarini SP, Panziera W, Driemeier D. Histological and immunohistochemical features of carcinomas with pulmonary involvement in cattle. Vet Pathol 2024; 61:179-189. [PMID: 37638494 DOI: 10.1177/03009858231192373] [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] [Indexed: 08/29/2023]
Abstract
Primary pulmonary neoplasms in cattle are rare. There are few studies on the pathological findings of these neoplasms in this species. This study aimed to describe the histological and immunohistochemical findings of primary and metastatic pulmonary carcinomas in cattle. We conducted a retrospective study of 19 cases of epithelial neoplasms with pulmonary involvement. Histologically, most of the neoplasms were classified as primary pulmonary neoplasms, including different adenocarcinoma subtypes (4/19, 21%) and adenosquamous carcinomas (3/19, 16%), followed by squamous cell carcinoma (6/19, 32%), metastatic uterine adenocarcinoma (4/19, 21%), metastatic hepatocellular carcinoma (1/19, 5%), and metastatic cholangiocarcinoma (1/19, 5%). By immunohistochemistry, all neoplasms were positive for pancytokeratin, and 4/19 (21%) were positive for vimentin. Primary pulmonary neoplasms had immunoreactivity for thyroid transcription factor-1 (6/7), while only 2 of these cases were positive for napsin A. All cases with squamous differentiation (9/9) had immunoreactivity for cytokeratin (CK) 5/6, while only 7 of these cases were positive for p40. CK20, CK7, and CK8/18 showed varied immunoreactivity in the primary and metastatic pulmonary carcinomas but were important markers to confirm the diagnosis of primary mucinous adenocarcinoma and metastatic cholangiocarcinoma. HepPar-1 was only positive in the metastatic hepatocellular carcinoma. The limited number of cases of metastatic uterine adenocarcinomas in this study precluded identification of a specific immunophenotype for this tumor. Immunohistochemistry proved to be an important tool to confirm the proper classification of these neoplasms.
Collapse
Affiliation(s)
- Andréia Vielmo
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | - Welden Panziera
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - David Driemeier
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
9
|
Keawvilai P, Kueanjinda P, Klomsing J, Palaga T. Coculturing liver cancer cells and monocytes in spheroids conditions monocytes to adopt tumor-associated macrophage phenotypes that favor tumor growth via cholesterol metabolism. J Leukoc Biol 2024; 115:344-357. [PMID: 37742062 DOI: 10.1093/jleuko/qiad114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023] Open
Abstract
Tumor-infiltrating immune cells and their crosstalk with cancer cells in the tumor microenvironment (TME) play a crucial role in shaping tumor progression and response to therapy. We utilized 3-dimensional liver cancer spheroids incorporating human primary monocytes to investigate the crosstalk between tumor-associated macrophages (TAMs) and Hepatocellular carcinoma (HCC) cells, HepG2 and PLC/PRF/5. Using multiplexed gene expression panels, the critical pathways involved in shaping primary human monocytes to adopt TAMs phenotypes were identified. The specific inhibitor for an identified pathway was used to explore its involvement in polarization of TAMs. In the cocultured spheroids comprising the human HCC cell lines, the infiltrating monocytes resembled protumor M2-like macrophage phenotypes. Gene expression panels of the infiltrating monocytes demonstrated that the upregulated genes were enriched in the cholesterol metabolism pathway. Cholesterol metabolism-related genes were upregulated together with the nuclear receptors, PPARG and LXR. When lysosomal acid lipase (LAL), the key enzyme necessary for the hydrolysis of lipoprotein, was inhibited, infiltrating monocytes in 3-dimensional spheroid coculture showed significantly decreased M2 marker and lipid uptake receptor expression as well as increased cellular lipid content, which indicated that cholesterol metabolism was important for conditioning the TAMs. Moreover, LAL inhibition reduced the spheroid growth and invasiveness of HCC cell lines. Small interfering RNA-mediated LAL silencing in monocytes yielded similar results upon spheroid coculture. These data indicated that liver cancer cells and infiltrating monocytes participate in crosstalk via cholesterol metabolism to condition monocytes toward TAMs, which favors tumor growth and survival, thereby promoting liver cancer progression.
Collapse
Affiliation(s)
- Pornlapat Keawvilai
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Patipark Kueanjinda
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Rama 4 Road, Pathumwan, Bangkok 10330, Thailand
| | - Jeerameth Klomsing
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Tanapat Palaga
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
10
|
Wang D, Zhang F, He J, Li C, Liu X, Zhang M, Huang H, Xiong Z, Duan H, Huang X, Wang M. Phosphorylated vimentin at Ser72 is associated with epithelial-mesenchymal transition in lupus nephritis. Int J Rheum Dis 2024; 27:e14990. [PMID: 38078507 DOI: 10.1111/1756-185x.14990] [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: 10/14/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 01/31/2024]
Abstract
OBJECTIVES To measure the expression of vimentin and its phosphorylated forms in lupus nephritis (LN) and explore their potential role in LN development. METHODS Lupus renal biopsies from LN patients and normal renal biopsies from kidney transplant donors were collected. The expression of vimentin and its phosphorylated forms (p-vimentin (Ser39, Ser56, Ser72, Ser83, and Tyr117)) were measured by Western blots and immunohistochemistry. To construct stable cell line that overexpress vimentin and its phosphorylated forms, an immortalized proximal tubule epithelial cell line (HK-2 cells) was utilized. The roles of vimentin and its phosphorylated forms on the migration of HK-2 cells were examined by transwell migration assay and wound healing analysis. RESULTS We first observed a significant upregulation of vimentin protein in TGFβ1-induced HK-2 cells. This finding was further confirmed in renal tissues obtained from LN patients and animal model. Interestingly, among the five phosphorylated forms of vimentin, only vimentin phosphorylated at Ser72 was upregulated in LN. Through the establishment of stable vimentin and its phosphorylated forms overexpression in HK-2 cells, we found that the overexpression of vimentin and its phosphorylated forms at Ser72 significantly enhances the cell migration. CONCLUSIONS Vimentin phosphorylated on Ser72 is important for renal epithelial cell migration, which would enhance the progression of vimentin-induced epithelial-mesenchymal transition during LN development.
Collapse
Affiliation(s)
- Daji Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Zhang
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing He
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Rheumatology and Immunology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Chuyi Li
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xingjiao Liu
- Department of Rheumatology and Immunology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Meng Zhang
- Department of Pathology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haihui Huang
- Department of Pathology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hongxia Duan
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Huang
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Meiying Wang
- Department of Rheumatology and Immunology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
11
|
Bopape M, Tiloke C, Ntsapi C. Moringa oleifera and Autophagy: Evidence from In Vitro Studies on Chaperone-Mediated Autophagy in HepG 2 Cancer Cells. Nutr Cancer 2023; 75:1822-1847. [PMID: 37850743 DOI: 10.1080/01635581.2023.2270215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer in Sub-Saharan African countries, including South Africa (SA). Given the limitations in current HCC therapeutics, there is an increasing need for alternative adjuvant therapeutic options. As such, several cell survival mechanisms, such as autophagy, have been identified as potential adjuvant therapeutic targets in HCC treatment. Of the three most established autophagic pathways, the upregulation of chaperone-mediated autophagy (CMA) has been extensively described in various cancer cells, including HCC cells. CMA promotes tumor growth and chemotherapeutic drug resistance, thus contributing to HCC tumorigenesis. Therefore, the modulation of CMA serves as a promising adjuvant target for current HCC therapeutic strategies. Phytochemical extracts found in the medicinal plant, Moringa oleifera (MO), have been shown to induce apoptosis in numerous cancer cells, including HCC. MO leaves have the greatest abundance of phytochemicals displaying anticancer potential. However, the potential interaction between the pro-apoptotic effects of MO aqueous leaf extract and the survival-promoting role of CMA in an in vitro model of HCC remains unclear. This review aims to summarize the latest findings on the role of CMA, and MO in the progression of HCC.
Collapse
Affiliation(s)
- Matlola Bopape
- Department of Basic Medical Sciences, University of the Free State, Bloemfontein, South Africa
| | - Charlette Tiloke
- Department of Basic Medical Sciences, University of the Free State, Bloemfontein, South Africa
| | - Claudia Ntsapi
- Department of Basic Medical Sciences, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
12
|
Liu B, Zheng X, Li J, Yao P, Guo P, Liu W, Zhao G. Atovaquone inhibits colorectal cancer metastasis by regulating PDGFRβ/NF-κB signaling pathway. BMC Cancer 2023; 23:1070. [PMID: 37932661 PMCID: PMC10629062 DOI: 10.1186/s12885-023-11585-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Colorectal cancer is a common malignant tumour. Invasive growth and distant metastasis are the main characteristics of its malignant biological behaviour, and they are also the primary factors leading to death in colon cancer patients. Atovaquone is an antimalarial drug, and its anticancer effect has recently been demonstrated in several cancer models in vitro and in vivo, but it has not been examined in the treatment of colorectal cancer. METHODS To elucidate the effect of atovaquone on colorectal cancer. We used RNA transcriptome sequencing, RT‒PCR and Western blot experiments to examine the expression of NF-κB (p-P65), EMT-related proteins and related inflammatory factors (IL1B, IL6, CCL20, CCL2, CXCL8, CXCL6, IL6ST, FAS, IL10 and IL1A). The effect of atovaquone on colorectal cancer metastasis was validated using an animal model of lung metastases. We further used transcriptome sequencing, the GCBI bioinformatics database and the STRING database to predict relevant target proteins. Furthermore, pathological sections were collected from relevant cases for immunohistochemical verification. RESULTS This study showed that atovaquone could inhibit colorectal cancer metastasis and invasion in vivo and in vitro, inhibit the expression of E-cadherin protein, and promote the protein expression of N-cadherin, vimentin, ZEB1, Snail and Slug. Atovaquone could inhibit EMT by inhibiting NF-κB (p-P65) and related inflammatory factors. Further bioinformatics analysis and verification showed that PDGFRβ was one of the targets of atovaquone. CONCLUSION In summary, atovaquone can inhibit the expression of NF-κB (p-P65) and related inflammatory factors by inhibiting the protein expression of p-PDGFRβ, thereby inhibiting colorectal cancer metastasis. Atovaquone may be a promising drug for the treatment of colorectal cancer metastasis.
Collapse
Affiliation(s)
- Bin Liu
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, 610051, Chengdu, Sichuan, China
| | - Xin Zheng
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, 610051, Chengdu, Sichuan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajun Li
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, 610051, Chengdu, Sichuan, China
| | - Peng Yao
- Department of Nephrology, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, 610051, Chengdu, Sichuan, China
| | - Peng Guo
- Chengdu Medical College, 610500, Chengdu, Sichuan, China
| | - Wei Liu
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, 610051, Chengdu, Sichuan, China
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, Sichuan, China.
| |
Collapse
|
13
|
Sato M, Tamauchi S, Yoshida K, Yoshihara M, Ikeda Y, Yoshikawa N, Kajiyama H. Unclear tumor border in magnetic resonance imaging as a prognostic factor of squamous cell cervical cancer. Sci Rep 2023; 13:15392. [PMID: 37717112 PMCID: PMC10505168 DOI: 10.1038/s41598-023-42787-7] [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: 03/05/2023] [Accepted: 09/14/2023] [Indexed: 09/18/2023] Open
Abstract
Magnetic resonance imaging (MRI) is used for pretreatment staging in cervical cancer. In the present study, we used pretreatment images to categorize operative cases into two groups and evaluated their prognosis. A total of 53 cervical cancer patients with squamous cell carcinoma who underwent radical hysterectomy were included in this study. Based on MRI, the patients were classified into two groups, namely clear and unclear tumor border. For each patient, the following characteristics were evaluated: overall survival; recurrence-free survival; lymph node metastasis; lymphovascular space invasion; and pathological findings, including immunohistochemical analysis of vimentin. The clear and unclear tumor border groups included 40 and 13 patients, respectively. Compared with the clear tumor border group, the unclear tumor border group was associated with higher incidence rates of recurrence (3/40 vs. 3/13, respectively), lymphovascular space invasion (24/40 vs. 13/13, respectively), lymph node metastasis (6/40 vs. 10/13, respectively), and positivity for vimentin (18/40 vs. 10/13, respectively). Despite the absence of significant difference in recurrence-free survival (p = 0.0847), the unclear tumor border group had a significantly poorer overall survival versus the clear tumor border group (p = 0.0062). According to MRI findings, an unclear tumor border in patients with squamous cell cervical cancer is linked to poorer prognosis, lymph node metastasis, and distant recurrence of metastasis.
Collapse
Affiliation(s)
- Mamiko Sato
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
14
|
Talukdar SN, McGregor B, Osan JK, Hur J, Mehedi M. Respiratory Syncytial Virus Infection Does Not Induce Epithelial-Mesenchymal Transition. J Virol 2023; 97:e0039423. [PMID: 37338373 PMCID: PMC10373540 DOI: 10.1128/jvi.00394-23] [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: 03/13/2023] [Accepted: 05/24/2023] [Indexed: 06/21/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection does not cause severe disease in most of us despite suffering from multiple RSV infections during our lives. However, infants, young children, older adults, and immunocompromised patients are unfortunately vulnerable to RSV-associated severe diseases. A recent study suggested that RSV infection causes cell expansion, resulting in bronchial wall thickening in vitro. Whether the virus-induced changes in the lung airway resemble epithelial-mesenchymal transition (EMT) is still unknown. Here, we report that RSV does not induce EMT in three different in vitro lung models: the epithelial A549 cell line, primary normal human bronchial epithelial cells, and pseudostratified airway epithelium. We found that RSV increases the cell surface area and perimeter in the infected airway epithelium, which is distinct from the effects of a potent EMT inducer, transforming growth factor β1 (TGF-β1), driving cell elongation-indicative of cell motility. A genome-wide transcriptome analysis revealed that both RSV and TGF-β1 have distinct modulation patterns of the transcriptome, which suggests that RSV-induced changes are distinct from EMT. IMPORTANCE We have previously shown that RSV infects ciliated cells on the apical side of the lung airway. RSV-induced cytoskeletal inflammation contributes to an uneven increase in the height of the airway epithelium, resembling noncanonical bronchial wall thickening. RSV infection changes epithelial cell morphology by modulating actin-protein 2/3 complex-driven actin polymerization. Therefore, it is prudent to investigate whether RSV-induced cell morphological changes contribute to EMT. Our data indicate that RSV does not induce EMT in at least three different epithelial in vitro models: an epithelial cell line, primary epithelial cells, and pseudostratified bronchial airway epithelium.
Collapse
Affiliation(s)
- Sattya N. Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Brett McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Jaspreet K. Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| |
Collapse
|
15
|
Talukdar SN, McGregor B, Osan JK, Hur J, Mehedi M. RSV infection does not induce EMT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532506. [PMID: 36993657 PMCID: PMC10055011 DOI: 10.1101/2023.03.13.532506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Respiratory syncytial virus (RSV) infection does not cause severe disease in most of us despite suffering from multiple RSV infections in our lives. However, infants, young children, older adults, and immunocompromised patients are unfortunately vulnerable to RSV-associated severe diseases. A recent study suggested that RSV infection causes cell expansion, resulting in bronchial wall thickening in vitro. Whether the virus-induced changes in the lung airway resemble epithelial-mesenchymal transition (EMT) is still unknown. Here, we report that RSV does not induce EMT in three different in vitro lung models: the epithelial A549 cell line, primary normal human bronchial epithelial cells, and pseudostratified airway epithelium. We found that RSV increases the cell surface area and perimeter in the infected airway epithelium, which is distinct from the effects of a potent EMT inducer, TGF-β1-driven cell elongation-indicative of cell motility. A genome-wide transcriptome analysis revealed that both RSV and TGF-β1 have distinct modulation patterns of the transcriptome, which suggests that RSV-induced changes are distinct from EMT.
Collapse
Affiliation(s)
- Sattya N. Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Brett McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Jaspreet K. Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| |
Collapse
|
16
|
van Tienderen GS, Rosmark O, Lieshout R, Willemse J, de Weijer F, Elowsson Rendin L, Westergren-Thorsson G, Doukas M, Groot Koerkamp B, van Royen ME, van der Laan LJ, Verstegen MM. Extracellular matrix drives tumor organoids toward desmoplastic matrix deposition and mesenchymal transition. Acta Biomater 2023; 158:115-131. [PMID: 36427688 DOI: 10.1016/j.actbio.2022.11.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/31/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022]
Abstract
Patient-derived tumor organoids have been established as promising tools for in vitro modelling of multiple tumors, including cholangiocarcinoma (CCA). However, organoids are commonly cultured in basement membrane extract (BME) which does not recapitulate the intricacies of the extracellular matrix (ECM). We combined CCA organoids (CCAOs) with native tumor and liver scaffolds, obtained by decellularization, to effectuate a model to study the interaction between epithelial tumor cells and their surrounding ECM. Decellularization resulted in removal of cells while preserving ECM structure and retaining important characteristics of the tissue origin, including stiffness and presence of desmoplasia. The transcriptome of CCAOs in a tumor scaffold much more resembled that of patient-paired CCA tissue in vivo compared to CCAOs cultured in BME or liver scaffolds. This was accompanied by an increase in chemoresistance to clinically-relevant chemotherapeutics. CCAOs in decellularized scaffolds revealed environment-dependent proliferation dynamics, driven by the occurrence of epithelial-mesenchymal transition. Furthermore, CCAOs initiated an environment-specific desmoplastic reaction by increasing production of multiple collagen types. In conclusion, convergence of organoid-based models with native ECM scaffolds will lead to better understanding of the in vivo tumor environment. STATEMENT OF SIGNIFICANCE: The extracellular matrix (ECM) influences various facets of tumor behavior. Understanding the exact role of the ECM in controlling tumor cell fate is pertinent to understand tumor progression and develop novel therapeutics. This is particularly the case for cholangiocarcinoma (CCA), whereby the ECM displays a distinct tumor environment, characterized by desmoplasia. However, current models to study the interaction between epithelial tumor cells and the environment are lacking. We have developed a fully patient-derived model encompassing CCA organoids (CCAOs) and human decellularized tumor and tumor-free liver ECM. The tumor ECM induced recapitulation of various aspects of CCA, including migration dynamics, transcriptome and proteome profiles, and chemoresistance. Lastly, we uncover that epithelial tumor cells contribute to matrix deposition, and that this phenomenon is dependent on the level of desmoplasia already present.
Collapse
Affiliation(s)
- Gilles S van Tienderen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Oskar Rosmark
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ruby Lieshout
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jorke Willemse
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Floor de Weijer
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Linda Elowsson Rendin
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Michail Doukas
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Luc Jw van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Monique Ma Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
17
|
Chiang CC, Yeh H, Lim SN, Lin WR. Transcriptome analysis creates a new era of precision medicine for managing recurrent hepatocellular carcinoma. World J Gastroenterol 2023; 29:780-799. [PMID: 36816628 PMCID: PMC9932421 DOI: 10.3748/wjg.v29.i5.780] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/23/2022] [Accepted: 01/10/2023] [Indexed: 02/06/2023] Open
Abstract
The high incidence of hepatocellular carcinoma (HCC) recurrence negatively impacts outcomes of patients treated with curative intent despite advances in surgical techniques and other locoregional liver-targeting therapies. Over the past few decades, the emergence of transcriptome analysis tools, including real-time quantitative reverse transcription PCR, microarrays, and RNA sequencing, has not only largely contributed to our knowledge about the pathogenesis of recurrent HCC but also led to the development of outcome prediction models based on differentially expressed gene signatures. In recent years, the single-cell RNA sequencing technique has revolutionized our ability to study the complicated crosstalk between cancer cells and the immune environment, which may benefit further investigations on the role of different immune cells in HCC recurrence and the identification of potential therapeutic targets. In the present article, we summarized the major findings yielded with these transcriptome methods within the framework of a causal model consisting of three domains: primary cancer cells; carcinogenic stimuli; and tumor microenvironment. We provided a comprehensive review of the insights that transcriptome analyses have provided into diagnostics, surveillance, and treatment of HCC recurrence.
Collapse
Affiliation(s)
- Chun-Cheng Chiang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, United States
| | - Hsuan Yeh
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Siew-Na Lim
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Wey-Ran Lin
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
18
|
Emerging role for R-loop formation in hepatocellular carcinoma. Genes Genomics 2023; 45:543-551. [PMID: 36635460 DOI: 10.1007/s13258-022-01360-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
The pathophysiological characteristics of hepatocellular carcinoma (HCC) is closely associated with genomic instability. Genomic instability has long been considered to be a hallmark of both human genetic disease and cancers. It is now well accepted that regulating R-loop formation to minimized levels is one of critical modulation to maintain genome integrity, and that improper regulation of R-loop metabolism causes genomic instability via DNA breakage, ultimately resulting in replicative senescence and even tumorigenesis. Given that R-loop is natural by-product formed during normal transcription condition, and that several types of cancer have defense mechanism against the genomic instability resulted from R-loop formation, modulating functional implication of proteins involved in the intrinsic and specific mechanisms of abnormal R-loop formation in cancers therefore could play an important part in appropriated therapeutic strategies for HCC cohorts. In this review, we highlight the latest understanding on how R-loops promote genomic instability and address how alterations in these pathways link to human HCC.
Collapse
|
19
|
3D microengineered vascularized tumor spheroids for drug delivery and efficacy testing. Acta Biomater 2022:S1742-7061(22)00665-1. [DOI: 10.1016/j.actbio.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/20/2022]
|
20
|
Park R, Park YI, Park Y, Lee S, So J, Park J. CTRP1 Knockout Attenuates Tumor Progression in A549 and HCT116 Cancer Cells. Cancers (Basel) 2022; 14:cancers14184495. [PMID: 36139655 PMCID: PMC9496675 DOI: 10.3390/cancers14184495] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary CTRP1 belongs to the C1q and TNF-related protein family, and we generated CTRP1 knockout cells to examine the role of CTRP1 in tumor progression. CTRP1 knockout attenuates cell growth, invasion and tumor growth in mice, suggesting that CTRP1 expression promotes tumor progression. Abstract C1q and TNF-related 1 (C1QTNF1/CTRP1) is an adiponectin-associated protein belonging to the C1q/TNF-related protein family. Recent studies have shown that the C1q and TNF-related protein (CTRP) family is involved in cancer progression; however, the specific role of CTRP1 in tumor progression has not yet been elucidated. To examine the role of CTRP1 in tumor progression, we generated CTRP1 knockout A549 and HCT116 cell lines, which reduced the expression levels of nuclear factor (NF)-κB-dependent and metastasis-promoting transcripts. We demonstrated that CTRP1 knockout inhibited the cell proliferation and invasion and tumor growth. Finally, database analysis showed that CTRP1 expression was upregulated in metastatic cancers and elevated levels of CTRP1 were associated with poor prognosis. These results suggest that CTRP1 expression contributes to NF-κB signaling and promotes tumor progression.
Collapse
Affiliation(s)
- Rackhyun Park
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
- Division of Biological Sciences, Yong-In University, Yongin 17092, Korea
| | - Yea-In Park
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
| | - Yeonjeong Park
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
| | - Siyun Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
| | - Jaeyeon So
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju 29493, Korea
- Correspondence: ; Tel.: +82-33-760-2560; Fax: +82-33-760-2183
| |
Collapse
|
21
|
Liang RP, Zhang XX, Zhao J, Lu QW, Zhu RT, Wang WJ, Li J, Bo K, Zhang CX, Sun YL. RING finger and WD repeat domain 3 regulates proliferation and metastasis through the Wnt/β-catenin signalling pathways in hepatocellular carcinoma. World J Gastroenterol 2022; 28:3435-3454. [PMID: 36158256 PMCID: PMC9346462 DOI: 10.3748/wjg.v28.i27.3435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/16/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) exhibits high invasiveness and mortality rates, and the molecular mechanisms of HCC have gained increasing research interest. The abnormal DNA damage response has long been recognized as one of the important factors for tumor occurrence and development. Recent studies have shown the potential of the protein RING finger and WD repeat domain 3 (RFWD3) that positively regulates p53 stability in response to DNA damage as a therapeutic target in cancers. AIM To investigate the relationship between HCC and RFWD3 in vitro and in vivo and explored the underlying molecular signalling transduction pathways. METHODS RFWD3 gene expression was analyzed in HCC tissues and adjacent normal tissues. Lentivirus was used to stably knockdown RFWD3 expression in HCC cell lines. After verifying the silencing efficiency, Celigo/cell cycle/apoptosis and MTT assays were used to evaluate cell proliferation and apoptosis. Subsequently, cell migration and invasion were assessed by wound healing and transwell assays. In addition, transduced cells were implanted subcutaneously and injected into the tail vein of nude mice to observe tumor growth and metastasis. Next, we used lentiviral-mediated rescue of RFWD3 shRNA to verify the phenotype. Finally, the microarray, ingenuity pathway analysis, and western blot analysis were used to analyze the regulatory network underlying HCC. RESULTS Compared with adjacent tissues, RFWD3 expression levels were significantly higher in clinical HCC tissues and correlated with tumor size and TNM stage (P < 0.05), which indicated a poor prognosis state. RFWD3 silencing in BEL-7404 and HCC-LM3 cells increased apoptosis, decreased growth, and inhibited the migration in shRNAi cells compared with those in shCtrl cells (P < 0.05). Furthermore, the in vitro results were supported by the findings of the in vivo experiments with the reduction of tumor cell invasion and migration. Moreover, the rescue of RFWD3 shRNAi resulted in the resumption of invasion and metastasis in HCC cell lines. Finally, gene expression profiling and subsequent experimental verification revealed that RFWD3 might influence the proliferation and metastasis of HCC via the Wnt/β-catenin signalling pathway. CONCLUSION We provide evidence for the expression and function of RFWD3 in HCC. RFWD3 affects the prognosis, proliferation, invasion, and metastasis of HCC by regulating the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Ruo-Peng Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiao-Xue Zhang
- Department of Physical Examination, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jie Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Qin-Wei Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rong-Tao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Wei-Jie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Kai Bo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chi-Xian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu-Ling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
22
|
Hausmann S, Geiser J, Valentini M. Mechanism of inhibition of bacterial RNA helicases by diazo dyes and implications for antimicrobial drug development. Biochem Pharmacol 2022; 204:115194. [DOI: 10.1016/j.bcp.2022.115194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
|
23
|
Chun KH. Molecular Targets and Signaling Pathways of microRNA-122 in Hepatocellular Carcinoma. Pharmaceutics 2022; 14:1380. [PMID: 35890276 PMCID: PMC9316959 DOI: 10.3390/pharmaceutics14071380] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading global causes of cancer mortality. MicroRNAs (miRNAs) are small interfering RNAs that alleviate the levels of protein expression by suppressing translation, inducing mRNA cleavage, and promoting mRNA degradation. miR-122 is the most abundant miRNA in the liver and is responsible for several liver-specific functions, including metabolism, cellular growth and differentiation, and hepatitis virus replication. Recent studies have shown that aberrant regulation of miR-122 is a key factor contributing to the development of HCC. In this review, the signaling pathways and the molecular targets of miR-122 involved in the progression of HCC have been summarized, and the importance of miR-122 in therapy has been discussed.
Collapse
Affiliation(s)
- Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Korea
| |
Collapse
|
24
|
Sheng J, Zhang J, Wang L, Tano V, Tang J, Wang X, Wu J, Song J, Zhao Y, Rong J, Cheng F, Wang J, Shen Y, Wen L, He J, Zhang H, Li T, Zhang Q, Bai X, Lu Z, Liang T. Topological analysis of hepatocellular carcinoma tumour microenvironment based on imaging mass cytometry reveals cellular neighbourhood regulated reversely by macrophages with different ontogeny. Gut 2022; 71:1176-1191. [PMID: 34253573 DOI: 10.1136/gutjnl-2021-324339] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/27/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) tumour microenvironment (TME) is highly complex with diverse cellular components organising into various functional units, cellular neighbourhoods (CNs). And we wanted to define CN of HCC while preserving the TME architecture, based on which, potential targets for novel immunotherapy could be identified. DESIGN A highly multiplexed imaging mass cytometry (IMC) panel was designed to simultaneously quantify 36 biomarkers of tissues from 134 patients with HCC and 7 healthy donors to generate 562 highly multiplexed histology images at single-cell resolution. Different function units were defined by topological analysis of TME. CN relevant to the patients' prognosis was identified as specific target for HCC therapy. Transgenic mouse models were used to validate the novel immunotherapy target for HCC. RESULTS Three major types of intratumour areas with distinct distribution patterns of tumorous, stromal and immune cells were identified. 22 cellular metaclusters and 16 CN were defined. CN composed of various types of cells formed regional function units and the regional immunity was regulated reversely by resident Kupffer cells and infiltrating macrophages with protumour and antitumour function, respectively. Depletion of Kupffer cells in mouse liver largely enhances the T cell response, reduces liver tumour growth and sensitises the tumour response to antiprogrammed cell death protein-1 treatment. CONCLUSION Our findings reveal for the first time the various topological function units of HCC TME, which also presents the largest depository of pathological landscape for HCC. This work highlights the potential of Kupffer cell-specific targeting rather than overall myeloid cell blocking as a novel immunotherapy for HCC treatment.
Collapse
Affiliation(s)
- Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang University Cancer Center, Zhejiang University, Hangzhou, People's Republic of China
| | - Junlei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Lin Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Vincent Tano
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jianghui Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Xun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jiangchao Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jinyuan Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yaxing Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jingxia Rong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Fei Cheng
- Pathology Department, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jianfeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Liang Wen
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Junjun He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Hui Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Taohong Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang University Cancer Center, Zhejiang University, Hangzhou, People's Republic of China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang University Cancer Center, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhimin Lu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang University Cancer Center, Zhejiang University, Hangzhou, People's Republic of China.,Institue of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China.,Zhejiang University Cancer Center, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
25
|
Surolia R, Antony VB. Pathophysiological Role of Vimentin Intermediate Filaments in Lung Diseases. Front Cell Dev Biol 2022; 10:872759. [PMID: 35573702 PMCID: PMC9096236 DOI: 10.3389/fcell.2022.872759] [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: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Vimentin intermediate filaments, a type III intermediate filament, are among the most widely studied IFs and are found abundantly in mesenchymal cells. Vimentin intermediate filaments localize primarily in the cytoplasm but can also be found on the cell surface and extracellular space. The cytoplasmic vimentin is well-recognized for its role in providing mechanical strength and regulating cell migration, adhesion, and division. The post-translationally modified forms of Vimentin intermediate filaments have several implications in host-pathogen interactions, cancers, and non-malignant lung diseases. This review will analyze the role of vimentin beyond just the epithelial to mesenchymal transition (EMT) marker highlighting its role as a regulator of host-pathogen interactions and signaling pathways for the pathophysiology of various lung diseases. In addition, we will also examine the clinically relevant anti-vimentin compounds and antibodies that could potentially interfere with the pathogenic role of Vimentin intermediate filaments in lung disease.
Collapse
Affiliation(s)
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
A Critical YAP in Malignancy of HCC Is Regulated by Evodiamine. Int J Mol Sci 2022; 23:ijms23031855. [PMID: 35163776 PMCID: PMC8837083 DOI: 10.3390/ijms23031855] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/18/2022] Open
Abstract
Liver cancer has relatively few early symptoms and is usually diagnosed in the advanced stage. Sorafenib is the only first-line anticancer drug approved by the Food and Drug Administration (FDA) for advanced HCC; however, its use is limited due to resistance. Therefore, the development of new drugs is essential to achieving customized treatment. Many studies have suggested that Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) is associated with metastasis and cancer formation and progression in various cancers. In the present study, YAP was overexpressed in various patient-derived hepatocarcinoma (HCC) tissues. In addition, this study examined whether evodiamine (which has anticancer effects) can inhibit YAP and, if so, modulate HCC. Evodiamine significantly reduced both the YAP level and cell growth of HCC in a dose-dependent manner. Biochemical analysis indicated mitochondria dysfunction-mediated apoptosis to be the cause of the reduction in HCC cell growth by evodiamine. YAP was overexpressed in metastatic HCC tissues as well when compared to primary HCC tissues. Migration and invasion analysis showed that evodiamine has anti-metastatic ability on Hep3B and Huh-7 cells and reduces the level of vimentin, an EMT marker. In conclusion, YAP is a critical target in HCC therapy, and evodiamine can be an effective HCC anticancer drug by reducing the YAP level.
Collapse
|
27
|
Su T, Wang T, Zhang N, Shen Y, Li W, Xing H, Yang M. Long non-coding RNAs in gastrointestinal cancers: implications for protein phosphorylation. Biochem Pharmacol 2022; 197:114907. [PMID: 35007523 DOI: 10.1016/j.bcp.2022.114907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/24/2022]
Abstract
Phosphorylation of proteins is one of the most extensively investigated post-translational protein modifications. Threonine, serine and tyrosine in proteins are the most commonly phosphorylated amino acids. Dysregulated cancer-related signaling pathways due to aberrant phosphorylation status of the key protein(s) in these pathways exist in most malignancies. Intensive studies in the recent decade have implicated long non-coding RNAs (lncRNAs) in the precise regulation of protein phosphorylation in cancers. In this review, we systematically delve into recent advance that underlines the multidimensional role of lncRNAs in modulating protein phosphorylation, regulating cancerous signaling and impacting prognosis of gastrointestinal (GI) cancers including hepatocellular carcinoma, colorectal cancer, gastric cancer, esophageal cancer, and pancreatic cancer. LncRNAs regulate protein phosphorylation via directly binding to the target protein(s), interacting with the partner protein(s) of the target protein(s) or lncRNAs-encoded small peptides. Although there are still extensive studies on disclosing the intricate interactions between lncRNAs and proteins and their impacts on protein phosphorylation, we believe that targeting lncRNAs controlling phosphorylation of key protein(s) in cancerous signaling pathways might provide novel paths for precision therapeutics of GI cancers in the future.
Collapse
Affiliation(s)
- Tao Su
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Teng Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Nasha Zhang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China
| | - Yue Shen
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenwen Li
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China
| | - Huaixin Xing
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China.
| | - Ming Yang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China.
| |
Collapse
|
28
|
Guo Y, Sun P, Guo W, Dong Z. Long Non-coding RNA LINC01503 Promotes Gastric Cardia Adenocarcinoma Progression via miR-133a-5p/VIM Axis and EMT Process. Dig Dis Sci 2021; 66:3391-3403. [PMID: 33200343 DOI: 10.1007/s10620-020-06690-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/21/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND LINC01503 has been reported to act as a candidate oncogenic lncRNA in several types of human cancer. However, the functions and underlying mechanisms of LINC01503 in gastric cardia adenocarcinoma (GCA) remain unclear. AIMS To investigate the roles and underlying mechanisms of LINC01503 in GCA progression. MATERIALS AND METHODS Gene expressions were detected by quantitative real-time PCR (qRT-PCR). Gain-of-function assays were performed to evaluate the function of LINC01503 in gastric cancer cells. Bioinformatics analysis, luciferase reporter assay, and RIP assay were performed to identify associations among LINC01503, miR-133a-5p, and VIM. RESULTS The expression level of LINC01503 was significantly elevated in GCA tissues and cell lines. High expression of LINC01503 was correlated with lymph node metastasis, TNM stage, and poor prognosis of GCA patients. Knockdown of LINC01503 significantly reduced proliferation, migration, and invasion ability in GC cells. LINC01503 might function as a competing endogenous RNA (ceRNA) via sponging miR-133a-5p to upregulate the expression of VIM. Furthermore, overexpression of LINC01503 promoted the progression of epithelial mesenchymal transition (EMT) in vitro. CONCLUSION LINC01503 serves as an oncogenic lncRNA to promote GCA progression via affecting LINC01503/miR-133a-5p/VIM axis and EMT process. LINC01503 not only has a critical role in GCA progression but also provide a novel potential biomarker in predicting prognosis for GCA patients.
Collapse
Affiliation(s)
- Yanli Guo
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, No.12, Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Pingping Sun
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wei Guo
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, No.12, Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Zhiming Dong
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, No.12, Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
29
|
Exacerbation of Liver Tumor Metastasis in twist1a+/ xmrk+ Double Transgenic Zebrafish following Lipopolysaccharide or Dextran Sulphate Sodium Exposure. Pharmaceuticals (Basel) 2021; 14:ph14090867. [PMID: 34577566 PMCID: PMC8468836 DOI: 10.3390/ph14090867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022] Open
Abstract
The poor prognosis for patients with hepatocellular carcinoma (HCC) is related directly to metastasis. The Twist1 gene encodes for a transcription factor essential to embryogenesis. It has also been shown to promote epithelial-to-mesenchymal transition (EMT), invasion, and metastasis; however, there is currently no in vivo evidence that Twist1 plays a role in the metastasis of liver tumors. Zebrafish are increasingly being used as an alternative cancer model. In the current study, an adult-stage zebrafish HCC model was used to examine the synergistic effects of twist1a and xmrk, a well characterized oncogene, during HCC metastasis. We also examined the effects of two inflammatory agents, lipopolysaccharides (LPS) and dextran sulfate sodium (DSS), on the hepatocyte-specific expression of transgenic twist1a and xmrk. The conditional overexpression of twist1a and xmrk was shown to promote liver tumor metastasis in zebrafish, resulting in increased apoptosis and cell proliferation as well as tumor maintenance and propagation independent of the inherent EMT-inducing activity of xmrk. Exposing twist1a+/xmrk+ transgenic zebrafish to LPS or DSS was shown to promote metastasis, indicating that the overexpression of twist1a and xmrk led to crosstalk between the signaling pathways involved in EMT. This study provides important evidence pertaining to the largely overlooked effects of signaling crosstalk between twist1a and xmrk in regulating HCC metastasis. Our results also suggest that the co-expression of twist1a/xmrk in conjunction with exposure to LPS or DSS enhances HCC metastasis, and provides a valuable in vivo platform by which to investigate tumor initiation and metastasis in the study of liver cancer.
Collapse
|
30
|
Lambrecht C, Ferreira GB, Omella JD, Libbrecht L, DE Vos R, Derua R, Mathieu C, Overbergh L, Waelkens E, Janssens V. Differential Proteomic Analysis of Hepatocellular Carcinomas from Ppp2r5d Knockout Mice and Normal (Knockout) Livers. Cancer Genomics Proteomics 2021; 17:669-685. [PMID: 33099469 DOI: 10.21873/cgp.20222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the major type of primary liver cancer. Mice lacking the tumor-suppressive protein phosphatase 2A subunit B56δ (Ppp2r5d) spontaneously develop HCC, correlating with increased c-MYC oncogenicity. MATERIALS AND METHODS We used two-dimensional difference gel electrophoresis-coupled matrix-assisted laser desorption/ionization time-of-flight mass spectrometry to identify differential proteomes of livers from wild-type, non-cancerous and HCC-affected B56δ knockout mice. RESULTS A total of 23 proteins were differentially expressed/regulated in liver between wild-type and non-cancerous knockout mice, and 119 between non-cancerous and HCC knockout mice ('cancer proteins'). Overlap with our reported differential transcriptome data was poor. Overall, 56% of cancer proteins were reported before in HCC proteomics studies; 44% were novel. Gene Ontology analysis revealed cancer proteins mainly associated with liver metabolism (18%) and mitochondria (15%). Ingenuity Pathway Analysis identified 'cancer' and 'gastrointestinal disease' as top hits. CONCLUSION We identified several proteins for further exploration as novel potential HCC biomarkers, and independently underscored the relevance of Ppp2r5d knockout mice as a valuable hepatocarcinogenesis model.
Collapse
Affiliation(s)
- Caroline Lambrecht
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Gabriela Bomfim Ferreira
- Clinical and Experimental Endocrinology, Department Clinical and Experimental Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Judit DomÈnech Omella
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Louis Libbrecht
- Department of Pathology, Université Catholique de Louvain (UCL), Brussels, Belgium
| | - Rita DE Vos
- Translational Cell and Tissue Research, Department Imaging and Pathology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department Clinical and Experimental Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Lut Overbergh
- Clinical and Experimental Endocrinology, Department Clinical and Experimental Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium .,LKI, KU Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
31
|
Wu W, Wang C, Wang F, Wang Y, Jin Y, Luo J, Wang M, Zhang C, Wang S, Zhang F, Li M. Silencing the COPB2 gene decreases the proliferation, migration and invasion of human triple-negative breast cancer cells. Exp Ther Med 2021; 22:792. [PMID: 34093748 PMCID: PMC8170640 DOI: 10.3892/etm.2021.10224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/23/2021] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is highly invasive, has a high rate of recurrence and is associated with a poor clinical outcome when compared with non-TNBC due to a lack of effective and targeted treatments. The coatomer protein complex subunit β2 (COPB2) is upregulated in various types of malignant cancer. The present study demonstrated that COPB2 expression levels were significantly upregulated in breast carcinoma HS-578T cells (clonal cells originating from TNBC) when compared with non-TNBC MCF-7 cells. HS-578T cells also exhibited higher rates of proliferation, invasion and transendothelial migration when compared with MCF-7 cells. Moreover, it was identified that genetically silencing the COPB2 gene using a lentivirus-short hairpin RNA inhibited the proliferative, colony formation, migratory and invasive properties of the TNBC HS-578T cells. Mediation of the COPB2 silencing effect may be associated with regulating the phosphorylation of serine/threonine kinase AKT in the PI3K/AKT signaling pathway. These results suggested the importance of COPB2 in promoting the proliferation of TNBC cells and identified COPB2 as a potential novel therapeutic target.
Collapse
Affiliation(s)
- Wencheng Wu
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chenyu Wang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Fengxia Wang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yan Wang
- Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yanling Jin
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jing Luo
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Min Wang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chenli Zhang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Shuya Wang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Fangfang Zhang
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Min Li
- Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
32
|
The CD44high Subpopulation of Multifraction Irradiation-Surviving NSCLC Cells Exhibits Partial EMT-Program Activation and DNA Damage Response Depending on Their p53 Status. Int J Mol Sci 2021; 22:ijms22052369. [PMID: 33673439 PMCID: PMC7956695 DOI: 10.3390/ijms22052369] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation (IR) is used for patients diagnosed with unresectable non-small cell lung cancer (NSCLC). However, radiotherapy remains largely palliative due to the survival of specific cell subpopulations. In the present study, the sublines of NSCLC cells, A549IR (p53wt) and H1299IR (p53null) survived multifraction X-ray radiation exposure (MFR) at a total dose of 60 Gy were investigated three weeks after the MFR course. We compared radiosensitivity (colony formation), expression of epithelial-mesenchymal transition (EMT) markers, migration activity, autophagy, and HR-dependent DNA double-strand break (DSB) repair in the bulk and entire CD44high/CD166high CSC-like populations of both parental and MFR survived NSCLC cells. We demonstrated that the p53 status affected: the pattern of expression of N-cadherin, E-cadherin, Vimentin, witnessing the appearance of EMT-like phenotype of MFR-surviving sublines; 1D confined migratory behavior (wound healing); the capability of an irradiated cell to continue to divide and form a colony of NSCLC cells before and after MFR; influencing the CD44/CD166 expression level in MFR-surviving NSCLC cells after additional single irradiation. Our data further emphasize the impact of p53 status on the decay of γH2AX foci and the associated efficacy of the DSB repair in NSCLC cells survived after MFR. We revealed that Rad51 protein might play a principal role in MFR-surviving of p53 null NSCLC cells promoting DNA DSB repair by homologous recombination (HR) pathway. The proportion of Rad51 + cells elevated in CD44high/CD166high population in MFR-surviving p53wt and p53null sublines and their parental cells. The p53wt ensures DNA-PK-mediated DSB repair for both parental and MFR-surviving cells irrespectively of a subsequent additional single irradiation. Whereas in the absence of p53, a dose-dependent increase of DNA-PK-mediated non-homologous end joining (NHEJ) occurred as an early post-irradiation response is more intensive in the CSC-like population MFR-surviving H1299IR, compared to their parental H1299 cells. Our study strictly observed a significantly higher content of LC3 + cells in the CD44high/CD166high populations of p53wt MFR-surviving cells, which enriched the CSC-like cells in contrast to their p53null counterparts. The additional 2 Gy and 5 Gy X-ray exposure leads to the dose-dependent increase in the proportion of LC3 + cells in CD44high/CD166high population of both parental p53wt and p53null, but not MFR-surviving NSCLC sublines. Our data indicated that autophagy is not necessarily associated with CSC-like cells’ radiosensitivity, emphasizing that careful assessment of other milestone processes (such as senescence and autophagy-p53-Zeb1 axis) of primary radiation responses may provide new potential targets modulated for therapeutic benefit through radiosensitizing cancer cells while rescuing normal tissue. Our findings also shed light on the intricate crosstalk between autophagy and the p53-related EMT, by which MFR-surviving cells might obtain an invasive phenotype and metastatic potential.
Collapse
|
33
|
Shidham VB, Layfield LJ. Cell-blocks and immunohistochemistry. Cytojournal 2021; 18:2. [PMID: 33598043 PMCID: PMC7881511 DOI: 10.25259/cytojournal_83_2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/25/2022] Open
Abstract
The interpretation of results on immunostained cell-block sections has to be compared with the cumulative published data derived predominantly from formalin-fixed paraffin-embedded (FFPE) tissue sections. Because of this, it is important to recognize that the fixation and processing protocol should not be different from the routinely processed FFPE surgical pathology tissue. Exposure to non-formalin fixatives or reagents may interfere with the diagnostic immunoreactivity pattern. The immunoprofile observed on such cell-blocks, which are not processed in a manner similar to the surgical pathology specimens, may not be representative resulting in aberrant results. The field of immunohistochemistry (IHC) is advancing continuously with the standardization of many immunomarkers. A variety of technical advances such as multiplex IHC with refined methodologies and automation is increasing its role in clinical applications. The recent addition of rabbit monoclonal antibodies has further improved sensitivity. As compared to the mouse monoclonal antibodies, the rabbit monoclonal antibodies have 10 to 100 fold higher antigen affinity. Most of the scenarios involve the evaluation of coordinate immunostaining patterns in cell-blocks with relatively scant diagnostic material without proper orientation which is usually retained in most of the surgical pathology specimens. These challenges are addressed if cell-blocks are prepared with some dedicated methodologies such as NextGen CelBloking™ (NGCB) kits. Cell-blocks prepared by NGCB kits also facilitate the easy application of the SCIP (subtractive coordinate immunoreactivity pattern) approach for proper evaluation of coordinate immunoreactivity. Various cell-block and IHC-related issues are discussed in detail.
Collapse
Affiliation(s)
- Vinod B. Shidham
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Center and Detroit Medical Center, Detroit, Michigan, USA
| | - Lester J. Layfield
- Department of Pathology and Anatomical Sciences, University of Missouri, One Hospital Drive, Columbia, Missouri, United States
| |
Collapse
|
34
|
Bai Y, Lian Y, Chen X, Wu J, Lai J, Qiu F, Zhou S, Zhu Z, Tian Y, Wang Y, Yang Y, Yan M. Immunohistochemical Signature Add Prognostic Value in Patients With Early and Intermediate Hepatocellular Carcinoma Underwent Curative Liver Resection. Front Oncol 2021; 10:616263. [PMID: 33585243 PMCID: PMC7874098 DOI: 10.3389/fonc.2020.616263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most lethal cancer worldwide; however, accurate prognostic tools are still lacking. We aimed to identify immunohistochemistry (IHC)-based signature as a prognostic classifier to predict recurrence and survival in patients with HCC at Barcelona Clinic Liver Cancer (BCLC) early- and immediate-stage. In total, 567 patients who underwent curative liver resection at two independent centers were enrolled. The least absolute shrinkage and selection operator regression model was used to identify significant IHC features, and penalized Cox regression was used to further narrow down the features in the training cohort (n = 201). The candidate IHC features were validated in internal (n = 101) and external validation cohorts (n = 265). Three IHC features, hepatocyte paraffin antigen 1, CD34, and Ki-67, were identified as candidate predictors for recurrence-free survival (RFS), and were used to categorize patients into low- and high-risk recurrence groups in the training cohort (P < 0.001). The discriminative performance of the 3-IHC_based classifier was validated using internal and external cohorts (P < 0.001). Furthermore, we developed a 3-IHC_based nomogram integrating the BCLC stage, microvascular invasion, and 3-IHC_based classifier to predict 2- and 5-year RFS in the training cohort; this nomogram exhibited acceptable area under the curve values for the training, internal validation, and external validation cohorts (2-year: 0.817, 0.787, and 0.810; 5-year: 0.726, 0.662, and 0.715; respectively). The newly developed 3-IHC_based classifier can effectively predict recurrence and survival in patients with early- and intermediate-stage HCC after curative liver resection.
Collapse
Affiliation(s)
- Yannan Bai
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yuane Lian
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoping Chen
- Department of Statistics, College of Mathematics and Informatics & FJKLMAA, Fujian Normal University, Fuzhou, China
| | - Jiayi Wu
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianlin Lai
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Funan Qiu
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Songqiang Zhou
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zijing Zhu
- Department of Statistics, College of Mathematics and Informatics & FJKLMAA, Fujian Normal University, Fuzhou, China
| | - Yifeng Tian
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yaodong Wang
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maolin Yan
- Department of Hepatobiliopancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
35
|
Iron elevates mesenchymal and metastatic biomarkers in HepG2 cells. Sci Rep 2020; 10:21926. [PMID: 33318518 PMCID: PMC7736862 DOI: 10.1038/s41598-020-78348-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/23/2020] [Indexed: 01/20/2023] Open
Abstract
Liver iron excess is observed in several chronic liver diseases and is associated with the development of hepatocellular carcinoma (HCC). However, apart from oxidative stress, other cellular mechanisms by which excess iron may mediate/increase HCC predisposition/progression are not known. HCC pathology involves epithelial to mesenchymal transition (EMT), the basis of cancer phenotype acquisition. Here, the effect of excess iron (holo-transferrin 0–2 g/L for 24 and 48 h) on EMT biomarkers in the liver-derived HepG2 cells was investigated. Holo-transferrin substantially increased intracellular iron. Unexpectedly, mRNA and protein expression of the epithelial marker E-cadherin either remained unaltered or increased. The mRNA and protein levels of metastasis marker N-cadherin and mesenchymal marker vimentin increased significantly. While the mRNA expression of EMT transcription factors SNAI1 and SNAI2 increased and decreased, respectively after 24 h, both factors increased after 48 h. The mRNA expression of TGF-β (EMT-inducer) showed no significant alterations. In conclusion, data showed direct link between iron and EMT. Iron elevated mesenchymal and metastatic biomarkers in HepG2 cells without concomitant decrement in the epithelial marker E-cadherin and altered the expression of the key EMT-mediating transcription factors. Such studies can help identify molecular targets to devise iron-related adjunctive therapies to ameliorate HCC pathophysiology.
Collapse
|
36
|
Sjöqvist M, Antfolk D, Suarez-Rodriguez F, Sahlgren C. From structural resilience to cell specification - Intermediate filaments as regulators of cell fate. FASEB J 2020; 35:e21182. [PMID: 33205514 PMCID: PMC7839487 DOI: 10.1096/fj.202001627r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
During the last decades intermediate filaments (IFs) have emerged as important regulators of cellular signaling events, ascribing IFs with functions beyond the structural support they provide. The organ and developmental stage‐specific expression of IFs regulate cell differentiation within developing or remodeling tissues. Lack of IFs causes perturbed stem cell differentiation in vasculature, intestine, nervous system, and mammary gland, in transgenic mouse models. The aberrant cell fate decisions are caused by deregulation of different stem cell signaling pathways, such as Notch, Wnt, YAP/TAZ, and TGFβ. Mutations in genes coding for IFs cause an array of different diseases, many related to stem cell dysfunction, but the molecular mechanisms remain unresolved. Here, we provide a comprehensive overview of how IFs interact with and regulate the activity, localization and function of different signaling proteins in stem cells, and how the assembly state and PTM profile of IFs may affect these processes. Identifying when, where and how IFs and cell signaling congregate, will expand our understanding of IF‐linked stem cell dysfunction during development and disease.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Freddy Suarez-Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
37
|
The Expressions and Mechanisms of Sarcomeric Proteins in Cancers. DISEASE MARKERS 2020; 2020:8885286. [PMID: 32670437 PMCID: PMC7346232 DOI: 10.1155/2020/8885286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
The sarcomeric proteins control the movement of cells in diverse species, whereas the deregulation can induce tumours in model organisms and occurs in human carcinomas. Sarcomeric proteins are recognized as oncogene and related to tumor cell metastasis. Recent insights into their expressions and functions have led to new cancer therapeutic opportunities. In this review, we appraise the evidence for the sarcomeric proteins as cancer genes and discuss cancer-relevant biological functions, potential mechanisms by which sarcomeric proteins activity is altered in cancer.
Collapse
|
38
|
Aldolase B suppresses hepatocellular carcinogenesis by inhibiting G6PD and pentose phosphate pathways. ACTA ACUST UNITED AC 2020; 1:735-747. [DOI: 10.1038/s43018-020-0086-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
|
39
|
Pandit H, Li Y, Zheng Q, Guo W, Yu Y, Li S, Martin RCG. Carcinogenetic initiation contributed by EpCAM+ cancer cells in orthotopic HCC models of immunocompetent and athymic mice. Oncotarget 2020; 11:2047-2060. [PMID: 32547703 PMCID: PMC7275786 DOI: 10.18632/oncotarget.27454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Overexpression of epithelial cell adhesion molecule (EpCAM) correlates with poor prognosis, therapeutic failure and early tumor recurrence in hepatocellular carcinoma (HCC) patients. The tumor microenvironment dictates the fate of tumor-initiating cancer stem cells (CSCs); however, very limited studies were attempted to evaluate CSC tumorigenesis in the liver microenvironment. Here, we have systemically investigated the role of EpCAM+ cancer cells in tumor initiation in orthotopic HCC models. RESULTS Control mice and the mice with bland steatosis failed to develop tumors. In the mice with steatohepatitis, EpCAM+ CSCs have shown significantly increased ability in terms of tumor initiation and growth, compared to that with EpCAM- non-CSCs inoculation (p < 0.005). For Hep3B inoculation, EpCAM-High group has shown significantly higher tumor growth compared with EpCAM-Low (p < 0.005). For HepG2 inoculation, both EpCAM-High and EpCAM-Low groups confirmed similar tumor incidence and growth. METHODS Diet-induced compromised microenvironments were established to mimic clinical fatty liver and non-alcoholic steatohepatitis (NASH) patients and the tumorigenic capabilities of Hepa1-6 cells were evaluated. CSCs were enriched by spheroid culture and labeled with copGFP for EpCAM+ CSCs and with mCherry for non-CSCs. FACS-sorted cells were inoculated into left liver lobes, and tumor growth was monitored by high-frequency ultrasound. The subpopulations of Hep3B and HepG2 cells in terms of EpCAM-Low and EpCAM-High were evaluated in the orthotopic model of athymic mice. CONCLUSIONS NASH microenvironment promotes the EpCAM+ CSCs initiated tumorigenesis in immunocompetent mouse model. Differential EpCAM expression demonstrates distinct tumor biology in athymic mouse models.
Collapse
Affiliation(s)
- Harshul Pandit
- Division of Surgical Oncology, Hiram C. Polk Jr. M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Yan Li
- Division of Surgical Oncology, Hiram C. Polk Jr. M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, Shenyang 110122, China
| | - Wei Guo
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Youxi Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Suping Li
- Division of Surgical Oncology, Hiram C. Polk Jr. M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Robert C G Martin
- Division of Surgical Oncology, Hiram C. Polk Jr. M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
40
|
Zhao Y, Zhao MF, Yang ML, Wu TY, Xu CJ, Wang JM, Li CJ, Li X. G Protein-Coupled Receptor 30 Mediates the Anticancer Effects Induced by Eicosapentaenoic Acid in Ovarian Cancer Cells. Cancer Res Treat 2020; 52:815-829. [PMID: 32138466 PMCID: PMC7373874 DOI: 10.4143/crt.2019.380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 03/03/2020] [Indexed: 01/23/2023] Open
Abstract
Purpose While numerous epidemiological studies have indicated that omega-3 polyunsaturated fatty acids have anticancer properties in various cancers, the effects and mechanisms of eicosapentaenoic acid (EPA) in ovarian cancer cell growth are poorly understood. Materials and Methods ES2 ovarian clear cell carcinoma cells and SKOV3 adenocarcinoma cells were treated with palmitic acid or EPA, followed by flow cytometry and cell counting to measure apoptosis and proliferation, respectively. A modified protein lipid overlay assay was used to further verify whether EPA was a ligand of G protein–coupled receptor 30 (GPR30) in ES2 cells. The levels of apoptosis-related genes, phosphorylated AKT, and phosphorylated ERK1/2 were detected to explore the underlying mechanism. Finally, inhibitory effect of EPA on tumor growth via GPR30 was determined in vitro and in vivo. Results EPA suppressed ES2 ovarian clear cell carcinoma cells growth via GPR30, a novel EPA receptor, by inducing apoptosis. As a ligand of GPR30, EPA activated the GPR30-cAMP–protein kinase A signaling pathway. When GPR30 was suppressed by siRNA or its inhibitor G15, the antiproliferative action of EPA was impaired. Furthermore, EPA inhibited tumor growth by blocking the activation of AKT and ERK. In the mouse xenograft model, EPA decreased tumor volume and weight through GPR30 by blocking tumor cell proliferation. Conclusion These results confirm that EPA is a tumor suppressor in human ovarian clear cell carcinoma cells and functions through a novel fatty acid receptor, GPR30, indicating a mechanistic linkage between omega-3 fatty acids and cancers.
Collapse
Affiliation(s)
- Yue Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Meng-Fei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Mei-Lin Yang
- Department of Obstetrics and Gynecology, Xiamen Chang Gung Hospital, Xiamen, China
| | - Tian-Yu Wu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Cong-Jian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jing-Mei Wang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xi Li
- Biology Science Institutes, Chongqing Medical University, Yuzhong, China
| |
Collapse
|
41
|
Liao H, Xiong T, Peng J, Xu L, Liao M, Zhang Z, Wu Z, Yuan K, Zeng Y. Classification and Prognosis Prediction from Histopathological Images of Hepatocellular Carcinoma by a Fully Automated Pipeline Based on Machine Learning. Ann Surg Oncol 2020; 27:2359-2369. [PMID: 31916093 DOI: 10.1245/s10434-019-08190-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The aim of this study was to develop quantitative feature-based models from histopathological images to distinguish hepatocellular carcinoma (HCC) from adjacent normal tissue and predict the prognosis of HCC patients after surgical resection. METHODS A fully automated pipeline was constructed using computational approaches to analyze the quantitative features of histopathological slides of HCC patients, in which the features were extracted from the hematoxylin and eosin (H&E)-stained whole-slide images of HCC patients from The Cancer Genome Atlas and tissue microarray images from West China Hospital. The extracted features were used to train the statistical models that classify tissue slides and predict patients' survival outcomes by machine-learning methods. RESULTS A total of 1733 quantitative image features were extracted from each histopathological slide. The diagnostic classifier based on 31 features was able to successfully distinguish HCC from adjacent normal tissues in both the test [area under the receiver operating characteristic curve (AUC) 0.988] and external validation sets (AUC 0.886). The random-forest prognostic model using 46 features was able to significantly stratify patients in each set into longer- or shorter-term survival groups according to their assigned risk scores. Moreover, the prognostic model we constructed showed comparable predicting accuracy as TNM staging systems in predicting patients' survival at different time points after surgery. CONCLUSIONS Our findings suggest that machine-learning models derived from image features can assist clinicians in HCC diagnosis and its prognosis prediction after hepatectomy.
Collapse
Affiliation(s)
- Haotian Liao
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Tianyuan Xiong
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Lin Xu
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Mingheng Liao
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Zhen Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenru Wu
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.
| | - Yong Zeng
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.
| |
Collapse
|
42
|
Mandal K, Gong Z, Rylander A, Shenoy VB, Janmey PA. Opposite responses of normal hepatocytes and hepatocellular carcinoma cells to substrate viscoelasticity. Biomater Sci 2020; 8:1316-1328. [PMID: 31903466 DOI: 10.1039/c9bm01339c] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cellular microenvironment plays a critical role in cell differentiation, proliferation, migration, and homeostasis. Recent studies have shown the importance of substrate viscosity in determining cellular function. Here, we study the mechanoresponse of normal hepatocytes and hepatocellular carcinoma cells (HCC) to elastic and viscoelastic substrates using the Huh7 cell line derived from a human liver tumor and primary human hepatocytes (PHH). Unlike PHH and fibroblasts, which respond to viscoelastic substrates by reducing spreading area and actin bundle assembly compared to purely elastic substrates of the same stiffness, Huh7 cells spread faster on viscoelastic substrates than on purely elastic substrates. The steady state spreading areas of Huh7 cells are larger on viscoelastic substrates, whereas the opposite effect occurs with PHH cells. The viscoelasticity of the microenvironment also promotes motility and multiple long protrusions in Huh7 cells. Pharmacologic disruption of the actin assembly makes cells unable to spread on either elastic or viscoelastic substrates. In contrast, upon vimentin perturbation, cells still spread to a limited degree on elastic substrates but are unable to spread on viscoelastic substrates. The time evolution of cell traction force shows that the peak occurs at an earlier time point on viscoelastic substrates compared to elastic substrates. However, the total force generation at steady state is the same on both substrates after 4 hours. Our data suggest that stress relaxation time scales of the viscoelastic substrate regulate cell dynamics and traction force generation, indicating different binding-unbinding rates of the proteins that form cell attachment sites in HCC cells and normal hepatocytes. These results suggest that liver cancer cells may have different characteristic lifetimes of binding to the substrate in comparision to normal cells, which might cause differences in cell spreading and motility within the diseased tissue.
Collapse
Affiliation(s)
- Kalpana Mandal
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia 19104, USA.
| | | | | | | | | |
Collapse
|
43
|
Kim BR, Ha J, Lee S, Park J, Cho S. Anti-cancer effects of ethanol extract of Reynoutria japonica Houtt. radix in human hepatocellular carcinoma cells via inhibition of MAPK and PI3K/Akt signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2019; 245:112179. [PMID: 31445130 DOI: 10.1016/j.jep.2019.112179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/13/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Reynoutria japonica Houtt. has been used as a traditional medicine of cancer in East Asia for thousands of years. However, the mechanism of the anti-cancer effect of R. japonica has not been investigated at the molecular level. The regulation of intracellular signaling pathways by the extract of R. japonica radix needs to be evaluated for a deeper understanding and application of the anti-cancer effect of R. japonica radix. AIM OF THE STUDY The purpose of this study was to evaluate the inhibitory effects of the ethanol extracts of R. japonica radix (ERJR) on cancer metastasis and the regulation mechanism of metastasis by ERJR in human hepatocellular carcinomas. MATERIALS AND METHODS Suppression of cancer metastasis by ERJR in SK-Hep1 and Huh7 cells were investigated. Prior to experiments, the cytotoxic effect of ERJR was examined by cell viability assays. To evaluate the inhibitory effects of ERJR on cancer metastasis, wound-healing assays, invasion assays, zymography, and multicellular tumor spheroids (MCTS) assays were performed. Molecular mechanisms in the suppressive regulation of metastasis by ERJR were verified by measuring the expression levels of metastatic markers, and the phosphorylation and protein levels of cancer metastasis-related signaling pathways. RESULTS In all experiments, ERJR was used at a maximum concentration of 20 μg/ml, which did not show cytotoxicity in SK-Hep1 and Huh7 cells. We examined the inhibitory effects of ERJR on cancer metastasis. In wound-healing and invasion assays, ERJR treatment effectively suppressed the wound-recovery of Huh7 cells and inhibited the invasion ability of SK-Hep1 cells. Also, ERJR treatment significantly decreased the enzymatic activity of matrix metalloproteinase-2 and -9 in SK-Hep1 cells. ERJR suppressed the growth of MCTS in SK-Hep1 cells in a dose-dependent manner. These results indicated that ERJR effectively inhibited the invasive and proliferative ability of SK-Hep1 and Huh7 cells. Moreover, ERJR treatment reduced the expression levels of Snail1, Twist1, N-cadherin, and Vimentin, which are metastatic markers, by inhibiting the activation of protein kinase B and mitogen-activated protein kinases in SK-Hep1 cells. CONCLUSIONS These results verified the molecular mechanism of ERJR that has been used in traditional anti-cancer remedy and suggest that it can be developed as a promising therapy for cancer metastasis in the future.
Collapse
Affiliation(s)
- Ba Reum Kim
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea
| | - Jain Ha
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea
| | - Sewoong Lee
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea
| | - Jiyoung Park
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea
| | - Sayeon Cho
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
44
|
Nissen NI, Karsdal M, Willumsen N. Post-translational modifications of vimentin reflect different pathological processes associated with non-small cell lung cancer and chronic obstructive pulmonary disease. Oncotarget 2019; 10:6829-6841. [PMID: 31827725 PMCID: PMC6887574 DOI: 10.18632/oncotarget.27332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Vimentin has shown to be highly implicated in cancer initiation and progression. Vimentin is often a target of post-translational modifications (PTMs) which can be disease specific, thus targeting these specific modifications can be of high biomarker potential. In this study we set out to evaluate the biological relevance and serum biomarker potential of citrullinated vimentin (VICM) and non-citrullinated vimentin (VIM) in non-small cell lung cancer (NSCLC) and chronic obstructive pulmonary disease (COPD). METHODS A competitive ELISA targeting VIM was developed and quantified in serum from patients with NSCLC and COPD. VIM was compared with levels of VICM in the same indications. RESULTS VIM was significantly increased in NSCLC (n = 100) compared to healthy controls (n = 67) in two independent cohorts (p = 0.0003 and p < 0.0001). Furthermore, VIM was highly increased in late stages of NSCLC (p = 0.001), however VIM was not increased in COPD patients (n = 10). Contrarily, serum levels of VICM was not increased in late stages of NSCLC, but highly elevated in patients with COPD (p < 0.0001). CONCLUSIONS These findings suggest a biomarker potential of VIM in NSCLC. Our findings also indicate that PTMs of vimentin are highly relevant and that targeting these modifications can have differential biomarker potential.
Collapse
Affiliation(s)
- Neel Ingemann Nissen
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark.,Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | | |
Collapse
|
45
|
Todenhöfer T, Pantel K, Stenzl A, Werner S. Pathophysiology of Tumor Cell Release into the Circulation and Characterization of CTC. Recent Results Cancer Res 2019; 215:3-24. [PMID: 31605221 DOI: 10.1007/978-3-030-26439-0_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The traditional model of metastatic progression postulates that the ability to form distant metastases is driven by random mutations in cells of the primary tumor.
Collapse
Affiliation(s)
- Tilman Todenhöfer
- Department of Urology, Eberhard-Karls-University, Tuebingen, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Arnulf Stenzl
- Department of Urology, Eberhard-Karls-University, Tuebingen, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Ho DWH, Tsui YM, Sze KMF, Chan LK, Cheung TT, Lee E, Sham PC, Tsui SKW, Lee TKW, Ng IOL. Single-cell transcriptomics reveals the landscape of intra-tumoral heterogeneity and stemness-related subpopulations in liver cancer. Cancer Lett 2019; 459:176-185. [PMID: 31195060 DOI: 10.1016/j.canlet.2019.06.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/23/2019] [Accepted: 06/06/2019] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is heterogeneous, rendering its current curative treatments ineffective. The emergence of single-cell genomics represents a powerful strategy in delineating the complex molecular landscapes of cancers. In this study, we demonstrated the feasibility and merit of using single-cell RNA sequencing to dissect the intra-tumoral heterogeneity and analyze the single-cell transcriptomic landscape to detect rare cell subpopulations of significance. Exploration of the inter-relationship among liver cancer stem cell markers showed two distinct major cell populations according to EPCAM expression, and the EPCAM+ cells had upregulated expression of multiple oncogenes. We also identified a CD24+/CD44+-enriched cell subpopulation within the EPCAM+ cells which had specific signature genes and might indicate a novel stemness-related cell subclone in HCC. Notably, knockdown of signature gene CTSE for CD24+/CD44+ cells significantly reduced self-renewal ability on HCC cells in vitro and the stemness-related role of CTSE was further confirmed by in vivo tumorigenicity assays in nude mice. In summary, single-cell genomics is a useful tool to delineate HCC intratumoral heterogeneity at better resolution. It can identify rare but important cell subpopulations, and may guide better precision medicine in the long run.
Collapse
Affiliation(s)
- Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Yu-Man Tsui
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Karen Man-Fong Sze
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Lo-Kong Chan
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Tan-To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Eva Lee
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Pak-Chung Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | | | - Terence Kin-Wah Lee
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong.
| |
Collapse
|
47
|
Zhou H, Xu J, Zhang C, Wen Y. Aberrant histone deacetylase 1 expression upregulates vimentin expression via an NF-κB-dependent pathway in hepatocellular carcinoma. Oncol Lett 2019; 18:339-347. [PMID: 31289505 PMCID: PMC6540068 DOI: 10.3892/ol.2019.10309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/12/2019] [Indexed: 12/14/2022] Open
Abstract
Aberrantly elevated expression levels of histone deacetylase 1 (HDAC1) and vimentin are closely associated with disease progression in hepatocellular carcinoma (HCC). It was previously demonstrated that knocking down expression of HDAC1 resulted in a concurrent decrease in the expression levels of vimentin. However, a causal link between these two proteins has not yet been demonstrated, to the best of our knowledge. In the present study, the association between HDAC1 and vimentin was investigated using an HDAC1 overexpression platform. HDAC1 and vimentin were significantly increased in HCC cells, and HDAC1 overexpression enhanced vimentin mRNA and protein expression levels in an HDAC1 dose-dependent manner. Subsequently, truncation and mutation of a vimentin promoter demonstrated that HDAC1-induced vimentin expression was dependent on a nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) binding site in the vimentin promoter sequence. Furthermore, HDAC1 induced vimentin expression by promoting NF-κB translocation between the cytoplasm and the nucleus, as opposed to modulating the total expression level of vimentin directly. The data in the present study demonstrated that HDAC1 is overexpressed in HCC and that HDAC1 may upregulate vimentin expression through the NF-κB signaling pathway, thus demonstrating a causal link between HDAC1 and vimentin in HCC, and may provide valuable information in understanding the pathogenesis of HCC.
Collapse
Affiliation(s)
- Huancheng Zhou
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Jiwei Xu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Caiyun Zhang
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Yuanzhang Wen
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| |
Collapse
|
48
|
Cao X, Xu L, Liu Q, Yang L, Li N, Li X. MicroRNA-1277 Inhibits Proliferation and Migration of Hepatocellular Carcinoma HepG2 Cells by Targeting and Suppressing BMP4 Expression and Reflects the Significant Indicative Role in Hepatocellular Carcinoma Pathology and Diagnosis After Magnetic Resonance Imaging Assessment. Oncol Res 2019; 27:301-309. [PMID: 29562958 PMCID: PMC7848408 DOI: 10.3727/096504018x15213058045841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Our study aimed to investigate the roles and possible regulatory mechanism of miR-1277 in the development of hepatocellular carcinoma (HCC). HCC patients were identified from patients who were diagnosed with focal liver lesions using magnetic resonance imaging (MRI). The expression levels of miR-1277 in the serum of HCC patients and HepG2 cells were measured. Then miR-1277 mimic, miR-1277 inhibitor, or scramble RNA was transfected into HepG2 cells. The effects of miR-1277 overexpression and suppression on HepG2 cell proliferation, migration, and invasion were then investigated. Additionally, the expression levels of epithelial-mesenchymal transition (EMT)-related markers, including E-cadherin, β-catenin, and vimentin, were detected. Target prediction and luciferase reporter assay were performed to explore the potential target of miR-1277. miR-1277 was significantly downregulated in the serum of HCC patients and HepG2 cells. Suppression of miR-1277 promoted HepG2 cell proliferation, migration, and invasion, whereas overexpression of miR-1277 had opposite effects. In addition, after miR-1277 was suppressed, the expressions of E-cadherin and β-catenin were significantly increased, while the expressions of vimentin were markedly decreased. Bone morphogenetic protein 4 (BMP4) was identified as the direct target of miR-1277. Knockdown of BMP4 reversed the effects of miR-1277 suppression on HepG2 cell migration and invasion, as well as the expressions of E-cadherin, β-catenin, and vimentin. Our results indicate that downregulation of miR-1277 may promote the migration and invasion of HepG2 cells by targeting BMP4 to induce EMT. Combination of MRI and miR-1277 level will facilitate the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Xinshan Cao
- *Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Ling Xu
- †Department of Liver Disease Center, Traditional Chinese Medicine Hospital of Binzhou City, Binzhou, Shandong, P.R. China
| | - Quanyuan Liu
- *Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Lijuan Yang
- ‡Department of Experiment Center of Tumor, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Na Li
- §Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Xiaoxiao Li
- *Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| |
Collapse
|
49
|
Sun X, Wang M, Zhang F, Kong X. Inhibition of NET-1 suppresses proliferation and promotes apoptosis of hepatocellular carcinoma cells by activating the PI3K/AKT signaling pathway. Exp Ther Med 2019; 17:2334-2340. [PMID: 30867719 DOI: 10.3892/etm.2019.7211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/20/2018] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to elucidate the underlying mechanism of neuroepithelial cell transforming 1 (NET-1), a member of the Ras homolog gene family, in hepatocellular carcinoma (HCC). To determine the association between the expression of NET-1 and the proliferation and migration of MHCC97-H cells, the cells were transfected with NET-1 small interfering (si)RNA and si negative control. Following transfection with NET-1 siRNA, the proliferation rate of MHCC97-H cells decreased significantly and the percentage of apoptotic cells increased. The HCC cell line MHCC97-H was used in the present study as it exhibited an increased expression level of NET-1 compared with the MHCC97-L cell line. Expression levels of apoptosis-associated proteins including apoptosis regulator Bax (Bax), cyclinD1, apoptosis regulator Bcl-2 (Bcl-2) and caspase-3 were determined. Expression levels of phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) and their phosphorylated forms were also measured by western blotting. Following NET-1 knockdown, the expression of Bax and cyclinD1 decreased, the expression of Bcl-2 and caspase-3 increased, and the PI3K/AKT signaling pathway was inhibited. The results of the present study suggest that inhibition of NET-1 can suppress the progression of HCC by targeting the PI3K/AKT signaling pathway. NET-1 expression level in HCC cells increased compared with normal liver cells.
Collapse
Affiliation(s)
- Xiangjun Sun
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Mingchun Wang
- Department of Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Fenghua Zhang
- Department of Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Xiao Kong
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
50
|
Kim DH, Park SJ, Lee SY, Yoon HS, Park CM. Silymarin Attenuates Invasion and Migration through the Regulation of Epithelial-mesenchymal Transition in Huh7 Cells. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2018. [DOI: 10.15324/kjcls.2018.50.3.337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Do-Hoon Kim
- Department of Clinical Laboratory Science, Dong-Eui University, Busan, Korea
| | - So-Jeong Park
- Department of Clinical Laboratory Science, Dong-Eui University, Busan, Korea
| | - Seung-Yeon Lee
- Department of Clinical Laboratory Science, Dong-Eui University, Busan, Korea
| | - Hyun-Seo Yoon
- Department of Dental Hygiene, Dong-Eui University, Busan, Korea
| | - Chung Mu Park
- Department of Clinical Laboratory Science, Dong-Eui University, Busan, Korea
| |
Collapse
|