1
|
Prescott JB, Liu KJ, Lander A, Pek NMQ, Jha SK, Bokelmann M, Begur M, Koh PW, Yang H, Lim B, Red-Horse K, Weissman IL, Loh KM, Ang LT. Metabolically purified human stem cell-derived hepatocytes reveal distinct effects of Ebola and Lassa viruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638665. [PMID: 40027809 PMCID: PMC11870522 DOI: 10.1101/2025.02.17.638665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Ebola and Lassa viruses require biosafety-level-4 (BSL4) containment, infect the liver, and cause deadly hemorrhagic fevers. The cellular effects of these viruses, and whether different families of hemorrhagic-fever viruses elicit similar effects, remain fundamental questions in BSL4 virology. Here, we introduce a new metabolic selection approach to create nearly-pure hepatocytes from human pluripotent stem cells, killing non-liver cells by withholding essential nutrients. Unexpectedly, Ebola and Lassa exerted starkly different effects on human hepatocytes. Ebola infection activated the integrated stress response (ISR) and WNT pathways in hepatocytes in vitro and killed them, whereas Lassa did not. Within non-human primates, Ebola likewise infected hepatocytes and activated ISR signaling in vivo . In summary, we present a single-cell transcriptional and chromatin accessibility roadmap of human hepatocyte differentiation, purification, and viral infection.
Collapse
|
2
|
Szwed M, Jost T, Majka E, Gharibkandi NA, Majkowska-Pilip A, Frey B, Bilewicz A, Fietkau R, Gaipl U, Marczak A, Lubgan D. Pt-Au Nanoparticles in Combination with Near-Infrared-Based Hyperthermia Increase the Temperature and Impact on the Viability and Immune Phenotype of Human Hepatocellular Carcinoma Cells. Int J Mol Sci 2025; 26:1574. [PMID: 40004038 PMCID: PMC11855494 DOI: 10.3390/ijms26041574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Near-infrared light (NIR)-responsive metal-based nanoparticles (NPs) could be used for tumour therapy. We examined how platinum (Pt), gold (Au), and core-shell Pt-Au NPs affect the viability of human hepatocellular carcinoma (HCC) cell lines (Hep3B, HepG2, and Huh7D-12) alone and in combination with NIR exposure. In addition, the expression of immune checkpoint molecules (ICMs) on the tumour cells was analysed. We revealed that the cytotoxicity and programmed cell death induction of Au and Pt-Au NPs toward HCC cells could be enhanced by NIR with 960 nm in a different way. Pt-Au NPs were the only particles that resulted in an additional temperature increase of up to 2 °C after NIR. Regarding the tumour cell immune phenotype, not all of the cells experienced changes in immune phenotype. NIR itself was the trigger of the alterations, while the NPs did not significantly affect the expression of most of the examined ICMs, such as PD-L1, PD-L1, HVEM, CD70, ICOS-L, Ox40-L, and TNFRSF9. The combination of Pt-Au NPs with NIR resulted in the most prominent increase of ICMs in HepG2 cells. We conclude that the thermotherapeutic effect of Pt-Au NP application and NIR could be beneficial in multimodal therapy settings in liver cancer for selected patients.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Tina Jost
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Emilia Majka
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Nasrin Abbasi Gharibkandi
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Agnieszka Majkowska-Pilip
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Rainer Fietkau
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Udo Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Agnieszka Marczak
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Dorota Lubgan
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
3
|
Soubeyrand S, Lau P, McPherson R. Distinct roles of Constitutive Photomorphogenesis Protein 1 homolog (COP1) in human hepatocyte models. Front Mol Biosci 2025; 12:1548582. [PMID: 39990870 PMCID: PMC11842253 DOI: 10.3389/fmolb.2025.1548582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Constitutive Photomorphogenesis Protein 1 homolog (COP1) is a conserved E3 ligase with key roles in several biological systems. Prior work in hepatocyte-derived tumors categorized COP1 as an oncogene, but its role in untransformed hepatocytes remains largely unexplored. Here, we have investigated the role of COP1 in primary human hepatocytes and two transformed hepatocyte models, HepG2 and HuH-7 cells. Methods The role of COP1 was tested by silencing and transduction experiments in HepG2, HuH-7, and primary human hepatocytes. Transcription array data of COP1-suppressed cells were generated and analyzed using clustering analyses. Cellular impacts were examined by proliferation assays, qRT-PCR, western blotting, reporter assays, and APOB enzyme-linked immunosorbent assays. Results and Discussion COP1 suppression had no noticeable impact on HepG2 and HuH-7 proliferation and was associated with contrasting rather than congruent transcriptome changes. Transcriptomic changes were consistent with perturbed metabolism in primary hepatocytes and HepG2 cells and impaired cell cycle regulation in HuH-7 cells. In HepG2 and primary hepatocytes but not in HuH-7 cells, COP1 suppression reduced the expression of important hepatic regulators and markers. COP1 downregulation reduced hepatic nuclear factor-4 alpha (HNF4A) abundance and function, as assessed by a lower abundance of key HNF4A targets, reduced APOB secretion, and reporter assays. HNF4A function could be restored by introducing a siRNA-resistant COP1 transgene, whereas HNF4A restoration partially rescued COP1 silencing in HepG2 cells. Our results identify and detail a pivotal regulatory role of COP1 in hepatocytes, in part through HNF4A.
Collapse
Affiliation(s)
| | - Paulina Lau
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
- Department of Medicine, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
4
|
Ma J, Mo J, Feng Y, Wang L, Jiang H, Li J, Jin C. Combination of transcriptomic and proteomic approaches helps unravel the mechanisms of luteolin in inducing liver cancer cell death via targeting AKT1 and SRC. Front Pharmacol 2024; 15:1450847. [PMID: 39234106 PMCID: PMC11371790 DOI: 10.3389/fphar.2024.1450847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Luteolin, a natural compound commonly used in traditional Chinese medicine, shows clinical potential as an anti-liver cancer agent. The mechanisms underlying the anti-liver cancer effect of luteolin are limited versus those reported for other cancers. Accordingly, this study was conducted to bridge the existing knowledge gap. Methods Transcriptomic and proteomic analyses of the response of the hepatocellular carcinoma cell line HuH-7 to luteolin were conducted, and a possible pathway was elucidated using confocal laser scanning microscopy (CLSM), flow cytometry, western blotting, qRT-PCR and bio-layer interferometry assay to systematically explore the possible mechanisms underlying the inhibition of the proliferation of liver cancer cells by luteolin. Results and Discussion Results showed that luteolin significantly inhibited HuH-7 cell proliferation. Transcriptomic and proteomic analyses collectively revealed that luteolin could promote cell cycle arrest and apoptosis in HuH-7 cells through transcription factors p53, nuclear factor kappa B (NF-κB), FOXO, ATF2, and TCF/LEF via AKT1, as well as the KEAP-NRF and SRC-STAT3 pathways. Furthermore, AKT1 and SRC were identified as the 2 targets of luteolin. Nuclear translocation of transcription factors p53 and NF-κB were affected by luteolin administration. Additionally, AKT1 activity affected normal metabolism in HuH-7 cells and resulted in the accumulation of reactive oxygen species, which activated MOMP and further promoted apoptosis. Our results systematically elucidate the mechanism of luteolin in inhibiting the proliferation of liver cancer cells, mainly through cell cycle arrest and apoptosis via targeting AKT1 and SRC.
Collapse
Affiliation(s)
- Junxia Ma
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Yifu Feng
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Junmin Li
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
5
|
Valente LC, Bacil GP, Riechelmann-Casarin L, Barbosa GC, Barbisan LF, Romualdo GR. Exploring in vitro modeling in hepatocarcinogenesis research: morphological and molecular features and similarities to the corresponding human disease. Life Sci 2024; 351:122781. [PMID: 38848937 DOI: 10.1016/j.lfs.2024.122781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/04/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The hepatocellular carcinoma (HCC) features a remarkable epidemiological burden, ranking as the third most lethal cancer worldwide. As the HCC-related molecular and cellular complexity unfolds as the disease progresses, the use of a myriad of in vitro models available is mandatory in translational preclinical research setups. In this review paper, we will compile cutting-edge information on the in vitro bioassays for HCC research, (A) emphasizing their morphological and molecular parallels with human HCC; (B) delineating the advantages and limitations of their application; and (C) offering perspectives on their prospective applications. While bidimensional (2D) (co) culture setups provide a rapid low-cost strategy for metabolism and drug screening investigations, tridimensional (3D) (co) culture bioassays - including patient-derived protocols as organoids and precision cut slices - surpass some of the 2D strategies limitations, mimicking the complex microarchitecture and cellular and non-cellular microenvironment observed in human HCC. 3D models have become invaluable tools to unveil HCC pathophysiology and targeted therapy. In both setups, the recapitulation of HCC in different etiologies/backgrounds (i.e., viral, fibrosis, and fatty liver) may be considered as a fundamental guide for obtaining translational findings. Therefore, a "multimodel" approach - encompassing the advantages of different in vitro bioassays - is encouraged to circumvent "model-biased" outcomes in preclinical HCC research.
Collapse
Affiliation(s)
- Leticia Cardoso Valente
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil
| | - Gabriel Prata Bacil
- São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Department of Structural and Functional Biology, Brazil
| | - Luana Riechelmann-Casarin
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil
| | | | - Luís Fernando Barbisan
- São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Department of Structural and Functional Biology, Brazil
| | - Guilherme Ribeiro Romualdo
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil.
| |
Collapse
|
6
|
Zhang R, He Z, Shi Y, Sun X, Chen X, Wang G, Zhang Y, Gao P, Wu Y, Lu S, Duan J, Sun S, Yang N, Fan W, Zhao K, Yang B, Xia Y, Zhang Y, Zhang Y, Yin H. Amplification editing enables efficient and precise duplication of DNA from short sequence to megabase and chromosomal scale. Cell 2024; 187:3936-3952.e19. [PMID: 38936359 DOI: 10.1016/j.cell.2024.05.056] [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: 12/25/2023] [Revised: 03/24/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
Duplication is a foundation of molecular evolution and a driver of genomic and complex diseases. Here, we develop a genome editing tool named Amplification Editing (AE) that enables programmable DNA duplication with precision at chromosomal scale. AE can duplicate human genomes ranging from 20 bp to 100 Mb, a size comparable to human chromosomes. AE exhibits activity across various cell types, encompassing diploid, haploid, and primary cells. AE exhibited up to 73.0% efficiency for 1 Mb and 3.4% for 100 Mb duplications, respectively. Whole-genome sequencing and deep sequencing of the junctions of edited sequences confirm the precision of duplication. AE can create chromosomal microduplications within disease-relevant regions in embryonic stem cells, indicating its potential for generating cellular and animal models. AE is a precise and efficient tool for chromosomal engineering and DNA duplication, broadening the landscape of precision genome editing from an individual genetic locus to the chromosomal scale.
Collapse
Affiliation(s)
- Ruiwen Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zhou He
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yajing Shi
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiangkun Sun
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinyu Chen
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Guoquan Wang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yizhou Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Pan Gao
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Ying Wu
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shuhan Lu
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Junyi Duan
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shangwu Sun
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Na Yang
- Center for Gene Diagnosis and Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wei Fan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China
| | - Bei Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China
| | - Yan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ying Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hao Yin
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
7
|
Nakamura H, Matsui T, Shinozawa T. Triclocarban induces lipid droplet accumulation and oxidative stress responses by inhibiting mitochondrial fatty acid oxidation in HepaRG cells. Toxicol Lett 2024; 396:11-18. [PMID: 38631510 DOI: 10.1016/j.toxlet.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
Mitochondrial fatty acid oxidation (mtFAO) plays an important role in hepatic energy metabolism. Severe mtFAO injury leads to nonalcoholic fatty liver disease (NAFLD) and liver failure. Several drugs have been withdrawn owing to safety issues, such as induction of fatty liver disease through mtFAO disruption. For instance, the antimicrobial triclocarban (TCC), an environmental contaminant that was removed from the market due to its unknown safety in humans, induces NAFLD in rats and promotes hepatic FAO in mice. Therefore, there are no consistent conclusions regarding the effects of TCC on FAO and lipid droplet accumulation. We hypothesized that TCC induces lipid droplet accumulation by inhibiting mtFAO in human hepatocytes. Here, we evaluated mitochondrial respiration in HepaRG cells to investigate the effects of TCC on fatty acid-driven oxidation in cells, electron transport chain parameters, lipid droplet accumulation, and antioxidant genes. The results suggest that TCC increases oxidative stress gene expression (GCLM, p62, HO-1, and NRF2) through lipid droplet accumulation via mtFAO inhibition in HepaRG cells. The results of the present study provide further insights into the effect of TCC on human NAFLD through mtFAO inhibition, and further in vivo studies could be used to validate the mechanisms.
Collapse
Affiliation(s)
- Hitoshi Nakamura
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited
| | - Toshikatsu Matsui
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited
| | - Tadahiro Shinozawa
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited.
| |
Collapse
|
8
|
Lee A, Simon AA, Boyreau A, Allain-Courtois N, Lambert B, Pradère JP, Saltel F, Cognet L. Identification of Early Stage Liver Fibrosis by Modifications in the Interstitial Space Diffusive Microenvironment Using Fluorescent Single-Walled Carbon Nanotubes. NANO LETTERS 2024; 24:5603-5609. [PMID: 38669477 DOI: 10.1021/acs.nanolett.4c00955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
During liver fibrosis, recurrent hepatic injuries lead to the accumulation of collagen and other extracellular matrix components in the interstitial space, ultimately disrupting liver functions. Early stages of liver fibrosis may be reversible, but opportunities for diagnosis at these stages are currently limited. Here, we show that the alterations of the interstitial space associated with fibrosis can be probed by tracking individual fluorescent single-walled carbon nanotubes (SWCNTs) diffusing in that space. In a mouse model of early liver fibrosis, we find that nanotubes generally explore elongated areas, whose lengths decrease as the disease progresses, even in regions where histopathological examination does not reveal fibrosis yet. Furthermore, this decrease in nanotube mobility is a purely geometrical effect as the instantaneous nanotube diffusivity stays unmodified. This work establishes the promise of SWCNTs both for diagnosing liver fibrosis at an early stage and for more in-depth studies of the biophysical effects of the disease.
Collapse
Affiliation(s)
- Antony Lee
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, 33400 Talence, France
- CNRS - UMR 5298, LP2N, Institut d'Optique Graduate School, 33400 Talence, France
- CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Institut Curie, Université PSL, Sorbonne Université, 75005 Paris, France
| | - Apolline A Simon
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, 33400 Talence, France
- CNRS - UMR 5298, LP2N, Institut d'Optique Graduate School, 33400 Talence, France
- CNRS, Bordeaux INP, ICMCB, UMR 5026, Univ. Bordeaux, 33600 Pessac, France
| | - Adeline Boyreau
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, 33400 Talence, France
- CNRS - UMR 5298, LP2N, Institut d'Optique Graduate School, 33400 Talence, France
| | - Nathalie Allain-Courtois
- Inserm, UMR1312, BRIC, Bordeaux Institute of Oncology, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Benjamin Lambert
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, 33400 Talence, France
- CNRS - UMR 5298, LP2N, Institut d'Optique Graduate School, 33400 Talence, France
| | - Jean-Philippe Pradère
- Institut RESTORE - UMR 1301 - Inserm/5070-CNRS/EFS, Univ. P. Sabatier, 31037 Toulouse, France
| | - Frédéric Saltel
- Inserm, UMR1312, BRIC, Bordeaux Institute of Oncology, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Laurent Cognet
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, 33400 Talence, France
- CNRS - UMR 5298, LP2N, Institut d'Optique Graduate School, 33400 Talence, France
| |
Collapse
|
9
|
Santagata S, Rea G, Castaldo D, Napolitano M, Capiluongo A, D'Alterio C, Trotta AM, Ieranò C, Portella L, Di Maro S, Tatangelo F, Albino V, Guarino R, Cutolo C, Izzo F, Scala S. Hepatocellular carcinoma (HCC) tumor microenvironment is more suppressive than colorectal cancer liver metastasis (CRLM) tumor microenvironment. Hepatol Int 2024; 18:568-581. [PMID: 37142825 PMCID: PMC11014815 DOI: 10.1007/s12072-023-10537-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/08/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND AND PURPOSE While HCC is an inflammation-associated cancer, CRLM develops on permissive healthy liver microenvironment. To evaluate the immune aspects of these two different environments, peripheral blood-(PB), peritumoral-(PT) and tumoral tissues-(TT) from HCC and CRLM patients were evaluated. METHODS 40 HCC and 34 CRLM were enrolled and freshly TT, PT and PB were collected at the surgery. PB-, PT- and TT-derived CD4+CD25+ Tregs, M/PMN-MDSC and PB-derived CD4+CD25- T-effector cells (Teffs) were isolated and characterized. Tregs' function was also evaluated in the presence of the CXCR4 inhibitor, peptide-R29, AMD3100 or anti-PD1. RNA was extracted from PB/PT/TT tissues and tested for FOXP3, CXCL12, CXCR4, CCL5, IL-15, CXCL5, Arg-1, N-cad, Vim, CXCL8, TGFβ and VEGF-A expression. RESULTS In HCC/CRLM-PB, higher number of functional Tregs, CD4+CD25hiFOXP3+ was detected, although PB-HCC Tregs exert a more suppressive function as compared to CRLM Tregs. In HCC/CRLM-TT, Tregs were highly represented with activated/ENTPD-1+Tregs prevalent in HCC. As compared to CRLM, HCC overexpressed CXCR4 and N-cadherin/vimentin in a contest rich in arginase and CCL5. Monocytic MDSCs were highly represented in HCC/CRLM, while high polymorphonuclear MDSCs were detected only in HCC. Interestingly, the function of CXCR4-PB-Tregs was impaired in HCC/CRLM by the CXCR4 inhibitor R29. CONCLUSION In HCC and CRLM, peripheral blood, peritumoral and tumoral tissues Tregs are highly represented and functional. Nevertheless, HCC displays a more immunosuppressive TME due to Tregs, MDSCs, intrinsic tumor features (CXCR4, CCL5, arginase) and the contest in which it develops. As CXCR4 is overexpressed in HCC/CRLM tumor/TME cells, CXCR4 inhibitors may be considered for double hit therapy in liver cancer patients.
Collapse
Affiliation(s)
- Sara Santagata
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Giuseppina Rea
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Daniela Castaldo
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Maria Napolitano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Anna Capiluongo
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Crescenzo D'Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Anna Maria Trotta
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Caterina Ieranò
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Salvatore Di Maro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100, Caserta, Italy
| | - Fabiana Tatangelo
- Pathology, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Vittorio Albino
- Divisions of Hepatobiliary Surgery, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Rita Guarino
- Divisions of Hepatobiliary Surgery, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Carmen Cutolo
- Divisions of Hepatobiliary Surgery, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Francesco Izzo
- Divisions of Hepatobiliary Surgery, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 80131, Naples, Italy.
| |
Collapse
|
10
|
Somatic XIST activation and features of X chromosome inactivation in male human cancers. Cell Syst 2022; 13:932-944.e5. [PMID: 36356577 DOI: 10.1016/j.cels.2022.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/09/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
Abstract
Expression of the non-coding RNA XIST is essential for initiating X chromosome inactivation (XCI) during early development in female mammals. As the main function of XCI is to enable dosage compensation of chromosome X genes between the sexes, XCI and XIST expression are generally absent in male normal tissues, except in germ cells and in individuals with supernumerary X chromosomes. Via a systematic analysis of public sequencing data of both cancerous and normal tissues, we report that XIST is somatically activated in a subset of male human cancers across diverse lineages. Some of these cancers display hallmarks of XCI, including silencing of gene expression, reduced chromatin accessibility, and increased DNA methylation across chromosome X, suggesting that the developmentally restricted, female-specific program of XCI can be somatically accessed in male cancers.
Collapse
|
11
|
Zinn E, Unzu C, Schmit PF, Turunen HT, Zabaleta N, Sanmiguel J, Fieldsend A, Bhatt U, Diop C, Merkel E, Gurrala R, Peacker B, Rios C, Messemer K, Santos J, Estelien R, Andres-Mateos E, Wagers AJ, Tipper C, Vandenberghe LH. Ancestral library identifies conserved reprogrammable liver motif on AAV capsid. Cell Rep Med 2022; 3:100803. [PMID: 36327973 PMCID: PMC9729830 DOI: 10.1016/j.xcrm.2022.100803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/18/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
Abstract
Gene therapy is emerging as a modality in 21st-century medicine. Adeno-associated viral (AAV) gene transfer is a leading technology to achieve efficient and durable expression of a therapeutic transgene. However, the structural complexity of the capsid has constrained efforts to engineer the particle toward improved clinical safety and efficacy. Here, we generate a curated library of barcoded AAVs with mutations across a variety of functionally relevant motifs. We then screen this library in vitro and in vivo in mice and nonhuman primates, enabling a broad, multiparametric assessment of every vector within the library. Among the results, we note a single residue that modulates liver transduction across all interrogated models while preserving transduction in heart and skeletal muscles. Moreover, we find that this mutation can be grafted into AAV9 and leads to profound liver detargeting while retaining muscle transduction-a finding potentially relevant to preventing hepatoxicities seen in clinical studies.
Collapse
Affiliation(s)
- Eric Zinn
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Carmen Unzu
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Pauline F Schmit
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Heikki T Turunen
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Nerea Zabaleta
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Julio Sanmiguel
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Allegra Fieldsend
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Urja Bhatt
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Cheikh Diop
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Erin Merkel
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Rakesh Gurrala
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Bryan Peacker
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher Rios
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Messemer
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer Santos
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Reynette Estelien
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Eva Andres-Mateos
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Amy J Wagers
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA; Joslin Diabetes Center, Boston, MA 02215, USA
| | - Christopher Tipper
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Bartolomeo CS, Lemes RMR, Morais RL, Pereria GC, Nunes TA, Costa AJ, de Barros Maciel RM, Braconi CT, Maricato JT, Janini LMR, Okuda LH, Lee KS, Prado CM, Ureshino RP, Stilhano RS. SARS-CoV-2 infection and replication kinetics in different human cell types: The role of autophagy, cellular metabolism and ACE2 expression. Life Sci 2022; 308:120930. [PMID: 36075471 PMCID: PMC9444585 DOI: 10.1016/j.lfs.2022.120930] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022]
Abstract
Aims This study evaluated SARS-CoV-2 replication in human cell lines derived from various tissues and investigated molecular mechanisms related to viral infection susceptibility and replication. Main methods SARS-CoV-2 replication in BEAS-2B and A549 (respiratory tract), HEK-293 T (kidney), HuH7 (liver), SH-SY5Y (brain), MCF7 (breast), Huvec (endothelial) and Caco-2 (intestine) was evaluated by RT-qPCR. Concomitantly, expression levels of ACE2 (Angiotensin Converting Enzyme) and TMPRSS2 were assessed through RT-qPCR and western blot. Proteins related to autophagy and mitochondrial metabolism were monitored in uninfected cells to characterize the cellular metabolism of each cell line. The effect of ACE2 overexpression on viral replication in pulmonary cells was also investigated. Key findings Our data show that HuH7, Caco-2 and MCF7 presented a higher viral load compared to the other cell lines. The increased susceptibility to SARS-CoV-2 infection seems to be associated not only with the differential levels of proteins intrinsically related to energetic metabolism, such as ATP synthase, citrate synthase, COX and NDUFS2 but also with the considerably higher TMPRSS2 mRNA expression. The two least susceptible cell types, BEAS-2B and A549, showed drastically increased SARS-CoV-2 replication capacity when ACE2 was overexpressed. These modified cell lines are relevant for studying SARS-CoV-2 replication in vitro. Significance Our data not only reinforce that TMPRSS2 expression and cellular energy metabolism are important molecular mechanisms for SARS-CoV-2 infection and replication, but also indicate that HuH7, MCF7 and Caco-2 are suitable models for mechanistic studies of COVID-19. Moreover, pulmonary cells overexpressing ACE2 can be used to understand mechanisms associated with SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Cynthia Silva Bartolomeo
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil; Department of Biosciences, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Robertha Mariana Rodrigues Lemes
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil; Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rafael Leite Morais
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gabriela Cruz Pereria
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Tamires Alves Nunes
- Department of Biosciences, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Angelica Jardim Costa
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rui Monteiro de Barros Maciel
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil; Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Carla Torres Braconi
- Department of Microbiology Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Juliana Terzi Maricato
- Department of Microbiology Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Luiz Mario Ramos Janini
- Department of Microbiology Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Liria Hiromi Okuda
- Instituto Biológico, Secretaria de Agricultura e Abastecimento, São Paulo, SP, Brazil
| | - Kil Sun Lee
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Carla Máximo Prado
- Department of Biosciences, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Rodrigo Portes Ureshino
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil; Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
13
|
Anaganti N, Chattopadhyay A, Poirier JT, Hussain MM. Generation of hepatoma cell lines deficient in microsomal triglyceride transfer protein. J Lipid Res 2022; 63:100257. [PMID: 35931202 PMCID: PMC9405095 DOI: 10.1016/j.jlr.2022.100257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/03/2022] [Accepted: 07/08/2022] [Indexed: 01/05/2023] Open
Abstract
The microsomal triglyceride transfer protein (MTP) is essential for the secretion of apolipoprotein B (apoB)48- and apoB100-containing lipoproteins in the intestine and liver, respectively. Loss of function mutations in MTP cause abetalipoproteinemia. Heterologous cells are used to evaluate the function of MTP in apoB secretion to avoid background MTP activity in liver and intestine-derived cells. However, these systems are not suitable to study the role of MTP in the secretion of apoB100-containing lipoproteins, as expression of a large apoB100 peptide using plasmids is difficult. Here, we report a new cell culture model amenable for studying the role of different MTP mutations on apoB100 secretion. The endogenous MTTP gene was ablated in human hepatoma Huh-7 cells using single guide RNA and RNA-guided clustered regularly interspaced short palindromic repeats-associated sequence 9 ribonucleoprotein complexes. We successfully established three different clones that did not express any detectable MTTP mRNA or MTP protein or activity. These cells were defective in secreting apoB-containing lipoproteins and accumulated lipids. Furthermore, we show that transfection of these cells with plasmids expressing human MTTP cDNA resulted in the expression of MTP protein, restoration of triglyceride transfer activity, and secretion of apoB100. Thus, these new cells can be valuable tools for studying structure-function of MTP, roles of different missense mutations in various lipid transfer activities of MTP, and their ability to support apoB100 secretion, compensatory changes associated with loss of MTP, and in the identification of novel proteins that may require MTP for their synthesis and secretion.
Collapse
Affiliation(s)
- Narasimha Anaganti
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Atrayee Chattopadhyay
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - John T Poirier
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
14
|
AL-Johani NS, Al-Zharani M, Aljarba NH, Alhoshani NM, Alkeraishan N, Alkahtani S. Antioxidant and Anti-Inflammatory Activities of Coenzyme-Q10 and Piperine against Cyclophosphamide-Induced Cytotoxicity in HuH-7 Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8495159. [PMID: 35872848 PMCID: PMC9300329 DOI: 10.1155/2022/8495159] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022]
Abstract
Cyclophosphamide (CP) alkylates DNA and RNA produce crosslinks that cause gene expression and protein synthesis inhibition to exert its anticancer effect. However, adverse effects of CP have restricted the CP application in cancer treatment. We investigate coenzyme-Q10 (Q10) and piperine (P) protective role on CP oxidant and inflammatory effect. HuH-7 cells were exposed to varying concentrations and combinations of Q10, P, and CP and evaluated intracellular ROS generation as well as inflammatory responses upon exposure. Our results showed Q10 and/or P suppressed both basal and CP-induced ROS generation without upsetting the balance in activities of SOD, catalase, and GSH levels. Analysis of proinflammatory cytokine gene expression showed that CP treatment alone only induced expression of IL-6β. However, coexposure of the cells to both Q10 and CP caused significant suppression of basal Cox-2 and TNF-α gene expression, while coexposure of the cells to CP and P with Co-Q10 suppressed basal IL-1β gene expression. Q10 also suppressed CP-induced expression of Cox-1. P and CP suppressed basal expression of IL-6β and IL-12β, while P and Q10 suppressed CP-induced IL1-α gene expression. Taken together, both Q10 and P seem to be inhibiting NFκβ pathway to suppress CP-mediated inflammation. In conclusion, Q10 and/or P induced suppression of ROS generation mediated by CP and also suppressed CP-induced inflammation by inhibiting expression of specific inflammatory cytokine.
Collapse
Affiliation(s)
- Norah S. AL-Johani
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammed Al-Zharani
- Imam Mohammad Ibn Saud Islamic University (IMSIU), College of Science, Biology Department, Riyadh 11623, Saudi Arabia
| | - Nada H. Aljarba
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P. O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Norah M. Alhoshani
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Nora Alkeraishan
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
15
|
Bär SI, Dittmer A, Nitzsche B, Ter-Avetisyan G, Fähling M, Klefenz A, Kaps L, Biersack B, Schobert R, Höpfner M. Chimeric HDAC and the cytoskeleton inhibitor broxbam as a novel therapeutic strategy for liver cancer. Int J Oncol 2022; 60:73. [PMID: 35485292 PMCID: PMC9097774 DOI: 10.3892/ijo.2022.5363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/11/2022] [Indexed: 12/24/2022] Open
Abstract
Broxbam, also known as N-hydroxy-4-{1-methoxy-4-[4′-(3′-bromo-4′,5′-dimethoxyphenyl)-oxazol-5′-yl]-2-phenoxy} butanamide, is a novel chimeric inhibitor that contains two distinct pharmacophores in its molecular structure. It has been previously demonstrated to inhibit the activity of histone deacetylases (HDAC) and tubulin polymerisation, two critical components required for cancer growth and survival. In the present study, the potential suitability of broxbam for the treatment of liver cancer was investigated. The effects of broxbam on cell proliferation and apoptosis, in addition to the under-lying molecular mechanism of action, were first investigated in primary liver cancer cell lines Huh7, HepG2, TFK1 and EGI1. Real-time proliferation measurements made using the iCEL-Ligence system and viable cell number counting following crystal violet staining) revealed that broxbam time- and dose-dependently reduced the proliferation of liver cancer cell lines with IC50 values <1 µM. In addition, a significant inhibition of the growth of hepatoblastoma microtumours on the chorioallantoic membranes (CAM) of fertilised chicken eggs by broxbam was observed according to results from the CAM assay, suggesting antineoplastic potency in vivo. Broxbam also exerted apoptotic effects through p53- and mitochondria-driven caspase-3 activation in Huh7 and HepG2 cells according to data from western blotting (p53 and phosphorylated p53), mitochondrial membrane potential measurements (JC-1 assay) and fluorometric capsase-3 measurements. Notably, no contribution of unspecific cytotoxic effects mediated by broxbam were observed from LDH-release measurements. HDAC1, -2, -4 and -6 expression was measured by western blotting and the HDAC inhibitory potency of broxbam was next evaluated using subtype-specific HDAC enzymatic assays, which revealed a largely pan-HDAC inhibitory activity with the most potent inhibition observed on HDAC6. Silencing HDAC6 expression in Huh7 cells led to a drop in the expression of the proliferation markers Ki-67 and E2F3, suggesting that HDAC6 inhibition by broxbam may serve a predomi-nant role in their antiproliferative effects on liver cancer cells. Immunofluorescence staining of cytoskeletal proteins (α-tubulin & actin) of broxbam-treated HepG2 cells revealed a pronounced inhibition of tubulin polymerisation, which was accompanied by reduced cell migration as determined by wound healing scratch assays. Finally, data from zebrafish angiogenesis assays revealed marked antiangiogenic effects of broxbam in vivo, as shown by the suppression of subintestinal vein growth in zebrafish embryos. To conclude, the pleiotropic anticancer activities of this novel chimeric HDAC- and tubulin inhibitor broxbam suggest that this compound is a promising candidate for liver cancer treatment, which warrants further pre-clinical and clinical evaluation.
Collapse
Affiliation(s)
- Sofia Isolde Bär
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Alexandra Dittmer
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Bianca Nitzsche
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Gohar Ter-Avetisyan
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Michael Fähling
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Adrian Klefenz
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Leonard Kaps
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Michael Höpfner
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| |
Collapse
|
16
|
Li L, Wang Q, He Y, Sun L, Yang Y, Pang X. Astragaloside IV suppresses migration and invasion of TGF-β 1-induced human hepatoma HuH-7 cells by regulating Nrf2/HO-1 and TGF-β 1/Smad3 pathways. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:397-405. [PMID: 35092472 DOI: 10.1007/s00210-021-02199-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
Astragaloside IV (AS-IV), one of the major compounds extract from Astragalus membranaceus, has shown attractive anti-cancer effects in certain malignancies. Oxidative stress (OS) is considered as a crucial factor in promoting the progression of hepatocellular carcinoma (HCC). In response to OS, nuclear factor erythroid 2-related factor 2 (Nrf2) upregulates and induces heme oxygenase 1 (HO-1) to combat oxidative damages. The phosphorylation of the COOH-terminal of Smad3 (pSmad3C) activates p21 to resist HCC progression, while the phosphorylation of the linker region of Smad3 (pSmad3L) up-regulates c-Myc transcription to exert promoting effect towards HCC. This study aimed to explore whether AS-IV suppresses migration and invasion of human hepatoma HuH-7 cells by regulating Nrf2/HO-1 and TGF-β1/Smad3 pathways. HuH-7 cells were induced with TGF-β1 (9 or 40 pM) to establish HCC model in vitro and pretreated with AS-IV at different concentration (5, 10, and 20 μM) for 24 h. Cell proliferation, migration, invasion, and intracellular reactive oxygen species (ROS) of HuH-7 cells were measured. The expression of Nrf2, pSmad3C, Nrf2/pNrf2, HO-1, pSmad3C/3L, c-Myc, and p21 were detected. Exposure of HuH-7 cells to TGF-β1 enhanced the cell proliferation, migration, invasion, and ROS production. Pretreatment with AS-IV (5, 10, and 20 μM) significantly reduced the cell proliferation, migration, invasion, and ROS production in HuH-7 cells. Furthermore, AS-IV increased the expressions of Nrf2/pNrf2, HO-1, pSmad3C, and p21, meanwhile reduced the expressions of pSmad3L and c-Myc. In conclusion, our study suggested that AS-IV inhibit HuH-7 cells migration and invasion, which related to activate Nrf2/HO-1 pathway, up-regulation pSmad3C/p21 pathway, and down-regulation pSmad3L/c-Myc pathway. The present research supports the notion that AS-IV may be a latent agent for the treatment of HCC.
Collapse
Affiliation(s)
- Lili Li
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Qin Wang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yinghao He
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Liangjie Sun
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yan Yang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Xiaonan Pang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
- Department of Oncology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
17
|
Oncogenic Mutation BRAF V600E Changes Phenotypic Behavior of THLE-2 Liver Cells through Alteration of Gene Expression. Int J Mol Sci 2022; 23:ijms23031548. [PMID: 35163468 PMCID: PMC8836259 DOI: 10.3390/ijms23031548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/10/2022] Open
Abstract
The accumulation of mutations in cancer driver genes, such as tumor suppressors or proto-oncogenes, affects cellular homeostasis. Disturbances in the mechanism controlling proliferation cause significant augmentation of cell growth and division due to the loss of sensitivity to the regulatory signals. Nowadays, an increasing number of cases of liver cancer are observed worldwide. Data provided by the International Cancer Genome Consortium (ICGC) have indicated many alterations within gene sequences, whose roles in tumor development are not well understood. A comprehensive analysis of liver cancer (virus-associated hepatocellular carcinoma) samples has identified new and rare mutations in B-Raf proto-oncogene (BRAF) in Japanese HCC patients, as well as BRAF V600E mutations in French HCC patients. However, their function in liver cancer has never been investigated. Here, using functional analysis and next generation sequencing, we demonstrate the tumorigenic effect of BRAF V600E on hepatocytes (THLE-2 cell line). Moreover, we identified genes such as BMP6, CXCL11, IL1B, TBX21, RSAD2, MMP10, and SERPIND1, which are possibly regulated by the BRAF V600E-mediated, mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) signaling pathway. Through several functional assays, we demonstrate that BRAF L537M, D594A, and E648G mutations alone are not pathogenic in liver cancer. The investigation of genome mutations and the determination of their impact on cellular processes and functions is crucial to unraveling the molecular mechanisms of liver cancer development.
Collapse
|
18
|
The Curious Case of the HepG2 Cell Line: 40 Years of Expertise. Int J Mol Sci 2021; 22:13135. [PMID: 34884942 PMCID: PMC8658661 DOI: 10.3390/ijms222313135;select dbms_pipe.receive_message(chr(115)||chr(108)||chr(113)||chr(84),5) from dual--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Liver cancer is the third leading cause of cancer death worldwide. Representing such a dramatic impact on our lives, liver cancer is a significant public health concern. Sustainable and reliable methods for preventing and treating liver cancer require fundamental research on its molecular mechanisms. Cell lines are treated as in vitro equivalents of tumor tissues, making them a must-have for basic research on the nature of cancer. According to recent discoveries, certified cell lines retain most genetic properties of the original tumor and mimic its microenvironment. On the other hand, modern technologies allowing the deepest level of detail in omics landscapes have shown significant differences even between samples of the same cell line due to cross- and mycoplasma infection. This and other observations suggest that, in some cases, cell cultures are not suitable as cancer models, with limited predictive value for the effectiveness of new treatments. HepG2 is a popular hepatic cell line. It is used in a wide range of studies, from the oncogenesis to the cytotoxicity of substances on the liver. In this regard, we set out to collect up-to-date information on the HepG2 cell line to assess whether the level of heterogeneity of the cell line allows in vitro biomedical studies as a model with guaranteed production and quality.
Collapse
|
19
|
The Curious Case of the HepG2 Cell Line: 40 Years of Expertise. Int J Mol Sci 2021; 22:13135. [PMID: 34884942 PMCID: PMC8658661 DOI: 10.3390/ijms222313135;select dbms_pipe.receive_message(chr(80)||chr(106)||chr(79)||chr(120),5) from dual--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Liver cancer is the third leading cause of cancer death worldwide. Representing such a dramatic impact on our lives, liver cancer is a significant public health concern. Sustainable and reliable methods for preventing and treating liver cancer require fundamental research on its molecular mechanisms. Cell lines are treated as in vitro equivalents of tumor tissues, making them a must-have for basic research on the nature of cancer. According to recent discoveries, certified cell lines retain most genetic properties of the original tumor and mimic its microenvironment. On the other hand, modern technologies allowing the deepest level of detail in omics landscapes have shown significant differences even between samples of the same cell line due to cross- and mycoplasma infection. This and other observations suggest that, in some cases, cell cultures are not suitable as cancer models, with limited predictive value for the effectiveness of new treatments. HepG2 is a popular hepatic cell line. It is used in a wide range of studies, from the oncogenesis to the cytotoxicity of substances on the liver. In this regard, we set out to collect up-to-date information on the HepG2 cell line to assess whether the level of heterogeneity of the cell line allows in vitro biomedical studies as a model with guaranteed production and quality.
Collapse
|
20
|
Arzumanian VA, Kiseleva OI, Poverennaya EV. The Curious Case of the HepG2 Cell Line: 40 Years of Expertise. Int J Mol Sci 2021; 22:13135. [PMID: 34884942 PMCID: PMC8658661 DOI: 10.3390/ijms222313135] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is the third leading cause of cancer death worldwide. Representing such a dramatic impact on our lives, liver cancer is a significant public health concern. Sustainable and reliable methods for preventing and treating liver cancer require fundamental research on its molecular mechanisms. Cell lines are treated as in vitro equivalents of tumor tissues, making them a must-have for basic research on the nature of cancer. According to recent discoveries, certified cell lines retain most genetic properties of the original tumor and mimic its microenvironment. On the other hand, modern technologies allowing the deepest level of detail in omics landscapes have shown significant differences even between samples of the same cell line due to cross- and mycoplasma infection. This and other observations suggest that, in some cases, cell cultures are not suitable as cancer models, with limited predictive value for the effectiveness of new treatments. HepG2 is a popular hepatic cell line. It is used in a wide range of studies, from the oncogenesis to the cytotoxicity of substances on the liver. In this regard, we set out to collect up-to-date information on the HepG2 cell line to assess whether the level of heterogeneity of the cell line allows in vitro biomedical studies as a model with guaranteed production and quality.
Collapse
|
21
|
Ozten O, Guney Eskiler G, Sonmez F, Yıldız MZ. Investigation of the therapeutic effect of 5-aminolevulinic acid based photodynamic therapy on hepatocellular carcinoma. Lasers Med Sci 2021; 37:1325-1332. [PMID: 34392466 DOI: 10.1007/s10103-021-03398-8] [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: 04/24/2021] [Accepted: 08/06/2021] [Indexed: 11/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous type of cancer and current treatment options limit successful therapy outcomes. Photodynamic therapy (PDT) has attracted attention as an alternative approach in the treatment of different types of cancer. However, there is no study in the literature regarding the effect of PDT on HCC, in vitro. Therefore, the aim of this study was to determine the cytotoxic and apoptotic effects of 5-aminolevulinic acid (5-ALA)/PDT on two different HCC cell lines in terms of hepatitis B virus (HBV) infection. The therapeutic effects of 5-ALA-based PDT on HCC cell lines (Huh-7 and SNU-449) were evaluated by PpIX-fluorescence accumulation, WST-1 analysis, Annexin V analysis, and acridine orange/ethidium bromide staining after irradiation with different light doses through diode laser. The results showed that 1 mM 5-ALA displayed higher PpIX fluorescence in the SNU-449 cell line than the Huh-7 cell line after 4 h of incubation. After irradiation with different light doses (3, 6, 9, and 12 J/cm2), 5-ALA significantly reduced the proliferation of HCC cells and induced apoptotic cell death (p < 0.01). Furthermore, SNU-449 cells were more responsive to 5-ALA-based PDT than Huh-7 cells due to possibly its molecular features as well as viral HBV status. Our preliminary data obtained from this study may contribute to the development of 5-ALA/PDT-based treatment strategies in the treatment of HCC. However, this study could be improved by the elucidation of the molecular mechanisms of cell death induced by 5-ALA/PDT in HCC cells, the use of different photosensitizer, light sources, and in vivo experiments.
Collapse
Affiliation(s)
- Ozge Ozten
- Department of Biomedical Engineering, Institute of Graduate Education, Sakarya University of Applied Sciences, 54187, Sakarya, Turkey
| | - Gamze Guney Eskiler
- Faculty of Medicine, Department of Medical Biology, Sakarya University, Korucuk Campus, 54290, Sakarya, Turkey.
| | - Fatih Sonmez
- Pamukova Vocational High School, Sakarya University of Applied Sciences, 54055, Sakarya, Turkey
| | - Mustafa Zahid Yıldız
- Faculty of Technology, Department of Electrical Electronics Engineering, Sakarya University of Applied Sciences, 54187, Sakarya, Turkey
| |
Collapse
|
22
|
Papachristou F, Anninou N, Koukoulis G, Paraskakis S, Sertaridou E, Tsalikidis C, Pitiakoudis M, Simopoulos C, Tsaroucha A. Differential effects of cisplatin combined with the flavonoid apigenin on HepG2, Hep3B, and Huh7 liver cancer cell lines. Mutat Res 2021; 866:503352. [PMID: 33985696 DOI: 10.1016/j.mrgentox.2021.503352] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
The potential of apigenin (APG) to enhance cisplatin's (CDDP) chemotherapeutic efficacy was investigated in HepG2, Hep3B, and Huh7 liver cancer cell lines. The presence of 20 μM APG sensitized all cell lines to CDDP treatment (degree of sensitization based on the MTT assay: HepG2>Huh7>Hep3B). As reflected by sister chromatid exchange levels, the degree of genetic instability as well as DNA repair by homologous recombination differed among cell lines. CDDP and 20 μM APG cotreatment exhibited a synergistic genotoxic effect on Hep3B cells and a less than additive effect on HepG2 and Huh7 cells. Cell cycle delays were noticed during the first mitotic division in Hep3B and Huh7 cells and the second mitotic division in HepG2 cells. CDDP and CDDP + APG treatments reduced the clonogenic capacity of all cell lines; however, there was a discordance in drug sensitivity compared with the MMT assay. Furthermore, a senescence-like phenotype was induced, especially in Hep3B and Huh7 cells. Unlike CDDP monotherapy, the combined treatment exhibited a significant anti-invasive and anti-migratory action in all cancer cell lines. The fact that the three liver cancer cell lines responded differently, yet positively, to CDDP + APG cotreatment could be attributed to variations they present in gene expression. Complex mechanisms seem to influence cellular responses and cell fate.
Collapse
Affiliation(s)
- Fotini Papachristou
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece; Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece.
| | - Nikolia Anninou
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Georgios Koukoulis
- Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Stefanos Paraskakis
- Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Eleni Sertaridou
- Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Christos Tsalikidis
- Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Michael Pitiakoudis
- Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Constantinos Simopoulos
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece; Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| | - Alexandra Tsaroucha
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece; Postgraduate Program in Hepatobiliary and Pancreatic Surgery, 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, 68 100, Greece
| |
Collapse
|
23
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
24
|
Kawamoto M, Yamaji T, Saito K, Shirasago Y, Satomura K, Endo T, Fukasawa M, Hanada K, Osada N. Identification of Characteristic Genomic Markers in Human Hepatoma HuH-7 and Huh7.5.1-8 Cell Lines. Front Genet 2020; 11:546106. [PMID: 33193621 PMCID: PMC7581915 DOI: 10.3389/fgene.2020.546106] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022] Open
Abstract
The human hepatoma-derived HuH-7 cell line and its derivatives (Huh7.5 and Huh7.5.1) have been widely used as a convenient experimental substitute for primary hepatocytes. In particular, these cell lines represent host cells suitable for propagating the hepatitis C virus (HCV) in vitro. The Huh7.5.1-8 cell line, a subline of Huh7.5.1, can propagate HCV more efficiently than its parental cells. To provide genomic information for cells' quality control, we performed whole-genome sequencing of HuH-7 and Huh7.5.1-8 and identified their characteristic genomic deletions, some of which are applicable to an in-house test for cell authentication. Among the genes related to HCV infection and replication, 53 genes were found to carry missense or loss-of-function mutations likely specific to the HuH-7 and/or Huh7.5.1-8. Eight genes, including DDX58 (RIG-I), BAX, EP300, and SPP1 (osteopontin), contained mutations observed only in Huh7.5.1-8 or mutations with higher frequency in Huh7.5.1-8. These mutations might be relevant to phenotypic differences between the two cell lines and may also serve as genetic markers to distinguish Huh7.5.1-8 cells from the ancestral HuH-7 cells.
Collapse
Affiliation(s)
- Masaki Kawamoto
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Toshiyuki Yamaji
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kyoko Saito
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshitaka Shirasago
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kazuhiro Satomura
- Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Toshinori Endo
- Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Masayoshi Fukasawa
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kentaro Hanada
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naoki Osada
- Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan.,Global Station for Big Data and Cybersecurity, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| |
Collapse
|
25
|
Sanclemente-Alaman I, Moreno-Jiménez L, Benito-Martín MS, Canales-Aguirre A, Matías-Guiu JA, Matías-Guiu J, Gómez-Pinedo U. Experimental Models for the Study of Central Nervous System Infection by SARS-CoV-2. Front Immunol 2020; 11:2163. [PMID: 32983181 PMCID: PMC7485091 DOI: 10.3389/fimmu.2020.02163] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The response to the SARS-CoV-2 coronavirus epidemic requires increased research efforts to expand our knowledge of the disease. Questions related to infection rates and mechanisms, the possibility of reinfection, and potential therapeutic approaches require us not only to use the experimental models previously employed for the SARS-CoV and MERS-CoV coronaviruses but also to generate new models to respond to urgent questions. DEVELOPMENT We reviewed the different experimental models used in the study of central nervous system (CNS) involvement in COVID-19 both in different cell lines that have enabled identification of the virus' action mechanisms and in animal models (mice, rats, hamsters, ferrets, and primates) inoculated with the virus. Specifically, we reviewed models used to assess the presence and effects of SARS-CoV-2 on the CNS, including neural cell lines, animal models such as mouse hepatitis virus CoV (especially the 59 strain), and the use of brain organoids. CONCLUSION Given the clear need to increase our understanding of SARS-CoV-2, as well as its potential effects on the CNS, we must endeavor to obtain new information with cellular or animal models, with an appropriate resemblance between models and human patients.
Collapse
Affiliation(s)
- Inmaculada Sanclemente-Alaman
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| | - Lidia Moreno-Jiménez
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| | - María Soledad Benito-Martín
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Canales-Aguirre
- Preclinical Evaluation Unit, Medical and Pharmaceutical Biotechnology, CIATEJ-CONACYT, Guadalajara, Mexico
| | - Jordi A. Matías-Guiu
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| | - Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Institute for Health Research, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
26
|
Pramfalk C, Jakobsson T, Verzijl CRC, Minniti ME, Obensa C, Ripamonti F, Olin M, Pedrelli M, Eriksson M, Parini P. Generation of new hepatocyte-like in vitro models better resembling human lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158659. [PMID: 32058035 DOI: 10.1016/j.bbalip.2020.158659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 11/21/2022]
Abstract
In contrast to human hepatocytes in vivo, which solely express acyl-coenzyme A:cholesterol acyltransferase (ACAT) 2, both ACAT1 and ACAT2 (encoded by SOAT1 and SOAT2) are expressed in primary human hepatocytes and in human hepatoma cell lines. Here, we aimed to create hepatocyte-like cells expressing the ACAT2, but not the ACAT1, protein to generate a model that - at least in this regard - resembles the human condition in vivo and to assess the effects on lipid metabolism. Using the Clustered Regularly Interspaced Short Palindromic Repeats technology, we knocked out SOAT1 in HepG2 and Huh7.5 cells. The wild type and SOAT2-only-cells were cultured with fetal bovine or human serum and the effects on lipoprotein and lipid metabolism were studied. In SOAT2-only-HepG2 cells, increased levels of cholesterol, triglycerides, apolipoprotein B and lipoprotein(a) in the cell media were detected; this was likely dependent of the increased expression of key genes involved in lipid metabolism (e.g. MTP, APOB, HMGCR, LDLR, ACACA, and DGAT2). Opposite effects were observed in SOAT2-only-Huh7.5 cells. Our study shows that the expression of SOAT1 in hepatocyte-like cells contributes to the distorted phenotype observed in HepG2 and Huh7.5 cells. As not only parameters of lipoprotein and lipid metabolism but also some markers of differentiation/maturation increase in the SOAT2-only-HepG2 cells cultured with HS, this cellular model represent an improved model for studies of lipid metabolism.
Collapse
Affiliation(s)
- Camilla Pramfalk
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Jakobsson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Cristy R C Verzijl
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Mirko E Minniti
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Clara Obensa
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Federico Ripamonti
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Maria Olin
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Mats Eriksson
- Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden; Metabolism Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Paolo Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden; Metabolism Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
27
|
Tabata Y, Shidoji Y. Hepatic monoamine oxidase B is involved in endogenous geranylgeranoic acid synthesis in mammalian liver cells. J Lipid Res 2020; 61:778-789. [PMID: 32094232 PMCID: PMC7193968 DOI: 10.1194/jlr.ra119000610] [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: 12/31/2019] [Revised: 02/15/2020] [Indexed: 12/16/2022] Open
Abstract
Geranylgeranoic acid (GGA) originally was identified in some animals and has been developed as an agent for preventing second primary hepatoma. We previously have also identified GGA as an acyclic diterpenoid in some medicinal herbs. Recently, we reported that in human hepatoma-derived HuH-7 cells, GGA is metabolically labeled from 13C-mevalonate. Several cell-free experiments have demonstrated that GGA is synthesized through geranylgeranial by oxygen-dependent oxidation of geranylgeraniol (GGOH), but the exact biochemical events giving rise to GGA in hepatoma cells remain unclear. Monoamine oxidase B (MOAB) has been suggested to be involved in GGOH oxidation. Here, using two human hepatoma cell lines, we investigated whether MAOB contributes to GGA biosynthesis. Using either HuH-7 cell lysates or recombinant human MAOB, we found that: 1) the MAO inhibitor tranylcypromine dose-dependently downregulates endogenous GGA levels in HuH-7 cells; and 2) siRNA-mediated MAOB silencing reduces intracellular GGA levels in HuH-7 and Hep3B cells. Unexpectedly, however, CRISPR/Cas9-generated MAOB-KO human hepatoma Hep3B cells had GGA levels similar to those in MAOB-WT cells. A sensitivity of GGA levels to siRNA-mediated MAOB downregulation was recovered when the MAOB-KO cells were transfected with a MAOB-expression plasmid, suggesting that MAOB is the enzyme primarily responsible for GGOH oxidation and that some other latent metabolic pathways may maintain endogenous GGA levels in the MAOB-KO hepatoma cells. Along with the previous findings, these results provide critical insights into the biological roles of human MAOB and provide evidence that hepatic MAOB is involved in endogenous GGA biosynthesis via GGOH oxidation.
Collapse
Affiliation(s)
- Yuki Tabata
- Molecular and Cellular Biology, Graduate School of Human Health Science, University of Nagasaki, Nagayo, Nagasaki 851-2195, Japan
| | - Yoshihiro Shidoji
- Molecular and Cellular Biology, Graduate School of Human Health Science, University of Nagasaki, Nagayo, Nagasaki 851-2195, Japan. mailto:
| |
Collapse
|
28
|
Morgan K, Gamal W, Samuel K, Morley SD, Hayes PC, Bagnaninchi P, Plevris JN. Application of Impedance-Based Techniques in Hepatology Research. J Clin Med 2019; 9:jcm9010050. [PMID: 31878354 PMCID: PMC7019217 DOI: 10.3390/jcm9010050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
There are a variety of end-point assays and techniques available to monitor hepatic cell cultures and study toxicity within in vitro models. These commonly focus on one aspect of cell metabolism and are often destructive to cells. Impedance-based cellular assays (IBCAs) assess biological functions of cell populations in real-time by measuring electrical impedance, which is the resistance to alternating current caused by the dielectric properties of proliferating of cells. While the uses of IBCA have been widely reported for a number of tissues, specific uses in the study of hepatic cell cultures have not been reported to date. IBCA monitors cellular behaviour throughout experimentation non-invasively without labelling or damage to cell cultures. The data extrapolated from IBCA can be correlated to biological events happening within the cell and therefore may inform drug toxicity studies or other applications within hepatic research. Because tight junctions comprise the blood/biliary barrier in hepatocytes, there are major consequences when these junctions are disrupted, as many pathologies centre around the bile canaliculi and flow of bile out of the liver. The application of IBCA in hepatology provides a unique opportunity to assess cellular polarity and patency of tight junctions, vital to maintaining normal hepatic function. Here, we describe how IBCAs have been applied to measuring the effect of viral infection, drug toxicity /IC50, cholangiopathies, cancer metastasis and monitoring of the gut-liver axis. We also highlight key areas of research where IBCAs could be used in future applications within the field of hepatology.
Collapse
Affiliation(s)
- Katie Morgan
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
- Correspondence:
| | - Wesam Gamal
- James Nasmyth Building, Institute of Mechanical, Process and Energy Engineering, Heriot-Watt University School of Engineering and Physical Sciences, Edinburgh EH14 4AS, UK;
| | - Kay Samuel
- The Jack Copland Centre, Advanced Therapeutics, Scottish National Blood Transfusion Service, 52 Research Avenue North, Edinburgh EH14 4BE, UK;
| | - Steven D. Morley
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Peter C. Hayes
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Pierre Bagnaninchi
- MRC Centre for Regenerative Medicine 5 Little France Drive, Edinburgh EH16 4UU, UK;
| | - John N. Plevris
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| |
Collapse
|
29
|
Establishment and Characterization of a New Cell Line Permissive for Hepatitis C Virus Infection. Sci Rep 2019; 9:7943. [PMID: 31138826 PMCID: PMC6538753 DOI: 10.1038/s41598-019-44257-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 03/12/2019] [Indexed: 01/02/2023] Open
Abstract
Hepatitis C virus (HCV) cell culture systems have facilitated the development of efficient direct-acting antivirals against HCV. Huh-7.5, a subline of the human hepatoma cell line Huh-7, has been used widely to amplify HCV because HCV can efficiently replicate in these cells due to a defect in innate antiviral signalling. Recently, we established a novel cell line, KH, derived from human hepatocellular carcinoma, which showed atypical uptake of gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA) in a Gd-EOB-DTPA-enhanced magnetic resonance imaging study. KH cells expressed hepatocyte markers including microRNA-122 (miR-122) at a lower level than Huh-7.5 cells. We demonstrated that KH cells could support the entire life cycle of HCV; however, HCV replicated at a lower rate in KH cells compared to Huh-7.5 cells, and virus particles produced from KH cells seemed to have some disadvantages in viral assembly compared with those produced from Huh-7.5 cells. KH cells had more robust interferon-stimulated gene expression and induction upon HCV RNA transfection, interferon-α2b addition, and HCV infection than Huh-7.5 cells. Interestingly, both miR-122 supplementation and IRF3 knockout in KH cells boosted HCV replication to a similar level as in Huh-7.5 cells, suggesting that intact innate antiviral signalling and lower miR-122 expression limit HCV replication in KH cells. KH cells will enable a deeper understanding of the role of the innate immune response in persistent HCV infection.
Collapse
|
30
|
Zhao L, Fu L, Xu Z, Fan R, Xu R, Fu R, Zou S, Wang C, Zhang Y, Wang J, Bao J, Wang Z, Hou X, Zheng Y, Dai E, Wang F. The anticancer effects of cinobufagin on hepatocellular carcinoma Huh‑7 cells are associated with activation of the p73 signaling pathway. Mol Med Rep 2019; 19:4119-4128. [PMID: 30942456 PMCID: PMC6471725 DOI: 10.3892/mmr.2019.10108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
The Na+/K+-ATPase inhibitor cinobufagin exhibits numerous anticancer effects on hepatocellular carcinoma (HCC) cells expressing wild-type p53 via inhibition of aurora kinase A (AURKA) and activation of p53 signaling. However, the effects of cinobufagin on HCC cells expressing mutant p53 remain unclear. In the present study, the anticancer effects of cinobufagin were investigated on HCC Huh-7 cells with mutant p53, and the effects of AURKA overexpression or inhibition on the anticancer effects of cinobufagin were analyzed. Viability, cell cycle progression and apoptosis of cells were determined using an MTT assay, flow cytometry and Hoechst 33342 staining, respectively. The expression levels of p53 and p73 signaling-associated proteins were investigated via western blot analysis. The results demonstrated that the expression levels of AURKA, B-cell lymphoma 2 (Bcl-2), cyclin-dependent kinase 1, cyclin B1, proliferating cell nuclear antigen and heterogeneous nuclear ribonucleoprotein K, as well as the phosphorylation of p53 and mouse double minute 2 homolog, were significantly decreased in Huh-7 cells treated with 5 µmol/l cinobufagin for 24 h. Conversely, the expression levels of Bcl-2-associated X protein, p21, p53 upregulated modulator of apoptosis and phorbol-12-myristate-13-acetate-induced protein 1, were significantly increased by cinobufagin treatment. Overexpression or inhibition of AURKA suppressed or promoted the anticancer effects of cinobufagin on Huh-7 cells, respectively. These results indicated that cinobufagin may induce anticancer effects on Huh-7 cells via the inhibition of AURKA and p53 signaling, and via the activation of p73 signaling, in an AURKA-dependent manner.
Collapse
Affiliation(s)
- Lei Zhao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Lina Fu
- Department of Gastroenterology, Tianjin Fourth Central Hospital, Tianjin 300140, P.R. China
| | - Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Rong Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Ruicheng Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Rong Fu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Congcong Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Jiabao Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Jun Bao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Zhimei Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Xiaojie Hou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Yupiao Zheng
- Department of Gastroenterology and Hepatology, The Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| | - Erqing Dai
- Hepatology Department of Pingjin Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Fengmei Wang
- Department of Gastroenterology and Hepatology, The Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| |
Collapse
|