1
|
Jiang H, Foroozandeh P, Kaplan N, Xu D, Yang W, Qi X, Nalbant EK, Clutter ED, Zhu Y, Xu J, Schipma MJ, Ren Z, Peng H. IFITM1/OVOL1 Axis Is a Novel Regulator of the Expansion of the Limbal Epithelial Stem/Early Transient Amplifying Cell Population. FASEB J 2025; 39:e70648. [PMID: 40372397 DOI: 10.1096/fj.202500783r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/18/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Limbal epithelial stem cells (LESCs), located in the basal layer of the limbal epithelium, rarely proliferate under normal conditions. Upon proliferation, LESCs give rise to early transient amplifying (eTA) cells, which are thought to be morphologically and phenotypically indistinguishable from LESCs. Following corneal epithelial wounding, LESCs are activated to repair the corneal epithelium via expansion of eTA cells, a process crucial for maintaining corneal epithelial homeostasis and tissue transparency as well as essential for clear vision. To understand how this process is regulated, we conducted a single cell RNA sequencing assay of mouse corneal rims with and without injury and observed an expansion of the stem/eTA cell cluster after corneal injury. Interestingly, we found that Interferon Induced Transmembrane Protein 1 (IFITM1) was predominantly expressed in stem/eTA cells and was positively associated with such stem/eTA cell expansion after corneal wounding. In vivo knockdown of IFITM1 using an AAV (adeno-associated virus) vector significantly attenuated stem/eTA cell expansion and activation of stem/eTA cells to proliferate after mouse corneal wounding. In human limbal epithelial cell cultures, IFITM1 positively impacted the proliferation of stem/eTA cell-enriched limbal epithelial cells, contributing to expansion of the stem/eTA cell population. Such expansion was due, in part, to inhibition of OVOL1 (Ovo like zinc finger 1), a negative regulator of epithelial cell proliferation. These results provide key molecular insights into how stem cell activation and eTA cell expansion are regulated. Elucidating the IFITM1/OVOL1 pathway that governs stem/eTA cell proliferation not only deepens our knowledge of tissue homeostasis but also opens avenues for developing novel regenerative therapies.
Collapse
Affiliation(s)
- Huimin Jiang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Parisa Foroozandeh
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nihal Kaplan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dan Xu
- Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wending Yang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xiaolin Qi
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elif Kayaalp Nalbant
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elwin D Clutter
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yongling Zhu
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jian Xu
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Matthew John Schipma
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Han Peng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
2
|
Sun J, Xu H, Li B, Deng W, Han X, Zhong X, Zhu J, Jiang Y, Wang Z, Zhang D, Sun G. IFITM1 aggravates ConA-Induced autoimmune hepatitis by promoting NKT cell activation through increased AMPK-Dependent mitochondrial function. Int Immunopharmacol 2025; 144:113692. [PMID: 39602958 DOI: 10.1016/j.intimp.2024.113692] [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: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Although interferon-induced transmembrane 1 (IFITM1) is known for its crucial role in antiviral immunity, its involvement in autoimmune hepatitis (AIH) remains largely unexplored. In this study, we observed that IFITM1 expression is markedly upregulated in a Concanavalin A (ConA)-induced AIH model, with particularly high and markedly elevated expression in natural killer T (NKT) cells. To further understand the role of IFITM1, we examined the responses of IFITM1-/- mice in a model of ConA-induced liver injury. In comparison to wild-type mice, IFITM1-/- mice exhibited reduced sensitivity in this model, as evidenced by significantly ameliorated necrosis areas, lower serum aminotransferase levels, a reduced number of intrahepatic NKT cells, and decreased expression of inflammatory factors, such as IL-1β, IL-6, IFN-γ and TNF-α. Notably, by using IFITM1-GFP mice and IFITM1-/- mice, we demonstrated that IFITM1 expression in NKT cells is crucial for their proliferation, proinflammatory cytokine production, and cytotoxic functions. Furthermore, analysis of single-cell RNA sequencingdata revealed that IFITM1 is essential for mitochondrial function, which is mediated by the AMP-activated protein kinase (AMPK) pathway. We also validated the importance of IFITM1 for the AMPK pathway and mitochondrial ATP synthesis in vivo. Together, our findings elucidate that IFITM1 could regulate NKT cell activation and survival by promoting mitochondrial function during AIH.
Collapse
Affiliation(s)
- Jie Sun
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Haozhe Xu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Buer Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wanqing Deng
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaotong Han
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xinjie Zhong
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jingjing Zhu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuan Jiang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zeyu Wang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Dong Zhang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guangyong Sun
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
3
|
Chi H, Qu B, Prawira A, Richardt T, Maurer L, Hu J, Fu RM, Lempp FA, Zhang Z, Grimm D, Wu X, Urban S, Dao Thi VL. An hepatitis B and D virus infection model using human pluripotent stem cell-derived hepatocytes. EMBO Rep 2024; 25:4311-4336. [PMID: 39232200 PMCID: PMC11466959 DOI: 10.1038/s44319-024-00236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Current culture systems available for studying hepatitis D virus (HDV) are suboptimal. In this study, we demonstrate that hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) are fully permissive to HDV infection across various tested genotypes. When co-infected with the helper hepatitis B virus (HBV) or transduced to express the HBV envelope protein HBsAg, HLCs effectively release infectious progeny virions. We also show that HBsAg-expressing HLCs support the extracellular spread of HDV, thus providing a valuable platform for testing available anti-HDV regimens. By challenging the cells along the differentiation with HDV infection, we have identified CD63 as a potential HDV co-entry factor that was rate-limiting for HDV infection in immature hepatocytes. Given their renewable source and the potential to derive hPSCs from individual patients, we propose HLCs as a promising model for investigating HDV biology. Our findings offer new insights into HDV infection and expand the repertoire of research tools available for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Huanting Chi
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Bingqian Qu
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Angga Prawira
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Talisa Richardt
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Lars Maurer
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- Department of Infectious Diseases, Virology, Section Viral Vector Technologies, University Hospital Heidelberg, Cluster of Excellence CellNetworks, BioQuant, Center for Integrative Infectious Diseases Research (CIID), Heidelberg, Germany
| | - Jungen Hu
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Rebecca M Fu
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Florian A Lempp
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- Humabs Biomed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Zhenfeng Zhang
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- School of Public Health and Emergency Management, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Dirk Grimm
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Department of Infectious Diseases, Virology, Section Viral Vector Technologies, University Hospital Heidelberg, Cluster of Excellence CellNetworks, BioQuant, Center for Integrative Infectious Diseases Research (CIID), Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Xianfang Wu
- Infection Biology Program and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Stephan Urban
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
- Molecular Virology, Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany.
| | - Viet Loan Dao Thi
- Schaller Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany.
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
4
|
Pewkliang Y, Thongsri P, Suthivanich P, Thongbaiphet N, Keatkla J, Pasomsub E, Anurathapan U, Borwornpinyo S, Wongkajornsilp A, Hongeng S, Sa-Ngiamsuntorn K. Immortalized hepatocyte-like cells: A competent hepatocyte model for studying clinical HCV isolate infection. PLoS One 2024; 19:e0303265. [PMID: 38739590 PMCID: PMC11090328 DOI: 10.1371/journal.pone.0303265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
More than 58 million individuals worldwide are inflicted with chronic HCV. The disease carries a high risk of end stage liver disease, i.e., cirrhosis and hepatocellular carcinoma. Although direct-acting antiviral agents (DAAs) have revolutionized therapy, the emergence of drug-resistant strains has become a growing concern. Conventional cellular models, Huh7 and its derivatives were very permissive to only HCVcc (JFH-1), but not HCV clinical isolates. The lack of suitable host cells had hindered comprehensive research on patient-derived HCV. Here, we established a novel hepatocyte model for HCV culture to host clinically pan-genotype HCV strains. The immortalized hepatocyte-like cell line (imHC) derived from human mesenchymal stem cell carries HCV receptors and essential host factors. The imHC outperformed Huh7 as a host for HCV (JFH-1) and sustained the entire HCV life cycle of pan-genotypic clinical isolates. We analyzed the alteration of host markers (i.e., hepatic markers, cellular innate immune response, and cell apoptosis) in response to HCV infection. The imHC model uncovered the underlying mechanisms governing the action of IFN-α and the activation of sofosbuvir. The insights from HCV-cell culture model hold promise for understanding disease pathogenesis and novel anti-HCV development.
Collapse
Affiliation(s)
- Yongyut Pewkliang
- Faculty of Medicine Ramathibodi Hospital, Program in Translational Medicine, Mahidol University, Rama VI Road, Rajathevi, Bangkok, Thailand
| | - Piyanoot Thongsri
- Faculty of Medicine Ramathibodi Hospital, Program in Translational Medicine, Mahidol University, Rama VI Road, Rajathevi, Bangkok, Thailand
| | - Phichaya Suthivanich
- Faculty of Science, Excellent Center for Drug Discovery, Mahidol University, Rama VI Road, Rajathevi, Bangkok, Thailand
| | - Nipa Thongbaiphet
- Faculty of Medicine Ramathibodi Hospital, Department of Pathology, Virology Laboratory, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Jiraporn Keatkla
- Faculty of Medicine Ramathibodi Hospital, Department of Pathology, Virology Laboratory, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Ekawat Pasomsub
- Faculty of Medicine Ramathibodi Hospital, Department of Pathology, Virology Laboratory, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Usanarat Anurathapan
- Faculty of Medicine Ramathibodi Hospital, Department of Pediatrics, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Suparerk Borwornpinyo
- Faculty of Science, Excellent Center for Drug Discovery, Mahidol University, Rama VI Road, Rajathevi, Bangkok, Thailand
- Faculty of Science, Department of Biotechnology, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Adisak Wongkajornsilp
- Faculty of Medicine Siriraj Hospital, Department of Pharmacology, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Faculty of Medicine Ramathibodi Hospital, Department of Pediatrics, Mahidol University, Rajathevi, Bangkok, Thailand
| | - Khanit Sa-Ngiamsuntorn
- Faculty of Pharmacy, Department of Biochemistry, Mahidol University, Rajathevi, Bangkok, Thailand
| |
Collapse
|
5
|
Mueller F, Witteveldt J, Macias S. Antiviral Defence Mechanisms during Early Mammalian Development. Viruses 2024; 16:173. [PMID: 38399949 PMCID: PMC10891733 DOI: 10.3390/v16020173] [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] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/11/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024] Open
Abstract
The type-I interferon (IFN) response constitutes the major innate immune pathway against viruses in mammals. Despite its critical importance for antiviral defence, this pathway is inactive during early embryonic development. There seems to be an incompatibility between the IFN response and pluripotency, the ability of embryonic cells to develop into any cell type of an adult organism. Instead, pluripotent cells employ alternative ways to defend against viruses that are typically associated with safeguard mechanisms against transposable elements. The absence of an inducible IFN response in pluripotent cells and the constitutive activation of the alternative antiviral pathways have led to the hypothesis that embryonic cells are highly resistant to viruses. However, some findings challenge this interpretation. We have performed a meta-analysis that suggests that the susceptibility of pluripotent cells to viruses is directly correlated with the presence of receptors or co-receptors for viral adhesion and entry. These results challenge the current view of pluripotent cells as intrinsically resistant to infections and raise the fundamental question of why these cells have sacrificed the major antiviral defence pathway if this renders them susceptible to viruses.
Collapse
Affiliation(s)
- Felix Mueller
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, King’s Buildings, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK; (F.M.); (J.W.)
- Centre for Virus Research, MRC-University of Glasgow, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Jeroen Witteveldt
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, King’s Buildings, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK; (F.M.); (J.W.)
| | - Sara Macias
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, King’s Buildings, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK; (F.M.); (J.W.)
| |
Collapse
|
6
|
Meyers NL, Ashuach T, Lyons DE, Khalid MM, Simoneau CR, Erickson AL, Bouhaddou M, Nguyen TT, Kumar GR, Taha TY, Natarajan V, Baron JL, Neff N, Zanini F, Mahmoudi T, Quake SR, Krogan NJ, Cooper S, McDevitt TC, Yosef N, Ott M. Hepatitis C virus infects and perturbs liver stem cells. mBio 2023; 14:e0131823. [PMID: 37938000 PMCID: PMC10746249 DOI: 10.1128/mbio.01318-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 11/09/2023] Open
Abstract
IMPORTANCE The hepatitis C virus (HCV) causes liver disease, affecting millions. Even though we have effective antivirals that cure HCV, they cannot stop terminal liver disease. We used an adult stem cell-derived liver organoid system to understand how HCV infection leads to the progression of terminal liver disease. Here, we show that HCV maintains low-grade infections in liver organoids for the first time. HCV infection in liver organoids leads to transcriptional reprogramming causing cancer cell development and altered immune response. Our finding shows how HCV infection in liver organoids mimics HCV infection and patient pathogenesis. These results reveal that HCV infection in liver organoids contributes to liver disease progression.
Collapse
Affiliation(s)
| | - Tal Ashuach
- Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California Berkeley, Berkeley, California, USA
| | | | - Mir M. Khalid
- Gladstone Institute of Virology, San Francisco, California, USA
| | | | - Ann L. Erickson
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Mehdi Bouhaddou
- Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California, San Francisco, California, USA
| | - Thong T. Nguyen
- Gladstone Institute of Virology, San Francisco, California, USA
| | - G. Renuka Kumar
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Taha Y. Taha
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Vaishaali Natarajan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Jody L. Baron
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Norma Neff
- Chan Zuckerburg Biohub, San Francisco, California, USA
| | - Fabio Zanini
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stephen R. Quake
- Chan Zuckerburg Biohub, San Francisco, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Nevan J. Krogan
- Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California, San Francisco, California, USA
| | - Stewart Cooper
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Todd C. McDevitt
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
- Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California Berkeley, Berkeley, California, USA
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Chan Zuckerburg Biohub, San Francisco, California, USA
| |
Collapse
|
7
|
Georgieva M, Xenodochidis C, Krasteva N. Old age as a risk factor for liver diseases: Modern therapeutic approaches. Exp Gerontol 2023; 184:112334. [PMID: 37977514 DOI: 10.1016/j.exger.2023.112334] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Recent scientific interest has been directed towards age-related diseases, driven by the significant increase in global life expectancy and the growing population of individuals aged 65 and above. The ageing process encompasses various biological, physiological, environmental, psychological, behavioural, and social changes, leading to an augmented susceptibility to chronic illnesses. Cardiovascular, neurological, musculoskeletal, liver and oncological diseases are prevalent in the elderly. Moreover, ageing individuals demonstrate reduced regenerative capacity and decreased tolerance towards therapeutic interventions, including organ transplantation. Liver diseases, such as non-alcoholic fatty liver disease, alcoholic liver disease, hepatitis, fibrosis, and cirrhosis, have emerged as significant public health concerns. Paradoxically, these conditions remain underestimated despite their substantial global impact. Age-related factors are closely associated with the severity and unfavorable prognosis of various liver diseases, warranting further investigation to enhance clinical management and develop novel therapeutic strategies. This comprehensive review focuses specifically on age-related liver diseases, their treatment strategies, and contemporary practices. It provides a detailed account of the global burden, types, molecular mechanisms, and epigenetic alterations underlying these liver pathologies.
Collapse
Affiliation(s)
- Milena Georgieva
- Institute of Molecular Biology "Acad. Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| | - Charilaos Xenodochidis
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| |
Collapse
|
8
|
Lange F, Garn J, Anagho HA, Vondran FWR, von Hahn T, Pietschmann T, Carpentier A. Hepatitis D virus infection, innate immune response and antiviral treatments in stem cell-derived hepatocytes. Liver Int 2023; 43:2116-2129. [PMID: 37366005 DOI: 10.1111/liv.15655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Human pluripotent stem cell (hPSC)-derived hepatocyte-like cells (HLCs) are a valuable model to investigate host-pathogen interactions of hepatitis viruses in a mature and authentic environment. Here, we investigate the susceptibility of HLCs to the hepatitis delta virus (HDV). METHODS We differentiated hPSC into HLCs, and inoculated them with infectious HDV produced in Huh7NTCP . HDV infection and cellular response was monitored by RTqPCR and immunostaining. RESULTS Cells undergoing hepatic differentiation become susceptible to HDV after acquiring expression of the viral receptor Na+ -taurocholate co-transporting polypeptide (NTCP) during hepatic specification. Inoculation of HLCs with HDV leads to detection of intracellular HDV RNA and accumulation of the HDV antigen in the cells. Upon infection, the HLCs mounted an innate immune response based on induction of the interferons IFNB and L, and upregulation of interferon-stimulated genes. The intensity of this immune response positively correlated with the level of viral replication and was dependant on both the JAK/STAT and NFκB pathway activation. Importantly, this innate immune response did not inhibit HDV replication. However, pre-treatment of the HLCs with IFNα2b reduced viral infection, suggesting that ISGs may limit early stages of infection. Myrcludex efficiently abrogated infection and blocked innate immune activation. Lonafarnib treatment of HDV mono infected HLCs on the other hand led to exacerbated viral replication and innate immune response. CONCLUSION The HDV in vitro mono-infection model represents a new tool to study HDV replication, its host-pathogen interactions and evaluate new antiviral drugs in cells displaying mature hepatic functions.
Collapse
Affiliation(s)
- Frauke Lange
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between Medical School Hannover (MHH) and Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Jonathan Garn
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between Medical School Hannover (MHH) and Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Holda A Anagho
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between Medical School Hannover (MHH) and Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
- Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Florian W R Vondran
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Thomas von Hahn
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
- Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between Medical School Hannover (MHH) and Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Arnaud Carpentier
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between Medical School Hannover (MHH) and Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
Yuan Y, Cotton K, Samarasekera D, Khetani SR. Engineered Platforms for Maturing Pluripotent Stem Cell-Derived Liver Cells for Disease Modeling. Cell Mol Gastroenterol Hepatol 2023; 15:1147-1160. [PMID: 36738860 PMCID: PMC10034210 DOI: 10.1016/j.jcmgh.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/06/2023]
Abstract
Several liver diseases (eg, hepatitis B/C viruses, alcoholic/nonalcoholic fatty liver, malaria, monogenic diseases, and drug-induced liver injury) significantly impact global mortality and morbidity. Species-specific differences in liver functions limit the use of animals to fully elucidate/predict human outcomes; therefore, in vitro human liver models are used for basic and translational research to complement animal studies. However, primary human liver cells are in short supply and display donor-to-donor variability in viability/quality. In contrast, human hepatocyte-like cells (HLCs) differentiated from induced pluripotent stem cells and embryonic stem cells are a near infinite cell resource that retains the patient/donor's genetic background; however, conventional protocols yield immature phenotypes. HLC maturation can be significantly improved using advanced techniques, such as protein micropatterning to precisely control cell-cell interactions, controlled sized spheroids, organoids with multiple cell types and layers, 3-dimensional bioprinting to spatially control cell populations, microfluidic devices for automated nutrient exchange and to induce liver zonation via soluble factor gradients, and synthetic biology to genetically modify the HLCs to accelerate and enhance maturation. Here, we present design features and characterization for representative advanced HLC maturation platforms and then discuss HLC use for modeling various liver diseases. Lastly, we discuss desirable advances to move this field forward. We anticipate that with continued advances in this space, pluripotent stem cell-derived liver models will provide human-relevant data much earlier in preclinical drug development and reduce animal usage, help elucidate liver disease mechanisms for the discovery of efficacious and safe therapeutics, and be useful as cell-based therapies for patients suffering from end-stage liver failure.
Collapse
Affiliation(s)
- Yang Yuan
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois
| | - Kristen Cotton
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois
| | - Dinithi Samarasekera
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
10
|
A road to contemporary era of hepatitis B virus regimen replacing existing therapeutics exploiting plant secondary metabolites as emerging heroes in exploring drugs: An expedition for a functional cure. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
11
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
12
|
Cheng Y, Medina A, Yao Z, Basu M, Natekar JP, Lang J, Sanchez E, Nkembo MB, Xu C, Qian X, Nguyen PTT, Wen Z, Song H, Ming GL, Kumar M, Brinton MA, Li MMH, Tang H. Intrinsic antiviral immunity of barrier cells revealed by an iPSC-derived blood-brain barrier cellular model. Cell Rep 2022; 39:110885. [PMID: 35649379 PMCID: PMC9230077 DOI: 10.1016/j.celrep.2022.110885] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/27/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Physiological blood-tissue barriers play a critical role in separating the circulation from immune-privileged sites and denying access to blood-borne viruses. The mechanism of virus restriction by these barriers is poorly understood. We utilize induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (iBMECs) to study virus-blood-brain barrier (BBB) interactions. These iPSC-derived cells faithfully recapitulate a striking difference in in vivo neuroinvasion by two alphavirus isolates and are selectively permissive to neurotropic flaviviruses. A model of cocultured iBMECs and astrocytes exhibits high transendothelial electrical resistance and blocks non-neurotropic flaviviruses from getting across the barrier. We find that iBMECs constitutively express an interferon-induced gene, IFITM1, which preferentially restricts the replication of non-neurotropic flaviviruses. Barrier cells from blood-testis and blood-retinal barriers also constitutively express IFITMs that contribute to the viral resistance. Our application of a renewable human iPSC-based model for studying virus-BBB interactions reveals that intrinsic immunity at the barriers contributes to virus exclusion.
Collapse
Affiliation(s)
- Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Angelica Medina
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Zhenlan Yao
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mausumi Basu
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Jianshe Lang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Egan Sanchez
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Mezindia B Nkembo
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Chongchong Xu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Xuyu Qian
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phuong T T Nguyen
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mukesh Kumar
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Margo A Brinton
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Melody M H Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
13
|
Natarajan V, Simoneau CR, Erickson AL, Meyers NL, Baron JL, Cooper S, McDevitt TC, Ott M. Modelling T-cell immunity against hepatitis C virus with liver organoids in a microfluidic coculture system. Open Biol 2022; 12:210320. [PMID: 35232252 PMCID: PMC8889170 DOI: 10.1098/rsob.210320] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/25/2022] [Indexed: 01/09/2023] Open
Abstract
Hepatitis C virus (HCV) remains a global public health challenge with an estimated 71 million people chronically infected, with surges in new cases and no effective vaccine. New methods are needed to study the human immune response to HCV since in vivo animal models are limited and in vitro cancer cell models often show dysregulated immune and proliferative responses. Here, we developed a CD8+ T cell and adult stem cell liver organoid system using a microfluidic chip to coculture 3D human liver organoids embedded in extracellular matrix with HLA-matched primary human T cells in suspension. We then employed automated phase contrast and immunofluorescence imaging to monitor T cell invasion and morphological changes in the liver organoids. This microfluidic coculture system supports targeted killing of liver organoids when pulsed with a peptide specific for HCV non-structural protein 3 (NS3) (KLVALGINAV) in the presence of patient-derived CD8+ T cells specific for KLVALGINAV. This demonstrates the novel potential of the coculture system to molecularly study adaptive immune responses to HCV in an in vitro setting using primary human cells.
Collapse
Affiliation(s)
| | - Camille R. Simoneau
- The Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Ann L. Erickson
- Division of General and Transplant Hepatology, California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | | | - Jody L. Baron
- UCSF Liver Center, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Stewart Cooper
- Division of General and Transplant Hepatology, California Pacific Medical Center and Research Institute, San Francisco, CA, USA
- UCSF Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Todd C. McDevitt
- The Gladstone Institutes, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Melanie Ott
- The Gladstone Institutes, San Francisco, CA, USA
- UCSF Liver Center, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Graffmann N, Scherer B, Adjaye J. In vitro differentiation of pluripotent stem cells into hepatocyte like cells - basic principles and current progress. Stem Cell Res 2022; 61:102763. [DOI: 10.1016/j.scr.2022.102763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
|
15
|
Nicolay W, Moeller R, Kahl S, Vondran FWR, Pietschmann T, Kunz S, Gerold G. Characterization of RNA Sensing Pathways in Hepatoma Cell Lines and Primary Human Hepatocytes. Cells 2021; 10:3019. [PMID: 34831243 PMCID: PMC8616302 DOI: 10.3390/cells10113019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 11/23/2022] Open
Abstract
The liver is targeted by several human pathogenic RNA viruses for viral replication and dissemination; despite this, the extent of innate immune sensing of RNA viruses by human hepatocytes is insufficiently understood to date. In particular, for highly human tropic viruses such as hepatitis C virus, cell culture models are needed to study immune sensing. However, several human hepatoma cell lines have impaired RNA sensing pathways and fail to mimic innate immune responses in the human liver. Here we compare the RNA sensing properties of six human hepatoma cell lines, namely Huh-6, Huh-7, HepG2, HepG2-HFL, Hep3B, and HepaRG, with primary human hepatocytes. We show that primary liver cells sense RNA through retinoic acid-inducible gene I (RIG-I) like receptor (RLR) and Toll-like receptor 3 (TLR3) pathways. Of the tested cell lines, Hep3B cells most closely mimicked the RLR and TLR3 mediated sensing in primary hepatocytes. This was shown by the expression of RLRs and TLR3 as well as the expression and release of bioactive interferon in primary hepatocytes and Hep3B cells. Our work shows that Hep3B cells partially mimic RNA sensing in primary hepatocytes and thus can serve as in vitro model to study innate immunity to RNA viruses in hepatocytes.
Collapse
Affiliation(s)
- Wiebke Nicolay
- TWINCORE—Centre for Experimental and Clinical Infection Research, Institute for Experimental Virology, 30625 Hannover, Germany; (W.N.); (R.M.); (S.K.); (T.P.)
| | - Rebecca Moeller
- TWINCORE—Centre for Experimental and Clinical Infection Research, Institute for Experimental Virology, 30625 Hannover, Germany; (W.N.); (R.M.); (S.K.); (T.P.)
- Center for Emerging Infections and Zoonoses (RIZ), Institute of Biochemistry & Research, University of Veterinary Medicine Hannover, 30625 Hannover, Germany
| | - Sina Kahl
- TWINCORE—Centre for Experimental and Clinical Infection Research, Institute for Experimental Virology, 30625 Hannover, Germany; (W.N.); (R.M.); (S.K.); (T.P.)
| | - Florian W. R. Vondran
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, 30625 Hannover, Germany;
- German Centre for Infection Research (DZIF), 30100 Braunschweig, Germany
| | - Thomas Pietschmann
- TWINCORE—Centre for Experimental and Clinical Infection Research, Institute for Experimental Virology, 30625 Hannover, Germany; (W.N.); (R.M.); (S.K.); (T.P.)
| | - Stefan Kunz
- Institute of Microbiology, Lausanne University Hospital, CH-1011 Lausanne, Switzerland;
| | - Gisa Gerold
- TWINCORE—Centre for Experimental and Clinical Infection Research, Institute for Experimental Virology, 30625 Hannover, Germany; (W.N.); (R.M.); (S.K.); (T.P.)
- Center for Emerging Infections and Zoonoses (RIZ), Institute of Biochemistry & Research, University of Veterinary Medicine Hannover, 30625 Hannover, Germany
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden
| |
Collapse
|
16
|
Chang M, Bogacheva MS, Lou YR. Challenges for the Applications of Human Pluripotent Stem Cell-Derived Liver Organoids. Front Cell Dev Biol 2021; 9:748576. [PMID: 34660606 PMCID: PMC8517247 DOI: 10.3389/fcell.2021.748576] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
The current organoid culture systems allow pluripotent and adult stem cells to self-organize to form three-dimensional (3D) structures that provide a faithful recapitulation of the architecture and function of in vivo organs. In particular, human pluripotent stem cell-derived liver organoids (PSC-LOs) can be used in regenerative medicine and preclinical applications, such as disease modeling and drug discovery. New bioengineering tools, such as microfluidics, biomaterial scaffolds, and 3D bioprinting, are combined with organoid technologies to increase the efficiency of hepatic differentiation and enhance the functional maturity of human PSC-LOs by precise control of cellular microenvironment. Long-term stabilization of hepatocellular functions of in vitro liver organoids requires the combination of hepatic endodermal, endothelial, and mesenchymal cells. To improve the biological function and scalability of human PSC-LOs, bioengineering methods have been used to identify diverse and zonal hepatocyte populations in liver organoids for capturing heterogeneous pathologies. Therefore, constructing engineered liver organoids generated from human PSCs will be an extremely versatile tool in in vitro disease models and regenerative medicine in future. In this review, we aim to discuss the recent advances in bioengineering technologies in liver organoid culture systems that provide a timely and necessary study to model disease pathology and support drug discovery in vitro and to generate cell therapy products for transplantation.
Collapse
Affiliation(s)
- Mingyang Chang
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Mariia S. Bogacheva
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Yan-Ru Lou
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Kukla DA, Khetani SR. Bioengineered Liver Models for Investigating Disease Pathogenesis and Regenerative Medicine. Semin Liver Dis 2021; 41:368-392. [PMID: 34139785 DOI: 10.1055/s-0041-1731016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Owing to species-specific differences in liver pathways, in vitro human liver models are utilized for elucidating mechanisms underlying disease pathogenesis, drug development, and regenerative medicine. To mitigate limitations with de-differentiated cultures, bioengineers have developed advanced techniques/platforms, including micropatterned cocultures, spheroids/organoids, bioprinting, and microfluidic devices, for perfusing cell cultures and liver slices. Such techniques improve mature functions and culture lifetime of primary and stem-cell human liver cells. Furthermore, bioengineered liver models display several features of liver diseases including infections with pathogens (e.g., malaria, hepatitis C/B viruses, Zika, dengue, yellow fever), alcoholic/nonalcoholic fatty liver disease, and cancer. Here, we discuss features of bioengineered human liver models, their uses for modeling aforementioned diseases, and how such models are being augmented/adapted for fabricating implantable human liver tissues for clinical therapy. Ultimately, continued advances in bioengineered human liver models have the potential to aid the development of novel, safe, and efficacious therapies for liver disease.
Collapse
Affiliation(s)
- David A Kukla
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
18
|
Pasqua M, Di Gesù R, Chinnici CM, Conaldi PG, Francipane MG. Generation of Hepatobiliary Cell Lineages from Human Induced Pluripotent Stem Cells: Applications in Disease Modeling and Drug Screening. Int J Mol Sci 2021; 22:8227. [PMID: 34360991 PMCID: PMC8348238 DOI: 10.3390/ijms22158227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
The possibility to reproduce key tissue functions in vitro from induced pluripotent stem cells (iPSCs) is offering an incredible opportunity to gain better insight into biological mechanisms underlying development and disease, and a tool for the rapid screening of drug candidates. This review attempts to summarize recent strategies for specification of iPSCs towards hepatobiliary lineages -hepatocytes and cholangiocytes-and their use as platforms for disease modeling and drug testing. The application of different tissue-engineering methods to promote accurate and reliable readouts is discussed. Space is given to open questions, including to what extent these novel systems can be informative. Potential pathways for improvement are finally suggested.
Collapse
Affiliation(s)
- Mattia Pasqua
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Roberto Di Gesù
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Cinzia Maria Chinnici
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- Dipartimento della Ricerca, IRCCS ISMETT, 90127 Palermo, Italy;
| | | | - Maria Giovanna Francipane
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
19
|
Azar J, Bahmad HF, Daher D, Moubarak MM, Hadadeh O, Monzer A, Al Bitar S, Jamal M, Al-Sayegh M, Abou-Kheir W. The Use of Stem Cell-Derived Organoids in Disease Modeling: An Update. Int J Mol Sci 2021; 22:7667. [PMID: 34299287 PMCID: PMC8303386 DOI: 10.3390/ijms22147667] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Organoids represent one of the most important advancements in the field of stem cells during the past decade. They are three-dimensional in vitro culturing models that originate from self-organizing stem cells and can mimic the in vivo structural and functional specificities of body organs. Organoids have been established from multiple adult tissues as well as pluripotent stem cells and have recently become a powerful tool for studying development and diseases in vitro, drug screening, and host-microbe interaction. The use of stem cells-that have self-renewal capacity to proliferate and differentiate into specialized cell types-for organoids culturing represents a major advancement in biomedical research. Indeed, this new technology has a great potential to be used in a multitude of fields, including cancer research, hereditary and infectious diseases. Nevertheless, organoid culturing is still rife with many challenges, not limited to being costly and time consuming, having variable rates of efficiency in generation and maintenance, genetic stability, and clinical applications. In this review, we aim to provide a synopsis of pluripotent stem cell-derived organoids and their use for disease modeling and other clinical applications.
Collapse
Affiliation(s)
- Joseph Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Darine Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Maya M. Moubarak
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Samar Al Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Mohamed Jamal
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 66566, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| |
Collapse
|
20
|
Khoshdel-Rad N, Zahmatkesh E, Bikmulina P, Peshkova M, Kosheleva N, Bezrukov EA, Sukhanov RB, Solovieva A, Shpichka A, Timashev P, Vosough M. Modeling Hepatotropic Viral Infections: Cells vs. Animals. Cells 2021; 10:1726. [PMID: 34359899 PMCID: PMC8305759 DOI: 10.3390/cells10071726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
The lack of an appropriate platform for a better understanding of the molecular basis of hepatitis viruses and the absence of reliable models to identify novel therapeutic agents for a targeted treatment are the two major obstacles for launching efficient clinical protocols in different types of viral hepatitis. Viruses are obligate intracellular parasites, and the development of model systems for efficient viral replication is necessary for basic and applied studies. Viral hepatitis is a major health issue and a leading cause of morbidity and mortality. Despite the extensive efforts that have been made on fundamental and translational research, traditional models are not effective in representing this viral infection in a laboratory. In this review, we discuss in vitro cell-based models and in vivo animal models, with their strengths and weaknesses. In addition, the most important findings that have been retrieved from each model are described.
Collapse
Affiliation(s)
- Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Nastasia Kosheleva
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- FSBSI ‘Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Evgeny A. Bezrukov
- Department of Urology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.A.B.); (R.B.S.)
| | - Roman B. Sukhanov
- Department of Urology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.A.B.); (R.B.S.)
| | - Anna Solovieva
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| |
Collapse
|
21
|
Afarid M, Sanie-Jahromi F. Mesenchymal Stem Cells and COVID-19: Cure, Prevention, and Vaccination. Stem Cells Int 2021; 2021:6666370. [PMID: 34035820 PMCID: PMC8103964 DOI: 10.1155/2021/6666370] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/26/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
COVID-19 disease has been a global health problem since late 2019. There are many concerns about the rapid spread of this disease, and yet, there is no approved treatment for COVID-19. Several biological interventions have been under study recently to investigate efficient treatment for this viral disease. Besides, many efforts have been made to find a safe way to prevent and vaccinate people against COVID-19 disease. In severe cases, patients suffer from acute respiratory distress syndrome usually associated with an increased level of inflammatory cytokines, called a cytokine storm. It seems that reequilibrating the hyperinflammatory response of the host immune system and regeneration of damaged cells could be the main way to manage the disease. Mesenchymal stem cells (MSCs) have been recently under investigation in this regard, and the achieved clinical outcomes show promising evidence for stem cell-based therapy of COVID-19. MSCs are known for their potential for immunomodulation, defense against virus infection, and tissue regeneration. MSCs are a newly emerged platform for designing vaccines and show promising evidence in this area. In the present study, we provided a thorough research study on the most recent clinical studies based on stem cells in the treatment of COVID-19 while introducing stem cell exclusivities for use as an immune disorder or lung cell therapy and its potential application for protection and vaccination against COVID-19.
Collapse
Affiliation(s)
- Mehrdad Afarid
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
Arez F, Rodrigues AF, Brito C, Alves PM. Bioengineered Liver Cell Models of Hepatotropic Infections. Viruses 2021; 13:773. [PMID: 33925701 PMCID: PMC8146083 DOI: 10.3390/v13050773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatitis viruses and liver-stage malaria are within the liver infections causing higher morbidity and mortality rates worldwide. The highly restricted tropism of the major human hepatotropic pathogens-namely, the human hepatitis B and C viruses and the Plasmodium falciparum and Plasmodium vivax parasites-has hampered the development of disease models. These models are crucial for uncovering the molecular mechanisms underlying the biology of infection and governing host-pathogen interaction, as well as for fostering drug development. Bioengineered cell models better recapitulate the human liver microenvironment and extend hepatocyte viability and phenotype in vitro, when compared with conventional two-dimensional cell models. In this article, we review the bioengineering tools employed in the development of hepatic cell models for studying infection, with an emphasis on 3D cell culture strategies, and discuss how those tools contributed to the level of recapitulation attained in the different model layouts. Examples of host-pathogen interactions uncovered by engineered liver models and their usefulness in drug development are also presented. Finally, we address the current bottlenecks, trends, and prospect toward cell models' reliability, robustness, and reproducibility.
Collapse
MESH Headings
- Animals
- Bioengineering/methods
- Cell Culture Techniques
- Disease Models, Animal
- Disease Susceptibility
- Drug Discovery
- Hepatitis/drug therapy
- Hepatitis/etiology
- Hepatitis/metabolism
- Hepatitis/pathology
- Hepatitis, Viral, Human/etiology
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatocytes/metabolism
- Hepatocytes/parasitology
- Hepatocytes/virology
- Host-Pathogen Interactions
- Humans
- Liver/metabolism
- Liver/parasitology
- Liver/virology
- Liver Diseases, Parasitic/etiology
- Liver Diseases, Parasitic/metabolism
- Liver Diseases, Parasitic/pathology
Collapse
Affiliation(s)
- Francisca Arez
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F. Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
23
|
Bove G, Mehnert AK, Dao Thi VL. iPSCs for modeling hepatotropic pathogen infections. IPSCS FOR STUDYING INFECTIOUS DISEASES 2021:149-213. [DOI: 10.1016/b978-0-12-823808-0.00013-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
24
|
Carpentier A, Sheldon J, Vondran FWR, Brown RJ, Pietschmann T. Efficient acute and chronic infection of stem cell-derived hepatocytes by hepatitis C virus. Gut 2020; 69:1659-1666. [PMID: 32114504 PMCID: PMC7456736 DOI: 10.1136/gutjnl-2019-319354] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE AND DESIGN Human stem cell-derived hepatocyte-like cells (HLCs) have shown high potential as authentic model for dissection of the HCV life cycle and virus-induced pathogenesis. However, modest HCV replication, possibly due to robust innate immune responses, limits their broader use. To overcome these limitations and to dissect the mechanisms responsible for control of HCV, we analysed expression of key components of the interferon (IFN) system in HLCs, assessed permissiveness for different HCV strains and blocked innate immune signalling by pharmacological intervention. RESULTS Transcriptional profiling revealed that HLCs constitutively express messenger RNA of RLRs, and members of the IFN pathway. Moreover, HLCs upregulated IFNs and canonical interferon-regulated genes (IRGs) upon transfection with the double-stranded RNA mimic poly(I:C). Infection of HLCs with Jc1-HCVcc produced only limited viral progeny. In contrast, infection with p100, a Jc1-derived virus population with enhanced replication fitness and partial resistance to IFN, resulted in robust yet transient viraemia. Viral titres declined concomitant with a peak of IRG induction. Addition of ruxolitinib, a JAK/STAT inhibitor, permitted chronic infection and raised p100 infectious virus titres to 1×105 FFU/mL. IRGs expression profiling in infected HLCs revealed a landscape of HCV-dependent transcriptional changes similar to HCV-infected primary human hepatocytes, but distinct from Huh-7.5 cells. Withdrawal of ruxolitinib restored innate immune responses and resulted in HCV clearance. CONCLUSION This authentic human cell model is well suited to examine acute and chronic host-HCV interactions, particularly IFN-triggered antiviral effector functions and mechanisms of innate immune control of HCV infection.
Collapse
Affiliation(s)
- Arnaud Carpentier
- Institute of Experimental Virology, Twincore, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Julie Sheldon
- Institute of Experimental Virology, Twincore, Hannover, Germany
| | - Florian W R Vondran
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Richard Jp Brown
- Institute of Experimental Virology, Twincore, Hannover, Germany
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Thomas Pietschmann
- Institute of Experimental Virology, Twincore, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| |
Collapse
|
25
|
Utility of Common Marmoset ( Callithrix jacchus) Embryonic Stem Cells in Liver Disease Modeling, Tissue Engineering and Drug Metabolism. Genes (Basel) 2020; 11:genes11070729. [PMID: 32630053 PMCID: PMC7397002 DOI: 10.3390/genes11070729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
The incidence of liver disease is increasing significantly worldwide and, as a result, there is a pressing need to develop new technologies and applications for end-stage liver diseases. For many of them, orthotopic liver transplantation is the only viable therapeutic option. Stem cells that are capable of differentiating into all liver cell types and could closely mimic human liver disease are extremely valuable for disease modeling, tissue regeneration and repair, and for drug metabolism studies to develop novel therapeutic treatments. Despite the extensive research efforts, positive results from rodent models have not translated meaningfully into realistic preclinical models and therapies. The common marmoset Callithrix jacchus has emerged as a viable non-human primate model to study various human diseases because of its distinct features and close physiologic, genetic and metabolic similarities to humans. C. jacchus embryonic stem cells (cjESC) and recently generated cjESC-derived hepatocyte-like cells (cjESC-HLCs) could fill the gaps in disease modeling, liver regeneration and metabolic studies. They are extremely useful for cell therapy to regenerate and repair damaged liver tissues in vivo as they could efficiently engraft into the liver parenchyma. For in vitro studies, they would be advantageous for drug design and metabolism in developing novel drugs and cell-based therapies. Specifically, they express both phase I and II metabolic enzymes that share similar substrate specificities, inhibition and induction characteristics, and drug metabolism as their human counterparts. In addition, cjESCs and cjESC-HLCs are advantageous for investigations on emerging research areas, including blastocyst complementation to generate entire livers, and bioengineering of discarded livers to regenerate whole livers for transplantation.
Collapse
|
26
|
Fischer L, Lucendo-Villarin B, Hay DC, O’Farrelly C. Human PSC-Derived Hepatocytes Express Low Levels of Viral Pathogen Recognition Receptors, but Are Capable of Mounting an Effective Innate Immune Response. Int J Mol Sci 2020; 21:ijms21113831. [PMID: 32481600 PMCID: PMC7312201 DOI: 10.3390/ijms21113831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 01/19/2023] Open
Abstract
Hepatocytes are key players in the innate immune response to liver pathogens but are challenging to study because of inaccessibility and a short half-life. Recent advances in in vitro differentiation of hepatocyte-like cells (HLCs) facilitated studies of hepatocyte-pathogen interactions. Here, we aimed to define the anti-viral innate immune potential of human HLCs with a focus on toll-like receptor (TLR)-expression and the presence of a metabolic switch. We analysed cytoplasmic pattern recognition receptor (PRR)- and endosomal TLR-expression and activity and adaptation of HLCs to an inflammatory environment. We found that transcript levels of retinoic acid inducible gene I (RIG-I), melanoma differentiation antigen 5 (MDA5), and TLR3 became downregulated during differentiation, indicating the acquisition of a more tolerogenic phenotype, as expected in healthy hepatocytes. HLCs responded to activation of RIG-I by producing interferons (IFNs) and IFN-stimulated genes. Despite low-level expression of TLR3, receptor expression was upregulated in an inflammatory environment. TLR3 signalling induced expression of proinflammatory cytokines at the gene level, indicating that several PRRs need to interact for successful innate immune activation. The inflammatory responsiveness of HLCs was accompanied by the downregulation of cytochrome P450 3A and 1A2 activity and decreased serum protein production, showing that the metabolic switch seen in primary hepatocytes during anti-viral responses is also present in HLCs.
Collapse
Affiliation(s)
- Lena Fischer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
| | | | - David C. Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK;
- Correspondence: (D.C.H.); (C.O.)
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
- School of Medicine, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
- Correspondence: (D.C.H.); (C.O.)
| |
Collapse
|
27
|
Kakinuma S, Watanabe M. Analysis of the mechanism underlying liver diseases using human induced pluripotent stem cells. Immunol Med 2019; 42:71-78. [PMID: 31498713 DOI: 10.1080/25785826.2019.1657254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Results of recent studies have shown that disease models using human induced pluripotent stem (iPS) cells have recapitulated the pathophysiology of genetic liver diseases, viral hepatitis and hepatic fibrosis. The utilization of human iPS cells as a model of liver diseases has several substantial advantages compared with primary hepatocytes and cancer cell lines, such as the potential for unlimited expansion and similarity of biological characteristics to normal liver cells. In this review, we have focused on modeling liver diseases using human iPS cells and discussed the experimental evidence that supports the utility of such disease models, including that in our recent studies. Genetically modified or patient-derived human iPS cells can mimic congenital liver disease phenotypes. Human iPS-derived hepatic cells can be infected with the hepatitis viruses. The co-culture of human iPS-derived hepatocytes and mesenchyme partially mimics the process of liver fibrosis. Human iPS cell-derived hepatic cells and the co-culture system of such cells will contribute to the progress of studies on the pathophysiology of genetic and non-genetic liver diseases and development of novel therapeutic strategies for treating liver diseases.
Collapse
Affiliation(s)
- Sei Kakinuma
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University (TMDU) , Tokyo , Japan.,Department of Liver Disease Control, Tokyo Medical and Dental University (TMDU) , Tokyo , Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University (TMDU) , Tokyo , Japan
| |
Collapse
|
28
|
Butt SL, Moura VMBD, Susta L, Miller PJ, Hutcheson JM, Cardenas-Garcia S, Brown CC, West FD, Afonso CL, Stanton JB. Tropism of Newcastle disease virus strains for chicken neurons, astrocytes, oligodendrocytes, and microglia. BMC Vet Res 2019; 15:317. [PMID: 31484573 PMCID: PMC6727330 DOI: 10.1186/s12917-019-2053-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/18/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Newcastle disease (ND), which is caused by infections of poultry species with virulent strains of Avian orthoavulavirus-1, also known as avian paramyxovirus 1 (APMV-1), and formerly known as Newcastle disease virus (NDV), may cause neurological signs and encephalitis. Neurological signs are often the only clinical signs observed in birds infected with neurotropic strains of NDV. Experimental infections have shown that the replication of virulent NDV (vNDV) strains is in the brain parenchyma and is possibly confined to neurons and ependymal cells. However, little information is available on the ability of vNDV strains to infect subset of glial cells (astrocytes, oligodendrocytes, and microglia). The objective of this study was to evaluate the ability of NDV strains of different levels of virulence to infect a subset of glial cells both in vitro and in vivo. Thus, neurons, astrocytes and oligodendrocytes from the brains of day-old White Leghorn chickens were harvested, cultured, and infected with both non-virulent (LaSota) and virulent, neurotropic (TxGB) NDV strains. To confirm these findings in vivo, the tropism of three vNDV strains with varying pathotypes (SA60 [viscerotropic], TxGB [neurotropic], and Tx450 [mesogenic]) was assessed in archived formalin-fixed material from day-old chicks inoculated intracerebrally. RESULTS Double immunofluorescence for NDV nucleoprotein and cellular markers showed that both strains infected at least 20% of each of the cell types (neurons, astrocytes, and oligodendrocytes). At 24 h post-inoculation, TxGB replicated significantly more than LaSota. Double immunofluorescence (DIFA) with markers for neurons, astrocytes, microglia, and NDV nucleoprotein detected the three strains in all three cell types at similar levels. CONCLUSION These data indicate that similar to other paramyxoviruses, neurons and glial cells (astrocytes, oligodendrocytes, and microglia) are susceptible to vNDV infection, and suggest that factors other than cellular tropism are likely the major determinant of the neurotropic phenotype.
Collapse
Affiliation(s)
- Salman L Butt
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.,Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA
| | - Veridiana Maria Brianezi Dignani Moura
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA.,Animal Pathology, School of Veterinary Medicine and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - Leonardo Susta
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA.,Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | - Patti J Miller
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA.,Department of Population Health, College of Veterinary Medicine, Athens, GA, USA
| | - Jessica M Hutcheson
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA.,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Stivalis Cardenas-Garcia
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA.,Department of Population Health, College of Veterinary Medicine, Athens, GA, USA
| | - Corrie C Brown
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Franklin D West
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA.,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Claudio L Afonso
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, Athens, GA, USA.
| | - James B Stanton
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
29
|
Human Pluripotent Stem Cell-Derived Endoderm for Modeling Development and Clinical Applications. Cell Stem Cell 2019; 22:485-499. [PMID: 29625066 DOI: 10.1016/j.stem.2018.03.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The liver, lung, pancreas, and digestive tract all originate from the endoderm germ layer, and these vital organs are subject to many life-threatening diseases affecting millions of patients. However, primary cells from endodermal organs are often difficult to grow in vitro. Human pluripotent stem cells thus hold great promise for generating endoderm cells and their derivatives as tools for the development of new therapeutics against a variety of global healthcare challenges. Here we describe recent advances in methods for generating endodermal cell types from human pluripotent stem cells and their use for disease modeling and cell-based therapy.
Collapse
|
30
|
Fu RM, Decker CC, Dao Thi VL. Cell Culture Models for Hepatitis E Virus. Viruses 2019; 11:E608. [PMID: 31277308 PMCID: PMC6669563 DOI: 10.3390/v11070608] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/24/2019] [Accepted: 06/29/2019] [Indexed: 12/14/2022] Open
Abstract
Despite a growing awareness, hepatitis E virus (HEV) remains understudied and investigations have been historically hampered by the absence of efficient cell culture systems. As a result, the pathogenesis of HEV infection and basic steps of the HEV life cycle are poorly understood. Major efforts have recently been made through the development of HEV infectious clones and cellular systems that significantly advanced HEV research. Here, we summarize these systems, discussing their advantages and disadvantages for HEV studies. We further capitalize on the need for HEV-permissive polarized cell models to better recapitulate the entire HEV life cycle and transmission.
Collapse
Affiliation(s)
- Rebecca Menhua Fu
- Schaller Research Group at Department of Infectious Diseases and Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Charlotte Caroline Decker
- Schaller Research Group at Department of Infectious Diseases and Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Viet Loan Dao Thi
- Schaller Research Group at Department of Infectious Diseases and Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, 69120 Heidelberg, Germany.
| |
Collapse
|
31
|
Thomas E, Baumert TF. Hepatitis B Virus-Hepatocyte Interactions and Innate Immune Responses: Experimental Models and Molecular Mechanisms. Semin Liver Dis 2019; 39:301-314. [PMID: 31266064 PMCID: PMC7377277 DOI: 10.1055/s-0039-1685518] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Chronic hepatitis B virus (HBV) infection is a major cause of liver disease and cancer worldwide. While current therapeutic approaches can efficiently control viral infection, efficient curative antivirals are absent. The understanding of virus-hepatocyte interactions and sensing of viral infection is an important prerequisite for the development of novel antiviral therapies for cure. Hepatocyte intrinsic innate immunity provides a rapid first line of defense to combat viral infection through the upregulation of antiviral and inflammatory genes. However, the functional relevance of many of these antiviral signaling pathways in the liver and their role in HBV pathogenesis is still only partially understood. The recent identification of intracellular RNA and DNA sensing pathways and their involvement in disease biology, including viral pathogenesis and carcinogenesis, is currently transforming our understanding of virus-host interactions. Here the authors review the current knowledge on intrinsic antiviral innate immune responses including the role of viral nucleic acid sensing pathways in the liver. Since HBV has been designated as a "stealth virus," the study of the impact of HBV on signaling pathways in the hepatocyte is of significant interest to understand viral pathogenesis. Characterizing the mechanism underlying these HBV-host interactions and targeting related pathways to enhance antiviral innate responses may open new strategies to trigger noncytopathic clearance of covalently closed circular DNA to ultimately cure patients with chronic HBV infection.
Collapse
Affiliation(s)
- Emmanuel Thomas
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Laboratory of Excellence HEPSYS, University of Strasbourg, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
32
|
Xiang K, Xiao Y, Li Y, He L, Wang L, Zhuang H, Li T. The Effect of the Hepatitis B Virus Surface Protein Truncated sC69 ∗ Mutation on Viral Infectivity and the Host Innate Immune Response. Front Microbiol 2019; 10:1341. [PMID: 31249567 PMCID: PMC6584109 DOI: 10.3389/fmicb.2019.01341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022] Open
Abstract
Viruses could rapidly diversify into variants, which has long been known to facilitate viral adaption in the host. Recent studies showed that cooperation among variants and wild-type (WT) also increased viral fitness. Here, a mutant of sC69∗ in small hepatitis B surface protein (SHBs) that resulted in premature stop was investigated and the frequency of sC69∗ was 4.37% (19/435), most of which coexisted with the WT (78.95%, 15/19), indicating mixed viral populations. Functional studies showed that sC69∗ mutant was associated with lower viral spread, but could be rescued by coexisting with the WT. The sC69∗ mutant showed to attenuate host innate immune response during infection and poly (I:C) treatment such as IL29, ISG15, and RIG-I (p < 0.05). The lower immune response was not caused by the lower replication of sC69∗ mutant. Our data provide information that sC69∗ coexisting with the WT might facilitate the fitness and persistence of the viral quasispecies in the host.
Collapse
Affiliation(s)
- Kuanhui Xiang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yiwei Xiao
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yao Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lingyuan He
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Luwei Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hui Zhuang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Tong Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
33
|
Wang J, Qu B, Zhang F, Zhang C, Deng W, Dao Thi VL, Xia Y. Stem Cell-Derived Hepatocyte-Like Cells as Model for Viral Hepatitis Research. Stem Cells Int 2019; 2019:9605252. [PMID: 31281392 PMCID: PMC6594266 DOI: 10.1155/2019/9605252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Viral hepatitis, the leading cause of liver diseases worldwide, is induced upon infection with hepatotropic viruses, including hepatitis A, B, C, D, and E virus. Due to their obligate intracellular lifestyles, culture systems for efficient viral replication are vital. Although basic and translational research on viral hepatitis has been performed for many years, conventional hepatocellular culture systems are not optimal. These studies have greatly benefited from recent efforts on improving cell culture models for virus replication and infection studies. Here we summarize the use of human stem cell-derived hepatocyte-like cells for hepatotropic virus infection studies, including the dissection of virus-host interactions and virus-induced pathogenesis as well as the identification and validation of novel antiviral agents.
Collapse
Affiliation(s)
- Jingjing Wang
- State Key Laboratory of Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Bingqian Qu
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fang Zhang
- Department of Translational Medicine, Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Cindy Zhang
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Wanyan Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Viet Loan Dao Thi
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Yuchen Xia
- State Key Laboratory of Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Antiviral RNAi in Insects and Mammals: Parallels and Differences. Viruses 2019; 11:v11050448. [PMID: 31100912 PMCID: PMC6563508 DOI: 10.3390/v11050448] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022] Open
Abstract
The RNA interference (RNAi) pathway is a potent antiviral defense mechanism in plants and invertebrates, in response to which viruses evolved suppressors of RNAi. In mammals, the first line of defense is mediated by the type I interferon system (IFN); however, the degree to which RNAi contributes to antiviral defense is still not completely understood. Recent work suggests that antiviral RNAi is active in undifferentiated stem cells and that antiviral RNAi can be uncovered in differentiated cells in which the IFN system is inactive or in infections with viruses lacking putative viral suppressors of RNAi. In this review, we describe the mechanism of RNAi and its antiviral functions in insects and mammals. We draw parallels and highlight differences between (antiviral) RNAi in these classes of animals and discuss open questions for future research.
Collapse
|
35
|
Fischer L, Hay DC, O'Farrelly C. Innate immunity in stem cell-derived hepatocytes. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0220. [PMID: 29786555 DOI: 10.1098/rstb.2017.0220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2018] [Indexed: 12/24/2022] Open
Abstract
Stem cell-derived hepatocyte-like cells (HLCs) offer great opportunities for studies of host-pathogen interactions and tissue regeneration, as well as hepatotoxicity. To reliably predict the outcome of infection or to enhance graft survival, a finely tuned innate immune system is essential. Hepatocytes have long been considered solely metabolic and their critical innate immune potential is only recently gaining attention. Viral infection studies show that pathogen detection by cytosolic receptors leads to interferon (IFN) induction in primary hepatocytes and HLCs. IFN expression in HLCs is characterized by strong expression of type III IFN and low expression of type I IFN which is also a characteristic of primary hepatocytes. The response to IFN differs in HLCs with lower interferon-stimulated gene (ISG)-expression levels than in primary hepatocytes. Tumour necrosis factor-alpha (TNF-α) signalling is less studied in HLCs, but appears to be functional. Expression of toll-like receptors (TLR) 2-5, 7 and 9 has been reported in primary hepatocytes but has been poorly studied in HLCs. In summary, although they retain some immature features, HLCs are in many ways superior to hepatoma cell lines for cell-based modelling. In this review, we will provide an overview of innate immune signalling in HLCs and how this compares with primary hepatocytes.This article is part of the themed issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Lena Fischer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Republic of Ireland
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Republic of Ireland
| |
Collapse
|
36
|
Witteveldt J, Knol LI, Macias S. MicroRNA-deficient mouse embryonic stem cells acquire a functional interferon response. eLife 2019; 8:44171. [PMID: 31012846 PMCID: PMC6529217 DOI: 10.7554/elife.44171] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/22/2019] [Indexed: 12/18/2022] Open
Abstract
When mammalian cells detect a viral infection, they initiate a type I interferon (IFNs) response as part of their innate immune system. This antiviral mechanism is conserved in virtually all cell types, except for embryonic stem cells (ESCs) and oocytes which are intrinsically incapable of producing IFNs. Despite the importance of the IFN response to fight viral infections, the mechanisms regulating this pathway during pluripotency are still unknown. Here we show that, in the absence of miRNAs, ESCs acquire an active IFN response. Proteomic analysis identified MAVS, a central component of the IFN pathway, to be actively silenced by miRNAs and responsible for suppressing IFN expression in ESCs. Furthermore, we show that knocking out a single miRNA, miR-673, restores the antiviral response in ESCs through MAVS regulation. Our findings suggest that the interaction between miR-673 and MAVS acts as a switch to suppress the antiviral IFN during pluripotency and present genetic approaches to enhance their antiviral immunity. Living cells are under constant attack from disease-causing agents, such as viruses and bacteria. As a result, they have evolved various protective mechanisms to fight off these agents. One of the most important ways that an animal cell protects itself from infection is through the interferon response, which warns the cell of approaching viruses, prompting it to prepare to defend itself. Virtually all healthy cells have an active interferon response, except for stem cells, which have switched off this defensive mechanism, for unknown reasons. This makes stem cells more susceptible to infections. Stem cells are specialized cells that play an essential role in developing the early embryo. The two defining characteristics of these cells – their ability to divide indefinitely, and develop into all cell types – offers great therapeutic potential, as they can be used to ‘replace’ damaged cells and tissues. However, without an interferon response, stem cells are likely to become infected when moved into a new environment, counteracting their therapeutic benefits. Now, Witteveldt et al. investigate how stem cells turn off this viral defence mechanism, and whether turning it back on will affect their ability to divide and form new tissues. Using stem cells taken from the embryos of mice, Witteveldt et al. found that the interferon response is turned off by specific small molecules of RNA. These small RNA molecules block a protein in the pathway that recognizes viruses and activates a defence. Genetically engineering stem cells to be deficient in these small RNA molecules led to an increased resistance to viral infections. Importantly, modifying stem cells in this manner had no obvious impact on the characteristic traits that give stem cells their therapeutic potential. Temporarily increasing the interferon response of stem cells as they are moved into a new environment could potentially make stem cell treatments more effective. However, more work is needed to investigate whether the same approach can be applied to human cells, and determine what negative effects may be associated with turning on the interferon response.
Collapse
Affiliation(s)
- Jeroen Witteveldt
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lisanne I Knol
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sara Macias
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
37
|
Heterogeneity and coexistence of oncogenic mechanisms involved in HCV-associated B-cell lymphomas. Crit Rev Oncol Hematol 2019; 138:156-171. [PMID: 31092372 DOI: 10.1016/j.critrevonc.2019.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The association of HCV-infection with B-lymphomas is supported by the regression of most indolent/low-grade lymphomas following anti-viral therapy. Studies on direct and indirect oncogenic mechanisms have elucidated the pathogenesis of HCV-associated B-lymphoma subtypes. These include B-lymphocyte proliferation and sustained clonal expansion by HCV-envelope protein stimulation of B-cell receptors, and prolonged HCV-infected B-cell growth by overexpression of an anti-apoptotic BCL-2 oncogene caused by the increased frequency of t(14;18) chromosomal translocations in follicular lymphomas. HCV has been implicated in lymphomagenesis by a "hit-and-run" mechanism, inducing enhanced mutation rate in immunoglobulins and anti-oncogenes favoring immune escape, due to permanent genetic damage by double-strand DNA-breaks. More direct oncogenic mechanisms have been identified in cytokines and chemokines in relation to NS3 and Core expression, particularly in diffuse large B-cell lymphoma. By reviewing genetic alterations and disrupted signaling pathways, we intend to highlight how mutually non-contrasting mechanisms cooperate with environmental factors toward progression of HCV-lymphoma.
Collapse
|
38
|
Dao Thi VL, Wu X, Rice CM. Stem Cell-Derived Culture Models of Hepatitis E Virus Infection. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a031799. [PMID: 29686039 DOI: 10.1101/cshperspect.a031799] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Similar to other hepatotropic viruses, hepatitis E virus (HEV) has been notoriously difficult to propagate in cell culture, limiting studies to unravel its biology. Recently, major advances have been made by passaging primary HEV isolates and selecting variants that replicate efficiently in carcinoma cells. These adaptations, however, can alter HEV biology. We have explored human embryonic or induced pluripotent stem cell (hESC/iPSC)-derived hepatocyte-like cells (HLCs) as an alternative to conventional hepatoma and hepatocyte cell culture systems for HEV studies. HLCs are permissive for nonadapted HEV isolate genotypes (gt)1-4 replication and can be readily genetically manipulated. HLCs, therefore, enable studies of pan-genotype HEV biology and will serve as a platform for testing anti-HEV treatments. Finally, we discuss how hepatocyte polarity is likely an important factor in the maturation and spread of infectious HEV particles.
Collapse
Affiliation(s)
- Viet Loan Dao Thi
- Laboratory of Virology and Infectious Diseases, The Rockefeller University, New York, New York 10065
| | - Xianfang Wu
- Laboratory of Virology and Infectious Diseases, The Rockefeller University, New York, New York 10065
| | - Charles M Rice
- Laboratory of Virology and Infectious Diseases, The Rockefeller University, New York, New York 10065
| |
Collapse
|
39
|
Raasch M, Fritsche E, Kurtz A, Bauer M, Mosig AS. Microphysiological systems meet hiPSC technology - New tools for disease modeling of liver infections in basic research and drug development. Adv Drug Deliv Rev 2019; 140:51-67. [PMID: 29908880 DOI: 10.1016/j.addr.2018.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
Complex cell culture models such as microphysiological models (MPS) mimicking human liver functionality in vitro are in the spotlight as alternative to conventional cell culture and animal models. Promising techniques like microfluidic cell culture or micropatterning by 3D bioprinting are gaining increasing importance for the development of MPS to address the needs for more predictivity and cost efficiency. In this context, human induced pluripotent stem cells (hiPSCs) offer new perspectives for the development of advanced liver-on-chip systems by recreating an in vivo like microenvironment that supports the reliable differentiation of hiPSCs to hepatocyte-like cells (HLC). In this review we will summarize current protocols of HLC generation and highlight recently established MPS suitable to resemble physiological hepatocyte function in vitro. In addition, we are discussing potential applications of liver MPS for disease modeling related to systemic or direct liver infections and the use of MPS in testing of new drug candidates.
Collapse
|
40
|
Kim J, Koo BK, Yoon KJ. Modeling Host-Virus Interactions in Viral Infectious Diseases Using Stem-Cell-Derived Systems and CRISPR/Cas9 Technology. Viruses 2019; 11:v11020124. [PMID: 30704043 PMCID: PMC6409779 DOI: 10.3390/v11020124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Pathologies induced by viral infections have undergone extensive study, with traditional model systems such as two-dimensional (2D) cell cultures and in vivo mouse models contributing greatly to our understanding of host-virus interactions. However, the technical limitations inherent in these systems have constrained efforts to more fully understand such interactions, leading to a search for alternative in vitro systems that accurately recreate in vivo physiology in order to advance the study of viral pathogenesis. Over the last decade, there have been significant technological advances that have allowed researchers to more accurately model the host environment when modeling viral pathogenesis in vitro, including induced pluripotent stem cells (iPSCs), adult stem-cell-derived organoid culture systems and CRISPR/Cas9-mediated genome editing. Such technological breakthroughs have ushered in a new era in the field of viral pathogenesis, where previously challenging questions have begun to be tackled. These include genome-wide analysis of host-virus crosstalk, identification of host factors critical for viral pathogenesis, and the study of viral pathogens that previously lacked a suitable platform, e.g., noroviruses, rotaviruses, enteroviruses, adenoviruses, and Zika virus. In this review, we will discuss recent advances in the study of viral pathogenesis and host-virus crosstalk arising from the use of iPSC, organoid, and CRISPR/Cas9 technologies.
Collapse
Affiliation(s)
- Jihoon Kim
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| |
Collapse
|
41
|
Abstract
Apolipoprotein E (apoE) plays dual functions in the HCV life cycle by promoting HCV infection and virion assembly and production. ApoE is a structural component on the HCV envelope. It mediates HCV cell attachment through specific interactions with the cell surface receptors such as syndecan-1 (SDC-1) and SDC-2 heparan sulfate proteoglycans (HSPGs). It also interacts with NS5A and E2, resulting in an enhancement of HCV morphogenesis. It can bind HCV extracellularly and promotes HCV infection. It is critical for HCV cell-to-cell transmission and may also play a role in HCV persistence by interfering with the action of HCV-neutralizing antibodies. Other apolipoproteins particularly apoB and apoC1 were also found on the HCV envelope, but their roles in the HCV life cycle remain unclear. In the last decade, a number of genomic, immunological, structural, and cell biology methodologies have been developed and used for determining the importance of apoE in the HCV life cycle. These methods and protocols will continue to be valuable to further understand the importance and the underlying molecular mechanism of various apolipoproteins in HCV infection and pathogenesis.
Collapse
Affiliation(s)
- Luhua Qiao
- Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Guangxiang George Luo
- Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
42
|
Wu X, Dao Thi VL. Embryonic or Induced Pluripotent Stem Cell-Derived Hepatocellular Systems for HCV Culture. Methods Mol Biol 2019; 1911:121-135. [PMID: 30593622 DOI: 10.1007/978-1-4939-8976-8_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cell-derived hepatocyte-like cells (iHeps) constitute a powerful tool for modeling hepatotropic pathogen infections in cell culture. Meanwhile, CRISPR-Cas9 technology enables precise editing of stem cell genomes to generate patient-specific disease models and thus development of personalized experimental systems. Here we present a detailed stepwise protocol for the differentiation of stem cells to hepatocyte-like cells for HCV studies in cell culture. We also outline the use of an inducible iCRISPR platform for the rapid and efficient modification of host factors of interest to better understand their function during HCV infection.
Collapse
Affiliation(s)
- Xianfang Wu
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, USA.
| | - Viet Loan Dao Thi
- Center for Integrative Infectious Disease Research, University Hospital Heidelberg, Heidelberg, Germany. .,Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
43
|
Tricot T, Helsen N, Kaptein SJF, Neyts J, Verfaillie CM. Human stem cell-derived hepatocyte-like cells support Zika virus replication and provide a relevant model to assess the efficacy of potential antivirals. PLoS One 2018; 13:e0209097. [PMID: 30566505 PMCID: PMC6300258 DOI: 10.1371/journal.pone.0209097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/29/2018] [Indexed: 01/30/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy has been extensively linked to microcephaly in newborns. High levels of ZIKV RNA were, however, also detected in mice and non-human primates in organs other than the brain, such as the liver. As ZIKV is a flavivirus closely related to the dengue and yellow fever virus, which are known to cause hepatitis, we here examined whether human hepatocytes are susceptible to ZIKV infection. We demonstrated that both human pluripotent stem cell (hPSC)-derived hepatocyte-like cells (HLCs) and the Huh7 hepatoma cell line support the complete ZIKV replication cycle. Of three antiviral molecules that inhibit ZIKV infection in Vero cells, only 7-deaza-2'-C-methyladenosine (7DMA) inhibited ZIKV replication in hPSC-HLCs, while all drugs inhibited ZIKV infection in Huh7 cells. ZIKV-infected hPSC-HLCs but not Huh7 cells mounted an innate immune and NFκβ response, which may explain the more extensive cytopathic effect observed in Huh7 cells. In conclusion, ZIKV productively infects human hepatocytes in vitro. However, significant differences in the innate immune response against ZIKV and antiviral drug sensitivity were observed when comparing hPSC-HLCs and hepatoma cells, highlighting the need to assess ZIKV infection as well as antiviral activity not only in hepatoma cells, but also in more physiologically relevant systems.
Collapse
Affiliation(s)
- Tine Tricot
- Stem Cell Institute, University of Leuven (KU Leuven), Leuven, Belgium
| | - Nicky Helsen
- Stem Cell Institute, University of Leuven (KU Leuven), Leuven, Belgium
| | - Suzanne J F Kaptein
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Johan Neyts
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | | |
Collapse
|
44
|
Grandy R, Tomaz RA, Vallier L. Modeling Disease with Human Inducible Pluripotent Stem Cells. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:449-468. [PMID: 30355153 DOI: 10.1146/annurev-pathol-020117-043634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Understanding the physiopathology of disease remains an essential step in developing novel therapeutics. Although animal models have certainly contributed to advancing this enterprise, their limitation in modeling all the aspects of complex human disorders is one of the major challenges faced by the biomedical research field. Human induced pluripotent stem cells (hiPSCs) derived from patients represent a great opportunity to overcome this deficiency because these cells cover the genetic diversity needed to fully model human diseases. Here, we provide an overview of the history of hiPSC technology and discuss common challenges and approaches that we and others have faced when using hiPSCs to model disease. Our emphasis is on liver disease, and consequently, we review the progress made using this technology to produce functional liver cells in vitro and how these systems are being used to recapitulate a diversity of developmental, metabolic, genetic, and infectious liver disorders.
Collapse
Affiliation(s)
- Rodrigo Grandy
- Wellcome and MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0SZ, United Kingdom; .,Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
| | - Rute A Tomaz
- Wellcome and MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0SZ, United Kingdom; .,Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
| | - Ludovic Vallier
- Wellcome and MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0SZ, United Kingdom; .,Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
| |
Collapse
|
45
|
Eglen RM, Reisine T. Human iPS Cell-Derived Patient Tissues and 3D Cell Culture Part 1: Target Identification and Lead Optimization. SLAS Technol 2018; 24:3-17. [PMID: 30286296 DOI: 10.1177/2472630318803277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human-induced pluripotent stem cells (HiPSCs), and new technologies to culture them into functional cell types and tissues, are now aiding drug discovery. Patient-derived HiPSCs can provide disease models that are more clinically relevant and so more predictive than the currently available animal-derived or tumor cell-derived cells. These cells, consequently, exhibit disease phenotypes close to the human pathology, particularly when cultured under conditions that allow them to recapitulate the tissue architecture in three-dimensional (3D) systems. A key feature of HiPSCs is that they can be cultured under conditions that favor formation of multicellular spheroids or organoids. By culturing and differentiating in systems mimicking the human tissue in vivo, the HiPSC microenvironment further reflects patient in vivo physiology, pathophysiology, and ultimately pharmacological responsiveness. We assess the rationale for using HiPSCs in several phases of preclinical drug discovery, specifically in disease modeling, target identification, and lead optimization. We also discuss the growing use of HiPSCs in compound lead optimization, particularly in profiling compounds for their potential metabolic liability and off-target toxicities. Collectively, we contend that both approaches, HiPSCs and 3D cell culture, when used in concert, have exciting potential for the development of novel medicines.
Collapse
|
46
|
Asumda FZ, Hatzistergos KE, Dykxhoorn DM, Jakubski S, Edwards J, Thomas E, Schiff ER. Differentiation of hepatocyte-like cells from human pluripotent stem cells using small molecules. Differentiation 2018; 101:16-24. [PMID: 29626713 PMCID: PMC6055513 DOI: 10.1016/j.diff.2018.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/18/2018] [Accepted: 03/21/2018] [Indexed: 12/19/2022]
Abstract
A variety of approaches have been developed for the derivation of hepatocyte-like cells from pluripotent stem cells. Currently, most of these strategies employ step-wise differentiation approaches with recombinant growth-factors or small-molecule analogs to recapitulate developmental signaling pathways. Here, we tested the efficacy of a small-molecule based differentiation protocol for the generation of hepatocyte-like cells from human pluripotent stem cells. Quantitative gene-expression, immunohistochemical, and western blot analyses for SOX17, FOXA2, CXCR4, HNF4A, AFP, indicated the stage-specific differentiation into definitive endoderm, hepatoblast and hepatocyte-like derivatives. Furthermore, hepatocyte-like cells displayed morphological and functional features characteristic of primary hepatocytes, as indicated by the production of ALB (albumin) and α-1-antitrypsin (A1AT), as well as glycogen storage capacity by periodic acid-Schiff staining. Together, these data support that the small-molecule based hepatic differentiation protocol is a simple, reproducible, and inexpensive method to efficiently drive the differentiation of human pluripotent stem cells towards a hepatocyte-like phenotype, for downstream pharmacogenomic and regenerative medicine applications.
Collapse
Affiliation(s)
- Faizal Z Asumda
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Silvia Jakubski
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Jasmine Edwards
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Emmanuel Thomas
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Eugene R Schiff
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| |
Collapse
|
47
|
Schöbel A, Rösch K, Herker E. Functional innate immunity restricts Hepatitis C Virus infection in induced pluripotent stem cell-derived hepatocytes. Sci Rep 2018; 8:3893. [PMID: 29497123 PMCID: PMC5832748 DOI: 10.1038/s41598-018-22243-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/20/2018] [Indexed: 12/15/2022] Open
Abstract
Knowledge of activation and interplay between the hepatitis C virus (HCV) and the hosts’ innate immunity is essential to understanding the establishment of chronic HCV infection. Human hepatoma cell lines, widely used as HCV cell culture system, display numerous metabolic alterations and a defective innate immunity, hindering the detailed study of virus-host interactions. Here, we analysed the suitability of induced pluripotent stem cell (iPSC)-derived hepatocyte-like cells (iHLCs) as a physiologically relevant model to study HCV replication in vitro. Density gradients and triglyceride analysis revealed that iHLCs secreted very-low density lipoprotein (VLDL)-like lipoproteins, providing a putative platform for bona fide lipoviroparticles. iHLCs supported the full HCV life cycle, but in contrast to Huh7 and Huh7.5 cells, replication and viral RNA levels decreased continuously. Following HCV infection, interferon-stimulated gene (ISG)-expression significantly increased in iHLCs, whereas induction was almost absent in Huh7/7.5 cells. However, IFNα-stimulation equally induced ISGs in iHLCs and hepatoma cells. JAK-STAT pathway inhibition increased HCV replication in mature iHLCs, but not in Huh7 cells. Additionally, HCV replication levels where higher in STAT2-, but not STAT1-knockdown iHLCs. Our findings support iHLCs as a suitable model for HCV-host interaction regarding a functional innate immunity and lipoprotein synthesis.
Collapse
Affiliation(s)
- Anja Schöbel
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Kathrin Rösch
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Eva Herker
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany.
| |
Collapse
|
48
|
Wu X, Thi VLD, Liu P, Takacs CN, Xiang K, Andrus L, Gouttenoire J, Moradpour D, Rice CM. Pan-Genotype Hepatitis E Virus Replication in Stem Cell-Derived Hepatocellular Systems. Gastroenterology 2018; 154:663-674.e7. [PMID: 29277559 PMCID: PMC5811326 DOI: 10.1053/j.gastro.2017.10.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The 4 genotypes of hepatitis E virus (HEV) that infect humans (genotypes 1-4) vary in geographical distribution, transmission, and pathogenesis. Little is known about the properties of HEV or its hosts that contribute to these variations. Primary isolates grow poorly in cell culture; most studies have relied on variants adapted to cancer cell lines, which likely alter virus biology. We investigated the infection and replication of primary isolates of HEV in hepatocyte-like cells (HLCs) derived from human embryonic and induced pluripotent stem cells. METHODS Using a cell culture-adapted genotype 3 strain and primary isolates of genotypes 1 to 4, we compared viral replication kinetics, sensitivity to drugs, and ability of HEV to activate the innate immune response. We studied HLCs using quantitative reverse-transcriptase polymerase chain reaction and immunofluorescence assay and enzyme-linked immunosorbent assays. We used an embryonic stem cell line that can be induced to express the CRISPR-Cas9 machinery to disrupt the peptidylprolyl isomerase A gene, encoding cyclophilin A (CYPA), a protein reported to inhibit replication of cell culture-adapted HEV. We further modified this line to rescue expression of CYPA before terminal differentiation to HLCs and performed HEV infection studies. RESULTS HLCs were permissive for infection by nonadapted, primary isolates of HEV genotypes 1 to 4. HEV infection of HLCs induced a replication-dependent type III interferon response. Replication of primary HEV isolates, unlike the cell culture-adapted strain, was not affected by disruption of the peptidylprolyl isomerase A gene or exposure to the CYPA inhibitor cyclosporine A. CONCLUSIONS Cell culture adaptations alter the replicative capacities of HEV. HLCs offer an improved, physiologically relevant, and genetically tractable system for studying the replication of primary HEV isolates. HLCs could provide a model to aid development of HEV drugs and a system to guide personalized regimens, especially for patients with chronic hepatitis E who have developed resistance to ribavirin.
Collapse
Affiliation(s)
- Xianfang Wu
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Viet Loan Dao Thi
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Peng Liu
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA,Department of Scientific Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Constantin N. Takacs
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA,Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Kuanhui Xiang
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA,Department of Microbiology and Center of Infectious Disease, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China
| | - Linda Andrus
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Jérôme Gouttenoire
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Charles M Rice
- Laboratory of Virology and Infectious Diseases, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York.
| |
Collapse
|
49
|
Wu X, Dao Thi VL, Huang Y, Billerbeck E, Saha D, Hoffmann HH, Wang Y, Silva LAV, Sarbanes S, Sun T, Andrus L, Yu Y, Quirk C, Li M, MacDonald MR, Schneider WM, An X, Rosenberg BR, Rice CM. Intrinsic Immunity Shapes Viral Resistance of Stem Cells. Cell 2017; 172:423-438.e25. [PMID: 29249360 DOI: 10.1016/j.cell.2017.11.018] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/11/2017] [Accepted: 11/09/2017] [Indexed: 12/26/2022]
Abstract
Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance.
Collapse
Affiliation(s)
- Xianfang Wu
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Viet Loan Dao Thi
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yumin Huang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; Department of Hematology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Eva Billerbeck
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Debjani Saha
- Program in Immunogenomics, The Rockefeller University, New York, NY 10065, USA
| | - Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | | | - Stephanie Sarbanes
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Tony Sun
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Linda Andrus
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yingpu Yu
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Corrine Quirk
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Melody Li
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - William M Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Brad R Rosenberg
- Program in Immunogenomics, The Rockefeller University, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
50
|
Gural N, Mancio-Silva L, He J, Bhatia SN. Engineered Livers for Infectious Diseases. Cell Mol Gastroenterol Hepatol 2017; 5:131-144. [PMID: 29322086 PMCID: PMC5756057 DOI: 10.1016/j.jcmgh.2017.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/02/2017] [Indexed: 01/18/2023]
Abstract
Engineered liver systems come in a variety of platform models, from 2-dimensional cocultures of primary human hepatocytes and stem cell-derived progeny, to 3-dimensional organoids and humanized mice. Because of the species-specificity of many human hepatropic pathogens, these engineered systems have been essential tools for biologic discovery and therapeutic agent development in the context of liver-dependent infectious diseases. Although improvement of existing models is always beneficial, and the addition of a robust immune component is a particular need, at present, considerable progress has been made using this combination of research platforms. We highlight advances in the study of hepatitis B and C viruses and malaria-causing Plasmodium falciparum and Plasmodium vivax parasites, and underscore the importance of pairing the most appropriate model system and readout modality with the particular experimental question at hand, without always requiring a platform that recapitulates human physiology in its entirety.
Collapse
Key Words
- 2D, 2-dimensional
- 3D
- 3D, 3-dimensional
- EBOV, Ebola virus
- Falciparum
- HBC, hepatitis C virus
- HBV
- HBV, hepatitis B virus
- HCV
- HLC, hepatocyte-like cells
- Hepatotropic
- LASV, Lassa virus
- Liver
- Liver Models
- MPCC, micropatterned coculture system
- Malaria
- PCR, polymerase chain reaction
- Pathogen
- SACC, self-assembling coculture
- Vivax
- iHLC, induced pluripotent stem cell–derived hepatocyte-like cells
- in vitro
- in vivo
Collapse
Affiliation(s)
- Nil Gural
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, Massachusetts,Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liliana Mancio-Silva
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jiang He
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sangeeta N. Bhatia
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Broad Institute, Cambridge, Massachusetts,Howard Hughes Medical Institute, Chevy Chase, Maryland,Correspondence Address correspondence to: Sangeeta N. Bhatia, MD, PhD, Koch Institute for Integrative Cancer, Research at MIT, Building 76, Room 473, 500 Main Street, Cambridge, Massachusetts 02142.
| |
Collapse
|