151
|
Kostyusheva AP, Kostyushev DS, Brezgin SA, Zarifyan DN, Volchkova EV, Chulanov VP. Small Molecular Inhibitors of DNA Double Strand Break Repair Pathways Increase the ANTI-HBV Activity of CRISPR/Cas9. Mol Biol 2019. [DOI: 10.1134/s0026893319010072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
152
|
Lauschke VM, Shafagh RZ, Hendriks DFG, Ingelman-Sundberg M. 3D Primary Hepatocyte Culture Systems for Analyses of Liver Diseases, Drug Metabolism, and Toxicity: Emerging Culture Paradigms and Applications. Biotechnol J 2019; 14:e1800347. [PMID: 30957976 DOI: 10.1002/biot.201800347] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/26/2019] [Indexed: 02/06/2023]
Abstract
Recent research has shown that the maintenance of relevant liver functions ex vivo requires models in which the cells exhibit an in vivo-like phenotype, often achieved by reconstitution of appropriate cellular interactions. Multiple different models have been presented that differ in the cells utilized, media, and culture conditions. Furthermore, several technologically different approaches have been presented including bioreactors, chips, and plate-based systems in fluidic or static media constituting of chemically diverse materials. Using such models, the ability to predict drug metabolism, drug toxicity, and liver functionality have increased tremendously as compared to conventional in vitro models in which cells are cultured as 2D monolayers. Here, the authors highlight important considerations for microphysiological systems for primary hepatocyte culture, review current culture paradigms, and discuss their opportunities for studies of drug metabolism, hepatotoxicity, liver biology, and disease.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Reza Z Shafagh
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Department of Micro and Nanosystems, School of Electrical Engineering, Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Delilah F G Hendriks
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Present address: Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| |
Collapse
|
153
|
Zhang Z, Xu H, Mazza G, Zhang M, Frenguelli L, Liu Q, Al-Akkad W, Ren J, Zhao R, Ren F, Chen X, Huang A, Chen J. Decellularized human liver scaffold-based three-dimensional culture system facilitate hepatitis B virus infection. J Biomed Mater Res A 2019; 107:1744-1753. [PMID: 30963688 DOI: 10.1002/jbm.a.36690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/18/2019] [Accepted: 03/15/2019] [Indexed: 12/21/2022]
Abstract
Hepatitis B virus (HBV) study is hampered by lacking of idea cell model which support effective HBV infection and meanwhile recapitulate hepatocyte biology function in vivo. In this study, we developed decellularized human liver scaffolds for cell culture and further applied for HBV infection. As a result, primary human hepatocytes (PHHs) engrafted into liver scaffolds and maintained differentiation with stable albumin secretion and liver-specific gene expression. Comparing to mono-layer cell culture, scaffold-based three-dimensional (3D) culture system significantly augment HBV DNA (including cccDNA), RNA level as well as HBsAg secretion. Moreover, HepG2-NTCP cells cultured on 3D system exhibited higher infection efficiency and longer infection period in vitro. In addition, HBV DNA level was suppressed when anti-HBV medicine Entecavir (ETV) introduced into HepG2-NTCP 3D system. Herein, we evaluated the potential of decellularized human liver scaffold-based in 3D cell culture and disclosed that scaffold-based 3D culture system can facilitate HBV infection in vitro. This 3D culture system could be further applied in HBV-related study. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1744-1753, 2019.
Collapse
Affiliation(s)
- ZhenZhen Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, ChongQing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Department of Infectious Disease, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - HongMei Xu
- Ministry of Education Key Laboratory of Child Development and Disorders, ChongQing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Department of Infectious Disease, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - Giuseppe Mazza
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - MingMan Zhang
- Department of Hepatobiliary Surgery, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - Luca Frenguelli
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - QuanBo Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, ChongQing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Department of Infectious Disease, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - Walid Al-Akkad
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - JiHua Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, ChongQing, China
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, ChongQing, 400016, China
| | - RuiQiu Zhao
- Ministry of Education Key Laboratory of Child Development and Disorders, ChongQing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Department of Infectious Disease, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - Fang Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, ChongQing, China
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, ChongQing, 400016, China
| | - Xin Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, ChongQing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- The General Gard, Children's Hospital of ChongQing Medical University, ChongQing, China
| | - AiLong Huang
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, ChongQing, China
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, ChongQing, 400016, China
| | - Juan Chen
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, ChongQing, China
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, ChongQing, 400016, China
| |
Collapse
|
154
|
A global scientific strategy to cure hepatitis B. Lancet Gastroenterol Hepatol 2019; 4:545-558. [PMID: 30981686 DOI: 10.1016/s2468-1253(19)30119-0] [Citation(s) in RCA: 362] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Chronic hepatitis B virus (HBV) infection is a global public health challenge on the same scale as tuberculosis, HIV, and malaria. The International Coalition to Eliminate HBV (ICE-HBV) is a coalition of experts dedicated to accelerating the discovery of a cure for chronic hepatitis B. Following extensive consultation with more than 50 scientists from across the globe, as well as key stakeholders including people affected by HBV, we have identified gaps in our current knowledge and new strategies and tools that are required to achieve HBV cure. We believe that research must focus on the discovery of interventional strategies that will permanently reduce the number of productively infected cells or permanently silence the covalently closed circular DNA in those cells, and that will stimulate HBV-specific host immune responses which mimic spontaneous resolution of HBV infection. There is also a pressing need for the establishment of repositories of standardised HBV reagents and protocols that can be accessed by all HBV researchers throughout the world. The HBV cure research agenda outlined in this position paper will contribute markedly to the goal of eliminating HBV infection worldwide.
Collapse
|
155
|
Deng J, Zhang X, Chen Z, Luo Y, Lu Y, Liu T, Wu Z, Jin Y, Zhao W, Lin B. A cell lines derived microfluidic liver model for investigation of hepatotoxicity induced by drug-drug interaction. BIOMICROFLUIDICS 2019; 13:024101. [PMID: 31040885 PMCID: PMC6456354 DOI: 10.1063/1.5070088] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/28/2019] [Indexed: 05/18/2023]
Abstract
The poor metabolic ability of cell lines fails to meet the requirements of an in vitro model for drug interaction testing which is crucial for the development or clinical application of drugs. Herein, we describe a liver sinusoid-on-a-chip device composed of four kinds of transformed cell lines (HepG2 cells, LX-2 cells, EAhy926 cells, and U937 cells) that were ordered in a physiological distribution with artificial liver blood flow and biliary efflux flowing in the opposite direction. This microfluidic device applied three-dimensional culturing of HepG2 cells with high density (107 ml-1), forming a tightly connected monolayer of EAhy926 cells and achieving the active transport of drugs in HepG2 cells. Results showed that the device maintained synthetic and secretory functions, preserved cytochrome P450 1A1/2 and uridine diphosphate glucuronyltransferase enzymatic activities, as well as sensitivity of drug metabolism. The cell lines derived device enables the investigation of a drug-drug interaction study. We used it to test the hepatotoxicity of acetaminophen and the following combinations: "acetaminophen + rifampicin," "acetaminophen + omeprazole," and "acetaminophen + ciprofloxacin." The variations in hepatotoxicity of the combinations compared to acetaminophen alone, which is not found in a 96-well plate model, in the device were -17.15%, 14.88%, and -19.74%. In addition, this result was similar to the one tested by the classical primary hepatocyte plate model (-13.22%, 13.51%, and -15.81%). Thus, this cell lines derived liver model provides an alternative to investigate drug hepatotoxicity, drug-drug interaction.
Collapse
Affiliation(s)
- Jiu Deng
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Soochow 215123, China
| | - Zongzheng Chen
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Yao Lu
- Biotechnologhy Division, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116011, China
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Zhengzhi Wu
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yu Jin
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Bingcheng Lin
- Authors to whom correspondence should be addressed: ; ; and
| |
Collapse
|
156
|
Abstract
Diabetes mellitus is a multifactorial disease affecting increasing numbers of patients worldwide. Progression to insulin-dependent diabetes mellitus is characterized by the loss or dysfunction of pancreatic β-cells, but the pathomechanisms underlying β-cell failure in type 1 diabetes mellitus and type 2 diabetes mellitus are still poorly defined. Regeneration of β-cell mass from residual islet cells or replacement by β-like cells derived from stem cells holds great promise to stop or reverse disease progression. However, the development of new treatment options is hampered by our limited understanding of human pancreas organogenesis due to the restricted access to primary tissues. Therefore, the challenge is to translate results obtained from preclinical model systems to humans, which requires comparative modelling of β-cell biology in health and disease. Here, we discuss diverse modelling systems across different species that provide spatial and temporal resolution of cellular and molecular mechanisms to understand the evolutionary conserved genotype-phenotype relationship and translate them to humans. In addition, we summarize the latest knowledge on organoids, stem cell differentiation platforms, primary micro-islets and pseudo-islets, bioengineering and microfluidic systems for studying human pancreas development and homeostasis ex vivo. These new modelling systems and platforms have opened novel avenues for exploring the developmental trajectory, physiology, biology and pathology of the human pancreas.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Technical University of Munich, Medical Faculty, Munich, Germany.
| |
Collapse
|
157
|
Feld JJ, Gehring AJ. Host-pathogen interactions in chronic HBV infection and transplantation of HCV-positive organs. Nat Rev Gastroenterol Hepatol 2019; 16:77-78. [PMID: 30617296 DOI: 10.1038/s41575-018-0101-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jordan J Feld
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada. .,Institute for Medical Science, University of Toronto, Toronto, Canada.
| | - Adam J Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Institute for Medical Science, University of Toronto, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
158
|
Hu J, Lin YY, Chen PJ, Watashi K, Wakita T. Cell and Animal Models for Studying Hepatitis B Virus Infection and Drug Development. Gastroenterology 2019; 156:338-354. [PMID: 30243619 PMCID: PMC6649672 DOI: 10.1053/j.gastro.2018.06.093] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
Abstract
Many cell culture and animal models have been used to study hepatitis B virus (HBV) replication and its effects in the liver; these have facilitated development of strategies to control and clear chronic HBV infection. We discuss the advantages and limitations of systems for studying HBV and developing antiviral agents, along with recent advances. New and improved model systems are needed. Cell culture systems should be convenient, support efficient HBV infection, and reproduce responses of hepatocytes in the human body. We also need animals that are fully permissive to HBV infection, convenient for study, and recapitulate human immune responses to HBV and effects in the liver. High-throughput screening technologies could facilitate drug development based on findings from cell and animal models.
Collapse
Affiliation(s)
- Jianming Hu
- The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| | - You-Yu Lin
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Hepatitis Research Center, National Taiwan University Hospital, National Taiwan University.
| | | | - Takaji Wakita
- National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
159
|
Shapshak P, Balaji S, Kangueane P, Chiappelli F, Somboonwit C, Menezes LJ, Sinnott JT. Innovative Technologies for Advancement of WHO Risk Group 4 Pathogens Research. GLOBAL VIROLOGY III: VIROLOGY IN THE 21ST CENTURY 2019. [PMCID: PMC7122670 DOI: 10.1007/978-3-030-29022-1_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Paul Shapshak
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| | - Seetharaman Balaji
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka India
| | | | - Francesco Chiappelli
- Oral Biology and Medicine, CHS 63-090, UCLA School of Dentistry Oral Biology and Medicine, CHS 63-090, Los Angeles, CA USA
| | | | - Lynette J. Menezes
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| | - John T. Sinnott
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| |
Collapse
|
160
|
Abstract
In native tissues, various cell types organize and spatiotemporally function and communicate with neighboring or remote cells in a highly regulated way. How can we replicate these amazing functional structures in vitro? From the view of a chemist, the heterogeneous cells and extracellular matrix (ECM) could be regarded as various chemical substrate materials for "synthetic" reactions during tissue engineering. But how can we accelerate these reactions? Microfluidics provides ideal solutions. Microfluidics could be metaphorically regarded as a miniature "biofactory", whereas the on-chip critical chemical cues such as biomolecule gradients and physical cues such as geometrical confinement, topological guidance, and mechanical stimulations, along with the external stimulations such as light, electricity, acoustics, and magnetics, could be regarded as "catalytic cues" which can accelerate the "synthetic reactions" by precisely and effectively manipulating a series of cell behaviors including cell adhesion, migration, growth, proliferation, differentiation, cell-cell interaction, and cell-matrix interaction to reduce activation energy of the "synthetic reactions". Thus, on the microfluidics platform, the "biofactory", various "synthetic" reactions take place to change the substrate materials (cells and ECM) into products (tissues) in a nonlinear way, which is a typical feature of a biological process. By precisely organizing the substrate materials and spatiotemporally controlling the activity of the products, as a "biofactory", the microfluidics system can not only "synthesize" living tissues but also recreate physiological or pathophysiological processes such as immune responses, angiogenesis, wound healing, and tumor metastasis in vitro to bring insights into the mechanisms underlying these processes taking place in vivo. In this Account, we borrow the concept of chemical "synthesis" to describe how to "synthesize" artificial tissues using microfluidics from a chemist's view. Accelerated by the built-in physiochemical cues on microfluidics and external stimulations, various tissues could be "synthesized" on a microfluidics platform. We summarize that there are "step-by-step synthesis" and "one-step synthesis" on microfluidics for creating desired tissues with unprecedented precision, accuracy, and speed. In recent years, researchers developed various microfluidic techniques including creating adhesive domains for mediating reverse and precise adhesion, chemical gradients for directing cell growth, geometrical confinements and topological cues for manipulating cell migration, and mechanics for stimulating cell differentiation. By employing and orchestrating these on-chip tissue "synthetic" conditions, "step-by-step synthesis" could be realized on chips to develop multilayered tissues such as blood vessels. "One-step synthesis" on chips could develop functional three-dimensional tissue structures such as neural networks or nephron-like structures. Based on these on-chip studies, many critical physiological and pathophysiological processes such as wound healing, tumor metastasis, and atherosclerosis could be deeply investigated, and the drugs or therapeutic approaches could also be evaluated or screened conveniently. The "synthetic tissues on microfluidics" system would pave an avenue for precise creation of artificial tissues for not only fundamental research but also biomedical applications such as tissue engineering.
Collapse
Affiliation(s)
- Wenfu Zheng
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, P. R. China
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, P. R. China
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
- The University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
161
|
Verrier ER, Yim SA, Heydmann L, El Saghire H, Bach C, Turon-Lagot V, Mailly L, Durand SC, Lucifora J, Durantel D, Pessaux P, Manel N, Hirsch I, Zeisel MB, Pochet N, Schuster C, Baumert TF. Hepatitis B Virus Evasion From Cyclic Guanosine Monophosphate-Adenosine Monophosphate Synthase Sensing in Human Hepatocytes. Hepatology 2018; 68:1695-1709. [PMID: 29679386 PMCID: PMC6195855 DOI: 10.1002/hep.30054] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023]
Abstract
Chronic hepatitis B virus (HBV) infection is a major cause of chronic liver disease and cancer worldwide. The mechanisms of viral genome sensing and the evasion of innate immune responses by HBV infection are still poorly understood. Recently, the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) was identified as a DNA sensor. In this study, we investigated the functional role of cGAS in sensing HBV infection and elucidate the mechanisms of viral evasion. We performed functional studies including loss-of-function and gain-of-function experiments combined with cGAS effector gene expression profiling in an infectious cell culture model, primary human hepatocytes, and HBV-infected human liver chimeric mice. Here, we show that cGAS is expressed in the human liver, primary human hepatocytes, and human liver chimeric mice. While naked relaxed-circular HBV DNA is sensed in a cGAS-dependent manner in hepatoma cell lines and primary human hepatocytes, host cell recognition of viral nucleic acids is abolished during HBV infection, suggesting escape from sensing, likely during packaging of the genome into the viral capsid. While the hepatocyte cGAS pathway is functionally active, as shown by reduction of viral covalently closed circular DNA levels in gain-of-function studies, HBV infection suppressed cGAS expression and function in cell culture models and humanized mice. Conclusion: HBV exploits multiple strategies to evade sensing and antiviral activity of cGAS and its effector pathways.
Collapse
Affiliation(s)
- Eloi R. Verrier
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France,Corresponding authors: Prof. Thomas F. Baumert, MD, , Dr. Catherine Schuster, PhD, , and Dr. Eloi R. Verrier, PhD, , Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000 Strasbourg, France. Tel: +33 3 68 85 37 03; fax: +33 3 68 85 37 24
| | - Seung-Ae Yim
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Laura Heydmann
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Houssein El Saghire
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Charlotte Bach
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Vincent Turon-Lagot
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Laurent Mailly
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Sarah C. Durand
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Julie Lucifora
- Inserm, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, Centre Léon Bérard, Lyon, France
| | - David Durantel
- Inserm, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, Centre Léon Bérard, Lyon, France
| | - Patrick Pessaux
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France,Pôle Hépato-Digestif, Institut Hospitalo-Universitaire, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, F-75005 Paris, France,Inserm, U932, F-75005 Paris, France
| | - Ivan Hirsch
- Department of Genetics and Microbiology, Faculty of Science, Biocev, Charles University, 12844 Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry, CAS, IOCB & Gilead Research Center, 16610 Prague
| | - Mirjam B. Zeisel
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France
| | - Nathalie Pochet
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Cell Circuits Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Catherine Schuster
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France,Corresponding authors: Prof. Thomas F. Baumert, MD, , Dr. Catherine Schuster, PhD, , and Dr. Eloi R. Verrier, PhD, , Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000 Strasbourg, France. Tel: +33 3 68 85 37 03; fax: +33 3 68 85 37 24
| | - Thomas F. Baumert
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMRS 1110, F-67000 Strasbourg, France,Pôle Hépato-Digestif, Institut Hospitalo-Universitaire, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France,Corresponding authors: Prof. Thomas F. Baumert, MD, , Dr. Catherine Schuster, PhD, , and Dr. Eloi R. Verrier, PhD, , Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000 Strasbourg, France. Tel: +33 3 68 85 37 03; fax: +33 3 68 85 37 24
| |
Collapse
|
162
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
163
|
Nie YZ, Zheng YW, Miyakawa K, Murata S, Zhang RR, Sekine K, Ueno Y, Takebe T, Wakita T, Ryo A, Taniguchi H. Recapitulation of hepatitis B virus-host interactions in liver organoids from human induced pluripotent stem cells. EBioMedicine 2018; 35:114-123. [PMID: 30120080 PMCID: PMC6156717 DOI: 10.1016/j.ebiom.2018.08.014] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 07/23/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Therapies against hepatitis B virus (HBV) have improved in recent decades; however, the development of individualized treatments has been limited by the lack of individualized infection models. In this study, we used human induced pluripotent stem cell (hiPSC) to generate a functional liver organoid (LO) that inherited the genetic background of the donor, and evaluated its application in modeling HBV infection and exploring virus-host interactions. To establish a functional hiPSC-LO, we cultured hiPSC-derived endodermal, mesenchymal, and endothelial cells with a chemically defined medium in a three-dimensional microwell culture system. Based on cell-cell interactions, these cells could organize themselves and gradually differentiate into a functional organoid, which exhibited stronger hepatic functions than hiPSC derived hepatic like cell (HLC). Moreover, the functional LO demonstrated more susceptibility to HBV infection than hiPSC-HLC, and could maintain HBV propagation and produce infectious virus for a prolonged duration. Furthermore, we found that virus infection could cause hepatic dysfunction of hiPSC-LOs, with down-regulation of hepatic gene expression, induced release of early acute liver failure markers, and altered hepatic ultrastructure. Therefore, our study demonstrated that HBV infection in hiPSC-LOs could recapitulate virus life cycle and virus induced hepatic dysfunction, suggesting that hiPSC-LOs may provide a promising individualized infection model for the development of individualized treatment for hepatitis.
Collapse
Affiliation(s)
- Yun-Zhong Nie
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yun-Wen Zheng
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan; Department of Advanced Gastroenterological Surgical Science and Technology, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8575, Japan; Research Center of Stem Cells and Regenerative Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China,.
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Ran-Ran Zhang
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640 Tokyo, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan; Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan.
| |
Collapse
|
164
|
3D microfluidic in vitro model and bioinformatics integration to study the effects of Spatholobi Caulis tannin in cervical cancer. Sci Rep 2018; 8:12285. [PMID: 30115981 PMCID: PMC6095931 DOI: 10.1038/s41598-018-29848-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023] Open
Abstract
Cervical cancer is considered the fourth most common malignant disease in women. Recently, tannin from Spatholobi Caulis (TTS) has been shown to have potent anticancer and antiproliferative characteristics in a few preliminary studies. This experiment used 3D microfluidic, flow cytometry, and gene chip technology to study the efficacy and mechanism of action of TTS, as well as molecular docking technology to study the effect of drugs on related proteins. The cell survival rates of the five groups measured by the 3D microfluidic chip were 94%, 85%, 64%, 55%, and 42%, respectively. With the increase in drug concentration, the cell survival rate gradually decreased. Apoptosis rates detected in the five groups were 2.12%, 15.87%, 33.40%, 41.13%, and 55.10%, respectively. These data suggest that TTS can promote cell apoptosis. The percentages of cells in the G0/G1 phase were 43.39%, 55.07%, 59.57%, 64.56%, and 67.39% in the five groups, respectively. TTS was demonstrated to inhibit the conversion of cells from G0/G1 to S phase and G2/M phase and inhibit gene and protein synthesis to block cell proliferation. TTS can effectively modulate pathogenic proteins. The results confirmed the efficacy of TTS against HeLa cells and that TTS can be used as an adjunct in cervical cancer prevention and treatment.
Collapse
|
165
|
Tan G, Song H, Xu F, Cheng G. When Hepatitis B Virus Meets Interferons. Front Microbiol 2018; 9:1611. [PMID: 30072974 PMCID: PMC6058040 DOI: 10.3389/fmicb.2018.01611] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection imposes a severe burden on global public health. Currently, there are no curative therapies for millions of chronic HBV-infected patients (Lok et al., 2017). Interferon (IFN; including pegylated IFN) is an approved anti-HBV drug that not only exerts direct antiviral activity, but also augments immunity against HBV infection. Through a systematic review of the literature, here we summarize and present recent progress in research regarding the interactions between IFN and HBV as well as dissect the antiviral mechanisms of IFN. We focus on inhibition of HBV replication by IFN-stimulated genes (ISGs) as well as inhibition of IFN signaling by HBV and viral proteins. Finally, we briefly discuss current IFN-based HBV treatment strategies. This review may help to better understand the mechanisms involved in the therapeutic action of IFN as well as the crosstalk between IFN and HBV, and facilitate the development of both direct-acting and immunology-based new HBV drugs.
Collapse
Affiliation(s)
- Guangyun Tan
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hongxiao Song
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fengchao Xu
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Genhong Cheng
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States.,Center of System Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Suzhou Institute of Systems Medicine, Suzhou, China
| |
Collapse
|
166
|
Thomas H. Viral hepatitis: A new model for HBV infection of primary human hepatocytes. Nat Rev Gastroenterol Hepatol 2018; 15:190. [PMID: 29487421 DOI: 10.1038/nrgastro.2018.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|