1
|
Simoneau CR, Chen PY, Xing GK, Hayashi JM, Chen IP, Khalid MM, Meyers NL, Taha TY, Leon KE, Suryawanshi RK, McCavitt-Malvido M, Ashuach T, Fontaine KA, Rodriguez L, Joehnk B, Walcott K, Vasudevan S, Fang X, Maishan M, Schultz S, Roose JP, Matthay MA, Sil A, Arjomandi M, Yosef N, Ott M. NF-κB inhibitor alpha controls SARS-CoV-2 infection in ACE2-overexpressing human airway organoids. Sci Rep 2024; 14:15351. [PMID: 38961189 PMCID: PMC11222426 DOI: 10.1038/s41598-024-66003-2] [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: 05/23/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
As SARS-CoV-2 continues to spread worldwide, tractable primary airway cell models that recapitulate the cell-intrinsic response to arising viral variants are needed. Here we describe an adult stem cell-derived human airway organoid model overexpressing the ACE2 receptor (ACE2-OE) that supports robust viral replication while maintaining 3D architecture and cellular diversity of the airway epithelium. ACE2-OE organoids were infected with SARS-CoV-2 variants and subjected to single-cell RNA-sequencing. Interferon-lambda was upregulated in cells with low-level infection while the NF-kB inhibitor alpha gene (encoding IkBa) was consistently upregulated in infected cells, and its expression positively correlated with infection levels. Confocal microscopy showed more IkBa expression in infected than bystander cells, but found concurrent nuclear translocation of NF-kB that IkBa usually prevents. Overexpressing a nondegradable IkBa mutant reduced NF-kB translocation and increased viral infection. These data demonstrate the functionality of ACE2-OE organoids in SARS-CoV-2 research and underscore that the strength of the NF-kB feedback loop in infected cells controls viral replication.
Collapse
Affiliation(s)
- Camille R Simoneau
- Gladstone Institute of Virology, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Pei-Yi Chen
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Galen K Xing
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | - Irene P Chen
- Gladstone Institute of Virology, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Mir M Khalid
- Gladstone Institute of Virology, San Francisco, CA, USA
| | | | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Kristoffer E Leon
- Gladstone Institute of Virology, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Tal Ashuach
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | - Lauren Rodriguez
- ImmunoX CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Bastian Joehnk
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Keith Walcott
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | | | - Xiaohui Fang
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Mazharul Maishan
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Shawn Schultz
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Mehrdad Arjomandi
- Medical Service, San Francisco VA Healthcare System, San Francisco, CA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Chan-Zuckerberg Biohub, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Baptista LS, Mironov V, Koudan E, Amorim ÉA, Pampolha TP, Kasyanov V, Kovalev A, Senatov F, Granjeiro JM. Bioprinting Using Organ Building Blocks: Spheroids, Organoids, and Assembloids. Tissue Eng Part A 2024; 30:377-386. [PMID: 38062998 DOI: 10.1089/ten.tea.2023.0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Three-dimensional (3D) bioprinting, a promising advancement in tissue engineering technology, involves the robotic, layer-by-layer additive biofabrication of functional 3D tissue and organ constructs. This process utilizes biomaterials, typically hydrogels and living cells, following digital models. Traditional tissue engineering uses a classic triad of living cells, scaffolds, and physicochemical signals in bioreactors. A scaffold is a temporary, often biodegradable, support structure. Tissue engineering primarily falls into two categories: (i) scaffold based and (ii) scaffold free. The latter, scaffold-free 3D bioprinting, is gaining increasing popularity. Organ building blocks (OBB), capable of self-assembly and self-organization, such as tissue spheroids, organoids, and assembloids, have begun to be utilized in scaffold-free bioprinting. This article discusses the expanding range of OBB, presents the rapidly evolving collection of bioprinting and bioassembly methods using these OBB, and finally, outlines the advantages, challenges, and future perspectives of using OBB in organ printing.
Collapse
Affiliation(s)
- Leandra Santos Baptista
- Campus Duque de Caxias Prof Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality, and Technology (Inmetro), Rio de Janeiro, Brazil
- Laboratory of Eukaryotic Cell Biology, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
| | - Vladimir Mironov
- Campus Duque de Caxias Prof Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elizaveta Koudan
- Center for Biomedical Engineering, National University of Science and Technology "MISIS," Moscow, Russia
| | - Érica Almeida Amorim
- Campus Duque de Caxias Prof Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Gcell 3D, Rio de Janeiro, Brazil
- Precision Medicine Research Center, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tathiana Proença Pampolha
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality, and Technology (Inmetro), Rio de Janeiro, Brazil
- Laboratory of Eukaryotic Cell Biology, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
| | - Vladimir Kasyanov
- Joint Laboratory of Traumatology and Orthopaedics, Riga Stradins University, Riga, Latvia
| | - Alexei Kovalev
- Priorov Central National Institute of Traumatology and Orthopedics, Moscow, Russia
| | - Fedor Senatov
- Center for Biomedical Engineering, National University of Science and Technology "MISIS," Moscow, Russia
| | - José Mauro Granjeiro
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality, and Technology (Inmetro), Rio de Janeiro, Brazil
- Laboratory of Eukaryotic Cell Biology, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói, Brazil
| |
Collapse
|
3
|
Iqbal W, Wang Y, Sun P, Zhou X. Modeling Liver Development and Disease in a Dish. Int J Mol Sci 2023; 24:15921. [PMID: 37958904 PMCID: PMC10650907 DOI: 10.3390/ijms242115921] [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: 09/12/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Historically, biological research has relied primarily on animal models. While this led to the understanding of numerous human biological processes, inherent species-specific differences make it difficult to answer certain liver-related developmental and disease-specific questions. The advent of 3D organoid models that are either derived from pluripotent stem cells or generated from healthy or diseased tissue-derived stem cells have made it possible to recapitulate the biological aspects of human organs. Organoid technology has been instrumental in understanding the disease mechanism and complements animal models. This review underscores the advances in organoid technology and specifically how liver organoids are used to better understand human-specific biological processes in development and disease. We also discuss advances made in the application of organoid models in drug screening and personalized medicine.
Collapse
Affiliation(s)
- Waqas Iqbal
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Yaru Wang
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Pingnan Sun
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Xiaoling Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China; (W.I.); (Y.W.); (P.S.)
- Research Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
4
|
Ren B, Chiaravalloti TR, Belony NL, Romero DI, Chai W, Leon C, Wu L, Lamango NS, Offringa IA, Huang Y. Design and Realization of Lung Organoid Cultures for COVID-19 Applications. Biodes Manuf 2023; 6:646-660. [PMID: 38993804 PMCID: PMC11238720 DOI: 10.1007/s42242-023-00255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/25/2023] [Indexed: 07/13/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been spreading globally and threatening public health. Advanced in vitro models that recapitulate the architecture and functioning of specific tissues and organs are in high demand for COVID-19-related pathology studies and drug screening. Three-dimensional (3D) in vitro cultures such as self-assembled and engineered organoid cultures surpass conventional two-dimensional (2D) cultures and animal models with respect to the increased cellular complexity, better human-relevant environment, and reduced cost, thus presenting as promising platforms for understanding viral pathogenesis and developing new therapeutics. This review highlights the recent advances in self-assembled and engineered organoid technologies that are used for COVID-19 studies. The challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | | | - Nadine L Belony
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Diana I Romero
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Wenxuan Chai
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Christopher Leon
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32611
| | - Nazarius S Lamango
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307
| | - Ite A Offringa
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| |
Collapse
|
5
|
Sanyal S. Crossroads in virology: current challenges and future perspectives in the age of emerging viruses. Dis Model Mech 2023; 16:dmm050476. [PMID: 37728628 PMCID: PMC10538290 DOI: 10.1242/dmm.050476] [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] [Indexed: 09/21/2023] Open
Abstract
Ongoing global health challenges posed by emerging and re-emerging viruses have highlighted the critical importance of understanding virus-host interactions in countering these threats. Environmental changes, urbanisation and ecological disruption, coupled with the adaptable nature of viruses, facilitates the emergence and spread of new viruses. This Editorial emphasises the urgency of a concerted effort in understanding virus-host interactions to inform the development of therapeutics and vaccines, and help predict disease outcomes. Furthermore, efforts to monitor viral evolution, identify mutations of concern, and develop 'universal' vaccines and broad-spectrum antiviral drugs are needed to counter viral evolution and potentially prevent future viral emergences. Widespread public mistrust surrounding viruses and vaccines also calls for improvement in science communication. A 'One Health' approach that advocates the development of robust global health systems, interdisciplinary collaborations and equity in health access is therefore imperative for transforming the virology landscape.
Collapse
Affiliation(s)
- Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| |
Collapse
|
6
|
Santos AK, Scalzo S, de Souza RTV, Santana PHG, Marques BL, Oliveira LF, Filho DM, Kihara AH, da Costa Santiago H, Parreira RC, Birbrair A, Ulrich H, Resende RR. Strategic use of organoids and organs-on-chip as biomimetic tools. Semin Cell Dev Biol 2023; 144:3-10. [PMID: 36192310 DOI: 10.1016/j.semcdb.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022]
Abstract
Organoid development and organ-on-a-chip are technologies based on differentiating stem cells, forming 3D multicellular structures resembling organs and tissues in vivo. Hence, both can be strategically used for disease modeling, drug screening, and host-pathogen studies. In this context, this review highlights the significant advancements in the area, providing technical approaches to organoids and organ-on-a-chip that best imitate in vivo physiology.
Collapse
Affiliation(s)
- Anderson K Santos
- Department of Pediatrics, Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Sérgio Scalzo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Lucas F Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Daniel M Filho
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexander Birbrair
- Departmento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Henning Ulrich
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinópolis, Brazil.
| |
Collapse
|
7
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
8
|
Subramaniyan B, Gurung S, Bodas M, Moore AR, Larabee JL, Reuter D, Georgescu C, Wren JD, Myers DA, Papin JF, Walters MS. The Isolation and In Vitro Differentiation of Primary Fetal Baboon Tracheal Epithelial Cells for the Study of SARS-CoV-2 Host-Virus Interactions. Viruses 2023; 15:v15040862. [PMID: 37112842 PMCID: PMC10146425 DOI: 10.3390/v15040862] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/13/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
The mucociliary airway epithelium lines the human airways and is the primary site of host-environmental interactions in the lung. Following virus infection, airway epithelial cells initiate an innate immune response to suppress virus replication. Therefore, defining the virus-host interactions of the mucociliary airway epithelium is critical for understanding the mechanisms that regulate virus infection, including Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Non-human primates (NHP) are closely related to humans and provide a model to study human disease. However, ethical considerations and high costs can restrict the use of in vivo NHP models. Therefore, there is a need to develop in vitro NHP models of human respiratory virus infection that would allow for rapidly characterizing virus tropism and the suitability of specific NHP species to model human infection. Using the olive baboon (Papio anubis), we have developed methodologies for the isolation, in vitro expansion, cryopreservation, and mucociliary differentiation of primary fetal baboon tracheal epithelial cells (FBTECs). Furthermore, we demonstrate that in vitro differentiated FBTECs are permissive to SARS-CoV-2 infection and produce a potent host innate-immune response. In summary, we have developed an in vitro NHP model that provides a platform for the study of SARS-CoV-2 infection and other human respiratory viruses.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.G.); (D.A.M.)
| | - Manish Bodas
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Andrew R. Moore
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Jason L. Larabee
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Darlene Reuter
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (D.R.); (J.F.P.)
| | - Constantin Georgescu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (C.G.); (J.D.W.)
| | - Jonathan D. Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (C.G.); (J.D.W.)
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.G.); (D.A.M.)
| | - James F. Papin
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (D.R.); (J.F.P.)
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Matthew S. Walters
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
- Correspondence:
| |
Collapse
|
9
|
Lee JE, Jeong SY, Li Z, Kim HY, Kim HW, Yoo MJ, Jang HJ, Kim DK, Cho N, Yoo HM, Kim KH. Development of a screening platform to discover natural products active against SARS-CoV-2 infection using lung organoid models. Biomater Res 2023; 27:18. [PMID: 36855173 PMCID: PMC9974403 DOI: 10.1186/s40824-023-00357-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Natural products can serve as one of the alternatives, exhibiting high potential for the treatment and prevention of COVID-19, caused by SARS-CoV-2. Herein, we report a screening platform to test the antiviral efficacy of a natural product library against SARS-CoV-2 and verify their activity using lung organoids. METHODS Since SARS-CoV-2 is classified as a risk group 3 pathogen, the drug screening assay must be performed in a biosafety level 3 (BSL-3) laboratory. To circumvent this limitation, pseudotyped viruses (PVs) have been developed as replacements for the live SARS-CoV-2. We developed PVs containing spikes from Delta and Omicron variants of SARS-CoV-2 and improved the infection in an angiotensin-converting enzyme 2 (ACE2)-dependent manner. Human induced pluripotent stem cells (hiPSCs) derived lung organoids were generated to test the SARS-CoV-2 therapeutic efficacy of natural products. RESULTS Flavonoids from our natural product library had strong antiviral activity against the Delta- or Omicron-spike-containing PVs without affecting cell viability. We aimed to develop strategies to discover the dual function of either inhibiting infection at the beginning of the infection cycle or reducing spike stability following SARS-CoV-2 infection. When lung cells are already infected with the virus, the active flavonoids induced the degradation of the spike protein and exerted anti-inflammatory effects. Further experiments confirmed that the active flavonoids had strong antiviral activity in lung organoid models. CONCLUSION This screening platform will open new paths by providing a promising standard system for discovering novel drug leads against SARS-CoV-2 and help develop promising candidates for clinical investigation as potential therapeutics for COVID-19.
Collapse
Affiliation(s)
- Joo-Eun Lee
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Se Yun Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Zijun Li
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | - Hyun-Yi Kim
- NGeneS Inc., Ansan, 15495, Republic of Korea
| | - Hyun-Woo Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea
| | - Min Jeong Yoo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee Joo Jang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea.
- Department of Precision Measurement, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
10
|
Carius P, Jungmann A, Bechtel M, Grißmer A, Boese A, Gasparoni G, Salhab A, Seipelt R, Urbschat K, Richter C, Meier C, Bojkova D, Cinatl J, Walter J, Schneider‐Daum N, Lehr C. A Monoclonal Human Alveolar Epithelial Cell Line ("Arlo") with Pronounced Barrier Function for Studying Drug Permeability and Viral Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207301. [PMID: 36748276 PMCID: PMC10015904 DOI: 10.1002/advs.202207301] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Indexed: 06/18/2023]
Abstract
In the development of orally inhaled drug products preclinical animal models regularly fail to predict pharmacological as well as toxicological responses in humans. Models based on human cells and tissues are potential alternatives to animal experimentation allowing for the isolation of essential processes of human biology and making them accessible in vitro. Here, the generation of a novel monoclonal cell line "Arlo," derived from the polyclonal human alveolar epithelium lentivirus immortalized cell line hAELVi via single-cell printing, and its characterization as a model for the human alveolar epithelium as well as a building block for future complex in vitro models is described. "Arlo" is systematically compared in vitro to primary human alveolar epithelial cells (hAEpCs) as well as to the polyclonal hAELVi cell line. "Arlo" cells show enhanced barrier properties with high transepithelial electrical resistance (TEER) of ≈3000 Ω cm2 and a potential difference (PD) of ≈30 mV under air-liquid interface (ALI) conditions, that can be modulated. The cells grow in a polarized monolayer and express genes relevant to barrier integrity as well as homeostasis as is observed in hAEpCs. Successful productive infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a proof-of-principle study offers an additional, attractive application of "Arlo" beyond biopharmaceutical experimentation.
Collapse
Affiliation(s)
- Patrick Carius
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| | - Annemarie Jungmann
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Marco Bechtel
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Alexander Grißmer
- Department of Anatomy and Cellular BiologySaarland UniversityKirrberger StraßeBuilding 6166421Homburg SaarGermany
| | - Annette Boese
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
| | - Gilles Gasparoni
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Abdulrahman Salhab
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Ralf Seipelt
- Section of Thoracic Surgery of the Saar Lung CenterSHG Clinics VölklingenRichardstraße 5‐966333VölklingenGermany
| | - Klaus Urbschat
- Section of Thoracic Surgery of the Saar Lung CenterSHG Clinics VölklingenRichardstraße 5‐966333VölklingenGermany
| | - Clémentine Richter
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| | - Carola Meier
- Department of Anatomy and Cellular BiologySaarland UniversityKirrberger StraßeBuilding 6166421Homburg SaarGermany
| | - Denisa Bojkova
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Jindrich Cinatl
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Jörn Walter
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Nicole Schneider‐Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
| | - Claus‐Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| |
Collapse
|
11
|
Host Response of Syrian Hamster to SARS-CoV-2 Infection including Differences with Humans and between Sexes. Viruses 2023; 15:v15020428. [PMID: 36851642 PMCID: PMC9960357 DOI: 10.3390/v15020428] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the importance of having proper tools and models to study the pathophysiology of emerging infectious diseases to test therapeutic protocols, assess changes in viral phenotypes, and evaluate the effects of viral evolution. This study provided a comprehensive characterization of the Syrian hamster (Mesocricetus auratus) as an animal model for SARS-CoV-2 infection using different approaches (description of clinical signs, viral load, receptor profiling, and host immune response) and targeting four different organs (lungs, intestine, brain, and PBMCs). Our data showed that both male and female hamsters were susceptible to the infection and developed a disease similar to the one observed in patients with COVID-19 that included moderate to severe pulmonary lesions, inflammation, and recruitment of the immune system in the lungs and at the systemic level. However, all animals recovered within 14 days without developing the severe pathology seen in humans, and none of them died. We found faint evidence for intestinal and neurological tropism associated with the absence of lesions and a minimal host response in intestines and brains, which highlighted another crucial difference with the multiorgan impairment of severe COVID-19. When comparing male and female hamsters, we observed that males sustained higher viral RNA shedding and replication in the lungs, suffered from more severe symptoms and histopathological lesions, and triggered higher pulmonary inflammation. Overall, these data confirmed the Syrian hamster as a suitable model for mild to moderate COVID-19 and reflected sex-related differences in the response against the virus observed in humans.
Collapse
|
12
|
Yang W, Li Y, Shi F, Liu H. Human lung organoid: Models for respiratory biology and diseases. Dev Biol 2023; 494:26-34. [PMID: 36470449 DOI: 10.1016/j.ydbio.2022.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
The human respiratory system, consisting of the airway and alveoli, is one of the most complex organs directly interfaced with the external environment. The diverse epithelial cells lining the surface are usually the first cell barrier that comes into contact with pathogens that could lead to deadly pulmonary disease. There is an urgent need to understand the mechanisms of self-renewal and protection of these epithelial cells against harmful pathogens, such as SARS-CoV-2. Traditional models, including cell lines and mouse models, have extremely limited native phenotypic features. Therefore, in recent years, to mimic the complexity of the lung, airway and alveoli organoid technology has been developed and widely applied. TGF-β/BMP/SMAD, FGF and Wnt/β-catenin signaling have been proven to play a key role in lung organoid expansion and differentiation. Thus, we summarize the current novel lung organoid culture strategies and discuss their application for understanding the lung biological features and pathophysiology of pulmonary diseases, especially COVID-19. Lung organoids provide an excellent in vitro model and research platform.
Collapse
Affiliation(s)
- Wenhao Yang
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology Sichuan University, Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yingna Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology Sichuan University, Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fang Shi
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology Sichuan University, Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology Sichuan University, Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Hughes T, Dijkstra KK, Rawlins EL, Hynds RE. Open questions in human lung organoid research. Front Pharmacol 2023; 13:1083017. [PMID: 36712670 PMCID: PMC9880211 DOI: 10.3389/fphar.2022.1083017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Organoids have become a prominent model system in pulmonary research. The ability to establish organoid cultures directly from patient tissue has expanded the repertoire of physiologically relevant preclinical model systems. In addition to their derivation from adult lung stem/progenitor cells, lung organoids can be derived from fetal tissue or induced pluripotent stem cells to fill a critical gap in modelling pulmonary development in vitro. Recent years have seen important progress in the characterisation and refinement of organoid culture systems. Here, we address several open questions in the field, including how closely organoids recapitulate the tissue of origin, how well organoids recapitulate patient cohorts, and how well organoids capture diversity within a patient. We advocate deeper characterisation of models using single cell technologies, generation of more diverse organoid biobanks and further standardisation of culture media.
Collapse
Affiliation(s)
- Tessa Hughes
- Wellcome Trust/CRUK Gurdon Institute and Department Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Krijn K. Dijkstra
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Emma L. Rawlins
- Wellcome Trust/CRUK Gurdon Institute and Department Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Robert E. Hynds
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, Great Ormond Street UCL Institute of Child Health, University College London, London, United Kingdom
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
14
|
Baumgardt M, Hülsemann M, Löwa A, Fatykhova D, Hoffmann K, Kessler M, Mieth M, Hellwig K, Frey D, Langenhagen A, Voss A, Obermayer B, Wyler E, Dökel S, Gruber AD, Tölch U, Hippenstiel S, Hocke AC, Hönzke K. State-of-the-art analytical methods of viral infections in human lung organoids. PLoS One 2022; 17:e0276115. [PMID: 36538516 PMCID: PMC9767351 DOI: 10.1371/journal.pone.0276115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
Human-based organ models can provide strong predictive value to investigate the tropism, virulence, and replication kinetics of viral pathogens. Currently, such models have received widespread attention in the study of SARS-CoV-2 causing the COVID-19 pandemic. Applicable to a large set of organoid models and viruses, we provide a step-by-step work instruction for the infection of human alveolar-like organoids with SARS-CoV-2 in this protocol collection. We also prepared a detailed description on state-of-the-art methodologies to assess the infection impact and the analysis of relevant host factors in organoids. This protocol collection consists of five different sets of protocols. Set 1 describes the protein extraction from human alveolar-like organoids and the determination of protein expression of angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) and FURIN as exemplary host factors of SARS-CoV-2. Set 2 provides detailed guidance on the extraction of RNA from human alveolar-like organoids and the subsequent qPCR to quantify the expression level of ACE2, TMPRSS2, and FURIN as host factors of SARS-CoV-2 on the mRNA level. Protocol set 3 contains an in-depth explanation on how to infect human alveolar-like organoids with SARS-CoV-2 and how to quantify the viral replication by plaque assay and viral E gene-based RT-qPCR. Set 4 provides a step-by-step protocol for the isolation of single cells from infected human alveolar-like organoids for further processing in single-cell RNA sequencing or flow cytometry. Set 5 presents a detailed protocol on how to perform the fixation of human alveolar-like organoids and guides through all steps of immunohistochemistry and in situ hybridization to visualize SARS-CoV-2 and its host factors. The infection and all subsequent analytical methods have been successfully validated by biological replications with human alveolar-like organoids based on material from different donors.
Collapse
Affiliation(s)
- Morris Baumgardt
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Maren Hülsemann
- Berlin Institute of Health at Charité (BIH), BIH QUEST Center for Responsible Research, Berlin, Germany
| | - Anna Löwa
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Diana Fatykhova
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Karen Hoffmann
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Mirjana Kessler
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Department of Gynecology and Obstetrics, University Hospital, LMU, Munich, Germany
| | - Maren Mieth
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Katharina Hellwig
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Doris Frey
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Alina Langenhagen
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Anne Voss
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Simon Dökel
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D. Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Ulf Tölch
- Berlin Institute of Health at Charité (BIH), BIH QUEST Center for Responsible Research, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Andreas C. Hocke
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Katja Hönzke
- Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
15
|
Mostowy S. Louis Pasteur continues to shape the future of microbiology. Dis Model Mech 2022; 15:dmm050011. [PMID: 36504391 PMCID: PMC10655809 DOI: 10.1242/dmm.050011] [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] [Indexed: 12/14/2022] Open
Abstract
Louis Pasteur made seminal discoveries in microbiology, immunology and vaccinology that transformed clinical science and saved millions of lives. Since the 19th century, our ability to study infectious disease has undergone radical changes due to newly emerging technologies and infection models. In this Editorial, I consider Pasteur's impact on our ability to understand and combat infectious disease in the context of two modern-day pandemics: coronavirus disease 2019 (COVID-19) and antimicrobial resistance (AMR). During the COVID-19 pandemic, we witnessed remarkable ambition to understand severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and to innovate effective vaccines to prevent disease. For the comparatively overlooked pandemic of AMR, we require the same level of urgency to develop alternative approaches to combat antibiotic-resistant bacterial strains that cause millions of deaths annually. Pasteur's statement "chance only favours the mind which is prepared" is a principle that captures 'l'esprit Pasteur'. This principle should continue to guide modern-day research on infectious disease, and for this we need to support the development of predictive disease models and cutting-edge mechanistic research that prepare us for discovery and therapeutic impact.
Collapse
Affiliation(s)
- Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| |
Collapse
|
16
|
Baptista LS, Porrini C, Kronemberger GS, Kelly DJ, Perrault CM. 3D organ-on-a-chip: The convergence of microphysiological systems and organoids. Front Cell Dev Biol 2022; 10:1043117. [PMID: 36478741 PMCID: PMC9720174 DOI: 10.3389/fcell.2022.1043117] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/07/2022] [Indexed: 07/29/2023] Open
Abstract
Medicine today faces the combined challenge of an increasing number of untreatable diseases and fewer drugs reaching the clinic. While pharmaceutical companies have increased the number of drugs in early development and entering phase I of clinical trials, fewer actually successfully pass phase III and launch into the market. In fact, only 1 out of every 9 drugs entering phase I will launch. In vitro preclinical tests are used to predict earlier and better the potential of new drugs and thus avoid expensive clinical trial phases. The most recent developments favor 3D cell culture and human stem cell biology. These 3D humanized models known as organoids better mimic the 3D tissue architecture and physiological cell behavior of healthy and disease models, but face critical issues in production such as small-scale batches, greater costs (when compared to monolayer cultures) and reproducibility. To become the gold standard and most relevant biological model for drug discovery and development, organoid technology needs to integrate biological culture processes with advanced microtechnologies, such as microphysiological systems based on microfluidics technology. Microphysiological systems, known as organ-on-a-chip, mimic physiological conditions better than conventional cell culture models since they can emulate perfusion, mechanical and other parameters crucial for tissue and organ physiology. In addition, they reduce labor cost and human error by supporting automated operation and reduce reagent use in miniaturized culture systems. There is thus a clear advantage in combining organoid culture with microsystems for drug development. The main objective of this review is to address the recent advances in organoids and microphysiological systems highlighting crucial technologies for reaching a synergistic strategy, including bioprinting.
Collapse
Affiliation(s)
- Leandra S. Baptista
- Eden Tech, Paris, France
- Universidade Federal do Rio de Janeiro, Campus UFRJ Duque de Caxias Prof Geraldo Cidade, Rio de Janeiro, Brazil
| | | | - Gabriela S. Kronemberger
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | |
Collapse
|
17
|
Pires De Souza GA, Le Bideau M, Boschi C, Wurtz N, Colson P, Aherfi S, Devaux C, La Scola B. Choosing a cellular model to study SARS-CoV-2. Front Cell Infect Microbiol 2022; 12:1003608. [PMID: 36339347 PMCID: PMC9634005 DOI: 10.3389/fcimb.2022.1003608] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/03/2022] [Indexed: 08/04/2023] Open
Abstract
As new pathogens emerge, new challenges must be faced. This is no different in infectious disease research, where identifying the best tools available in laboratories to conduct an investigation can, at least initially, be particularly complicated. However, in the context of an emerging virus, such as SARS-CoV-2, which was recently detected in China and has become a global threat to healthcare systems, developing models of infection and pathogenesis is urgently required. Cell-based approaches are crucial to understanding coronavirus infection biology, growth kinetics, and tropism. Usually, laboratory cell lines are the first line in experimental models to study viral pathogenicity and perform assays aimed at screening antiviral compounds which are efficient at blocking the replication of emerging viruses, saving time and resources, reducing the use of experimental animals. However, determining the ideal cell type can be challenging, especially when several researchers have to adapt their studies to specific requirements. This review strives to guide scientists who are venturing into studying SARS-CoV-2 and help them choose the right cellular models. It revisits basic concepts of virology and presents the currently available in vitro models, their advantages and disadvantages, and the known consequences of each choice.
Collapse
Affiliation(s)
- Gabriel Augusto Pires De Souza
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Marion Le Bideau
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Céline Boschi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Nathalie Wurtz
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Sarah Aherfi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Christian Devaux
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Department of Biological Sciences (INSB), Centre National de la Recherche Scientifique, Marseille, France
| | - Bernard La Scola
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
18
|
Simoneau CR, Chen PY, Xing GK, Khalid MM, Meyers NL, Hayashi JM, Taha TY, Leon KE, Ashuach T, Fontaine KA, Rodriguez L, Joehnk B, Walcott K, Vasudevan S, Fang X, Maishan M, Schultz S, Roose J, Matthay MA, Sil A, Arjomandi M, Yosef N, Ott M. NF-κB inhibitor alpha has a cross-variant role during SARS-CoV-2 infection in ACE2-overexpressing human airway organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.02.502100. [PMID: 35982664 PMCID: PMC9387123 DOI: 10.1101/2022.08.02.502100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
As SARS-CoV-2 continues to spread worldwide, tractable primary airway cell models that accurately recapitulate the cell-intrinsic response to arising viral variants are needed. Here we describe an adult stem cell-derived human airway organoid model overexpressing the ACE2 receptor that supports robust viral replication while maintaining 3D architecture and cellular diversity of the airway epithelium. ACE2-OE organoids were infected with SARS-CoV-2 variants and subjected to single-cell RNA-sequencing. NF-κB inhibitor alpha was consistently upregulated in infected epithelial cells, and its mRNA expression positively correlated with infection levels. Confocal microscopy showed more IκBα expression in infected than bystander cells, but found concurrent nuclear translocation of NF-κB that IκBα usually prevents. Overexpressing a nondegradable IκBα mutant reduced NF-κB translocation and increased viral infection. These data demonstrate the functionality of ACE2-OE organoids in SARS-CoV-2 research and identify an incomplete NF-κB feedback loop as a rheostat of viral infection that may promote inflammation and severe disease.
Collapse
Affiliation(s)
- Camille R. Simoneau
- Gladstone Institute of Virology, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Pei-Yi Chen
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Galen K. Xing
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley CA, USA
| | - Mir M. Khalid
- Gladstone Institute of Virology, San Francisco, CA, USA
| | | | | | - Taha Y. Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Kristoffer E. Leon
- Gladstone Institute of Virology, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | - Tal Ashuach
- Center for Computational Biology, University of California, Berkeley, Berkeley CA, USA
| | | | - Lauren Rodriguez
- ImmunoX CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Bastian Joehnk
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Keith Walcott
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | | | - Xiaohui Fang
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Mazharul Maishan
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Shawn Schultz
- Center for Computational Biology, University of California, Berkeley, Berkeley CA, USA
| | - Jeroen Roose
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Michael A. Matthay
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Mehrdad Arjomandi
- Medical Service, San Francisco VA Healthcare System, San Francisco, CA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley CA, USA
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Tobin DM. Modelling infectious disease to support human health. Dis Model Mech 2022; 15:276457. [PMID: 36037003 PMCID: PMC9459390 DOI: 10.1242/dmm.049824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During the current COVID-19 pandemic, there has been renewed scientific and public focus on understanding the pathogenesis of infectious diseases and investigating vaccines and therapies to combat them. In addition to the tragic toll of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we also recognize increased threats from antibiotic-resistant bacterial strains, the effects of climate change on the prevalence and spread of human pathogens, and the recalcitrance of other infectious diseases – including tuberculosis, malaria, human immunodeficiency virus (HIV) and fungal infections – that continue to cause millions of deaths annually. Large amounts of funding have rightly been redirected toward vaccine development and clinical trials for COVID-19, but we must continue to pursue fundamental and translational research on other pathogens and host immunity. Now more than ever, we need to support the next generation of researchers to develop and utilize models of infectious disease that serve as engines of discovery, innovation and therapy. Summary: This Editorial considers how knowledge from animal and other models of infectious disease can impact our understanding of human biology and potential therapies, focusing largely on zebrafish. It also highlights ways in which DMM is supporting these areas.
Collapse
Affiliation(s)
- David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
20
|
Corsini NS, Knoblich JA. Human organoids: New strategies and methods for analyzing human development and disease. Cell 2022; 185:2756-2769. [PMID: 35868278 DOI: 10.1016/j.cell.2022.06.051] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023]
Abstract
For decades, insight into fundamental principles of human biology and disease has been obtained primarily by experiments in animal models. While this has allowed researchers to understand many human biological processes in great detail, some developmental and disease mechanisms have proven difficult to study due to inherent species differences. The advent of organoid technology more than 10 years ago has established laboratory-grown organ tissues as an additional model system to recapitulate human-specific aspects of biology. The use of human 3D organoids, as well as other advances in single-cell technologies, has revealed unprecedented insights into human biology and disease mechanisms, especially those that distinguish humans from other species. This review highlights novel advances in organoid biology with a focus on how organoid technology has generated a better understanding of human-specific processes in development and disease.
Collapse
Affiliation(s)
- Nina S Corsini
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
| | - Juergen A Knoblich
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria; Medical University of Vienna, Department of Neurology, Vienna, Austria.
| |
Collapse
|
21
|
Chia SPS, Kong SLY, Pang JKS, Soh BS. 3D Human Organoids: The Next "Viral" Model for the Molecular Basis of Infectious Diseases. Biomedicines 2022; 10:1541. [PMID: 35884846 PMCID: PMC9312734 DOI: 10.3390/biomedicines10071541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023] Open
Abstract
The COVID-19 pandemic has driven the scientific community to adopt an efficient and reliable model that could keep up with the infectious disease arms race. Coinciding with the pandemic, three dimensional (3D) human organoids technology has also gained traction in the field of infectious disease. An in vitro construct that can closely resemble the in vivo organ, organoid technology could bridge the gap between the traditional two-dimensional (2D) cell culture and animal models. By harnessing the multi-lineage characteristic of the organoid that allows for the recapitulation of the organotypic structure and functions, 3D human organoids have emerged as an essential tool in the field of infectious disease research. In this review, we will be providing a comparison between conventional systems and organoid models. We will also be highlighting how organoids played a role in modelling common infectious diseases and molecular mechanisms behind the pathogenesis of causative agents. Additionally, we present the limitations associated with the current organoid models and innovative strategies that could resolve these shortcomings.
Collapse
Affiliation(s)
- Shirley Pei Shan Chia
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Sharleen Li Ying Kong
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Jeremy Kah Sheng Pang
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| |
Collapse
|
22
|
Advances of Engineered Hydrogel Organoids within the Stem Cell Field: A Systematic Review. Gels 2022; 8:gels8060379. [PMID: 35735722 PMCID: PMC9222364 DOI: 10.3390/gels8060379] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Organoids are novel in vitro cell culture models that enable stem cells (including pluripotent stem cells and adult stem cells) to grow and undergo self-organization within a three-dimensional microenvironment during the process of differentiation into target tissues. Such miniature structures not only recapitulate the histological and genetic characteristics of organs in vivo, but also form tissues with the capacity for self-renewal and further differentiation. Recent advances in biomaterial technology, particularly hydrogels, have provided opportunities to improve organoid cultures; by closely integrating the mechanical and chemical properties of the extracellular matrix microenvironment, with novel synthetic materials and stem cell biology. This systematic review critically examines recent advances in various strategies and techniques utilized for stem-cell-derived organoid culture, with particular emphasis on the application potential of hydrogel technology in organoid culture. We hope this will give a better understanding of organoid cultures for modelling diseases and tissue engineering applications.
Collapse
|
23
|
Nchioua R, Schundner A, Kmiec D, Prelli Bozzo C, Zech F, Koepke L, Graf A, Krebs S, Blum H, Frick M, Sparrer KMJ, Kirchhoff F. SARS-CoV-2 Variants of Concern Hijack IFITM2 for Efficient Replication in Human Lung Cells. J Virol 2022; 96:e0059422. [PMID: 35543509 PMCID: PMC9175628 DOI: 10.1128/jvi.00594-22] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
It has recently been shown that an early SARS-CoV-2 isolate (NL-02-2020) hijacks interferon-induced transmembrane proteins (IFITMs) for efficient replication in human lung cells, cardiomyocytes, and gut organoids. To date, several "variants of concern" (VOCs) showing increased infectivity and resistance to neutralization have emerged and globally replaced the early viral strains. Here, we determined whether the five current SARS-CoV-2 VOCs (Alpha, Beta, Gamma, Delta, and Omicron) maintained the dependency on IFITM proteins for efficient replication. We found that depletion of IFITM2 strongly reduces viral RNA production by all VOCs in the human epithelial lung cancer cell line Calu-3. Silencing of IFITM1 had modest effects, while knockdown of IFITM3 resulted in an intermediate phenotype. Strikingly, depletion of IFITM2 generally reduced infectious virus production by more than 4 orders of magnitude. In addition, an antibody directed against the N terminus of IFITM2 inhibited SARS-CoV-2 VOC replication in induced pluripotent stem cell (iPSC)-derived alveolar epithelial type II cells, thought to represent major viral target cells in the lung. In conclusion, endogenously expressed IFITM proteins (especially IFITM2) are critical cofactors for efficient replication of genuine SARS-CoV-2 VOCs, including the currently dominant Omicron variant. IMPORTANCE Recent data indicate that SARS-CoV-2 requires endogenously expressed IFITM proteins for efficient infection. However, the results were obtained with an early SARS-CoV-2 isolate. Thus, it remained to be determined whether IFITMs are also important cofactors for infection of emerging SARS-CoV-2 VOCs that outcompeted the original strains in the meantime. This includes the Omicron VOC, which currently dominates the pandemic. Here, we show that depletion of endogenous IFITM2 expression almost entirely prevents productive infection of Alpha, Beta, Gamma, Delta, and Omicron SARS-CoV-2 VOCs in human lung cells. In addition, an antibody targeting the N terminus of IFITM2 inhibited SARS-CoV-2 VOC replication in iPSC-derived alveolar epithelial type II cells. Our results show that SARS-CoV-2 VOCs, including the currently dominant Omicron variant, are strongly dependent on IFITM2 for efficient replication, suggesting a key proviral role of IFITMs in viral transmission and pathogenicity.
Collapse
Affiliation(s)
- Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Annika Schundner
- Institute of General Physiology, Ulm University Medical Center, Ulm, Germany
| | - Dorota Kmiec
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University Medical Center, Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
24
|
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a devastating pandemic. Although most people infected with SARS-CoV-2 develop a mild to moderate disease with virus replication restricted mainly to the upper airways, some progress to having a life-threatening pneumonia. In this Review, we explore recent clinical and experimental advances regarding SARS-CoV-2 pathophysiology and discuss potential mechanisms behind SARS-CoV-2-associated acute respiratory distress syndrome (ARDS), specifically focusing on new insights obtained using novel technologies such as single-cell omics, organoid infection models and CRISPR screens. We describe how SARS-CoV-2 may infect the lower respiratory tract and cause alveolar damage as a result of dysfunctional immune responses. We discuss how this may lead to the induction of a 'leaky state' of both the epithelium and the endothelium, promoting inflammation and coagulation, while an influx of immune cells leads to overexuberant inflammatory responses and immunopathology. Finally, we highlight how these findings may aid the development of new therapeutic interventions against COVID-19.
Collapse
|
25
|
Goldstein ME, Scull MA. Modeling Innate Antiviral Immunity in Physiological Context. J Mol Biol 2022; 434:167374. [PMID: 34863779 PMCID: PMC8940657 DOI: 10.1016/j.jmb.2021.167374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022]
Abstract
An effective innate antiviral response is critical for the mitigation of severe disease and host survival following infection. In vivo, the innate antiviral response is triggered by cells that detect the invading pathogen and then communicate through autocrine and paracrine signaling to stimulate the expression of genes that inhibit viral replication, curtail cell proliferation, or modulate the immune response. In other words, the innate antiviral response is complex and dynamic. Notably, in the laboratory, culturing viruses and assaying viral life cycles frequently utilizes cells that are derived from tissues other than those that support viral replication during natural infection, while the study of viral pathogenesis often employs animal models. In recapitulating the human antiviral response, it is important to consider that variation in the expression and function of innate immune sensors and antiviral effectors exists across species, cell types, and cell differentiation states, as well as when cells are placed in different contexts. Thus, to gain novel insight into the dynamics of the host response and how specific sensors and effectors impact infection kinetics by a particular virus, the model system must be selected carefully. In this review, we briefly introduce key signaling pathways involved in the innate antiviral response and highlight how these differ between systems. We then review the application of tissue-engineered or 3D models for studying the antiviral response, and suggest how these in vitro culture systems could be further utilized to assay physiologically-relevant host responses and reveal novel insight into virus-host interactions.
Collapse
Affiliation(s)
- Monty E Goldstein
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA
| | - Margaret A Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
26
|
Li YR, Dunn ZS, Garcia G, Carmona C, Zhou Y, Lee D, Yu J, Huang J, Kim JT, Arumugaswami V, Wang P, Yang L. Development of off-the-shelf hematopoietic stem cell-engineered invariant natural killer T cells for COVID-19 therapeutic intervention. Stem Cell Res Ther 2022; 13:112. [PMID: 35313965 PMCID: PMC8935266 DOI: 10.1186/s13287-022-02787-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND New COVID-19 treatments are desperately needed as case numbers continue to rise and emergent strains threaten vaccine efficacy. Cell therapy has revolutionized cancer treatment and holds much promise in combatting infectious disease, including COVID-19. Invariant natural killer T (iNKT) cells are a rare subset of T cells with potent antiviral and immunoregulatory functions and an excellent safety profile. Current iNKT cell strategies are hindered by the extremely low presence of iNKT cells, and we have developed a platform to overcome this critical limitation. METHODS We produced allogeneic HSC-engineered iNKT (AlloHSC-iNKT) cells through TCR engineering of human cord blood CD34+ hematopoietic stem cells (HSCs) and differentiation of these HSCs into iNKT cells in an Ex Vivo HSC-Derived iNKT Cell Culture. We then established in vitro SARS-CoV-2 infection assays to assess AlloHSC-iNKT cell antiviral and anti-hyperinflammation functions. Lastly, using in vitro and in vivo preclinical models, we evaluated AlloHSC-iNKT cell safety and immunogenicity for off-the-shelf application. RESULTS We reliably generated AlloHSC-iNKT cells at high-yield and of high-purity; these resulting cells closely resembled endogenous human iNKT cells in phenotypes and functionalities. In cell culture, AlloHSC-iNKT cells directly killed SARS-CoV-2 infected cells and also selectively eliminated SARS-CoV-2 infection-stimulated inflammatory monocytes. In an in vitro mixed lymphocyte reaction (MLR) assay and an NSG mouse xenograft model, AlloHSC-iNKT cells were resistant to T cell-mediated alloreaction and did not cause GvHD. CONCLUSIONS Here, we report a method to robustly produce therapeutic levels of AlloHSC-iNKT cells. Preclinical studies showed that these AlloHSC-iNKT cells closely resembled endogenous human iNKT cells, could reduce SARS-CoV-2 virus infection load and mitigate virus infection-induced hyperinflammation, and meanwhile were free of GvHD-risk and resistant to T cell-mediated allorejection. These results support the development of AlloHSC-iNKT cells as a promising off-the-shelf cell product for treating COVID-19; such a cell product has the potential to target the new emerging SARS-CoV-2 variants as well as the future new emerging viruses.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zachary Spencer Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, Los Angeles, CA, 90089, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Camille Carmona
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jie Huang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jocelyn T Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, Los Angeles, CA, 90089, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
27
|
Ekanger CT, Zhou F, Bohan D, Lotsberg ML, Ramnefjell M, Hoareau L, Røsland GV, Lu N, Aanerud M, Gärtner F, Salminen PR, Bentsen M, Halvorsen T, Ræder H, Akslen LA, Langeland N, Cox R, Maury W, Stuhr LEB, Lorens JB, Engelsen AST. Human Organotypic Airway and Lung Organoid Cells of Bronchiolar and Alveolar Differentiation Are Permissive to Infection by Influenza and SARS-CoV-2 Respiratory Virus. Front Cell Infect Microbiol 2022; 12:841447. [PMID: 35360113 PMCID: PMC8964279 DOI: 10.3389/fcimb.2022.841447] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has led to the initiation of unprecedented research efforts to understand the pathogenesis mediated by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). More knowledge is needed regarding the cell type-specific cytopathology and its impact on cellular tropism. Furthermore, the impact of novel SARS-CoV-2 mutations on cellular tropism, alternative routes of entry, the impact of co-infections, and virus replication kinetics along the respiratory tract remains to be explored in improved models. Most applied virology models are not well suited to address the remaining questions, as they do not recapitulate the histoarchitecture and cellular composition of human respiratory tissues. The overall aim of this work was to establish from single biopsy specimens, a human adult stem cell-derived organoid model representing the upper respiratory airways and lungs and explore the applicability of this model to study respiratory virus infection. First, we characterized the organoid model with respect to growth pattern and histoarchitecture, cellular composition, and functional characteristics. Next, in situ expression of viral entry receptors, including influenza virus-relevant sialic acids and SARS-CoV-2 entry receptor ACE2 and TMPRSS2, were confirmed in organoids of bronchiolar and alveolar differentiation. We further showed successful infection by pseudotype influenza A H7N1 and H5N1 virus, and the ability of the model to support viral replication of influenza A H7N1 virus. Finally, successful infection and replication of a clinical isolate of SARS-CoV-2 were confirmed in the organoids by TCID50 assay and immunostaining to detect intracellular SARS-CoV-2 specific nucleocapsid and dsRNA. The prominent syncytia formation in organoid tissues following SARS-CoV-2 infection mimics the findings from infected human tissues in situ. We conclude that the human organotypic model described here may be particularly useful for virology studies to evaluate regional differences in the host response to infection. The model contains the various cell types along the respiratory tract, expresses respiratory virus entry factors, and supports successful infection and replication of influenza virus and SARS-CoV-2. Thus, the model may serve as a relevant and reliable tool in virology and aid in pandemic preparedness, and efficient evaluation of antiviral strategies.
Collapse
Affiliation(s)
- Camilla Tvedt Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Fan Zhou
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Maria Lie Lotsberg
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Maria Ramnefjell
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Laurence Hoareau
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Gro Vatne Røsland
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Ning Lu
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Marianne Aanerud
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Fabian Gärtner
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Pirjo Riitta Salminen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Section of Cardiothoracic Surgery, Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Mariann Bentsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Thomas Halvorsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Helge Ræder
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Nina Langeland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Cox
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | | | - James B. Lorens
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- *Correspondence: Agnete S. T. Engelsen,
| |
Collapse
|
28
|
Teo A, Chua CLL, Chan LLY. Airway models in a pandemic: Suitability of models in modeling SARS-CoV-2. PLoS Pathog 2022; 18:e1010432. [PMID: 35349597 PMCID: PMC8963546 DOI: 10.1371/journal.ppat.1010432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Louisa L. Y. Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
29
|
Tran BM, Grimley SL, McAuley JL, Hachani A, Earnest L, Wong SL, Caly L, Druce J, Purcell DFJ, Jackson DC, Catton M, Nowell CJ, Leonie L, Deliyannis G, Waters SA, Torresi J, Vincan E. Air-Liquid-Interface Differentiated Human Nose Epithelium: A Robust Primary Tissue Culture Model of SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:835. [PMID: 35055020 PMCID: PMC8776210 DOI: 10.3390/ijms23020835] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.
Collapse
Affiliation(s)
- Bang M. Tran
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia;
| | - Samantha L. Grimley
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Julie L. McAuley
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Linda Earnest
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Sharon L. Wong
- Molecular and Integrative Cystic Fibrosis Research Centre, School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; (S.L.W.); (S.A.W.)
| | - Leon Caly
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Julian Druce
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Damian F. J. Purcell
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - David C. Jackson
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Cameron J. Nowell
- Imaging, FACS and Analysis Core, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia;
| | - Laura Leonie
- Melbourne Histology Platform, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Georgia Deliyannis
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Shafagh A. Waters
- Molecular and Integrative Cystic Fibrosis Research Centre, School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; (S.L.W.); (S.A.W.)
- School of Women’s and Children’s Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children’s Hospital, Randwick, NSW 2031, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Elizabeth Vincan
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia;
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
30
|
Söderlund-Venermo M, Varma A, Guo D, Gladue DP, Poole E, Pujol FH, Pappu H, Romalde JL, Kramer L, Baz M, Venter M, Moore MD, Nevels MM, Ezzikouri S, Vakharia VN, Wilson WC, Malik YS, Shi Z, Abdel-Moneim AS. World Society for Virology first international conference: Tackling global virus epidemics. Virology 2022; 566:114-121. [PMID: 34902730 PMCID: PMC8646940 DOI: 10.1016/j.virol.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/18/2021] [Accepted: 11/26/2021] [Indexed: 01/04/2023]
Abstract
This communication summarizes the presentations given at the 1st international conference of the World Society for Virology (WSV) held virtually during 16-18 June 2021, under the theme of tackling global viral epidemics. The purpose of this biennial meeting is to foster international collaborations and address important viral epidemics in different hosts. The first day included two sessions exclusively on SARS-CoV-2 and COVID-19. The other two days included one plenary and three parallel sessions each. Last not least, 16 sessions covered 140 on-demand submitted talks. In total, 270 scientists from 49 countries attended the meeting, including 40 invited keynote speakers.
Collapse
Affiliation(s)
| | - Anupam Varma
- Advanced Centre for Plant Virology Indian Agricultural Research Institute, New Delhi, India
| | - Deyin Guo
- Center for Infection and Immunity Study, School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Emma Poole
- Department of Medicine, University of Cambridge, Level 5, Addenbrooke's Hospital, Hills Road, Cambridge, UK
| | - Flor H. Pujol
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - Hanu Pappu
- Department of Plant Pathology, Washington State University, Pullman, WA, USA
| | - Jesús L. Romalde
- Department of Microbiology and Parasitology, CRETUS & CIBUS-Faculty of Biology, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Laura Kramer
- Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Mariana Baz
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute, Australia and Research Center in Infectious Diseases of the CHU of Québec and Université Laval, Melbourne, Victoria, Québec City, Quebec, Canada
| | - Marietjie Venter
- Zoonotic Arbo- and Respiratory Virus Research Program, Centre for Viral Zoonosis, Department of Medical Virology, University of Pretoria, Pretoria, South Africa
| | - Matthew D. Moore
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Michael M. Nevels
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Vikram N. Vakharia
- Institute of Marine & Environmental Technology, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - William C. Wilson
- Foreign Arthropod Borne Animal Diseases Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS, USA
| | - Yashpal S. Malik
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Zhengli Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ahmed S. Abdel-Moneim
- Microbiology Department, Virology Division, College of Medicine, Taif University, Al-Taif, Saudi Arabia, Virology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt,Corresponding author
| |
Collapse
|
31
|
Matusali G, Trionfetti F, Bordoni V, Nardacci R, Falasca L, Colombo D, Terri M, Montaldo C, Castilletti C, Mariotti D, Del Nonno F, Capobianchi MR, Agrati C, Tripodi M, Strippoli R. Pleural Mesothelial Cells Modulate the Inflammatory/Profibrotic Response During SARS-CoV-2 Infection. Front Mol Biosci 2021; 8:752616. [PMID: 34901152 PMCID: PMC8662383 DOI: 10.3389/fmolb.2021.752616] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Although lung fibrosis has a major impact in COVID-19 disease, its pathogenesis is incompletely understood. In particular, no direct evidence of pleura implication in COVID-19-related fibrotic damage has been reported so far. In this study, the expression of epithelial cytokeratins and Wilms tumor 1 (WT1), specific markers of mesothelial cells (MCs), was analyzed in COVID-19 and unrelated pleura autoptic samples. SARS-CoV-2 replication was analyzed by RT-PCR and confocal microscopy in MeT5A, a pleura MC line. SARS-CoV-2 receptors were analyzed by RT-PCR and western blot. Inflammatory cytokines from the supernatants of SARS-CoV-2-infected MeT5A cells were analysed by Luminex and ELLA assays. Immunohistochemistry of COVID-19 pleura patients highlighted disruption of pleura monolayer and fibrosis of the sub-mesothelial stroma, with the presence of MCs with fibroblastoid morphology in the sub-mesothelial stroma, but no evidence of direct infection in vivo. Interestingly, we found evidence of ACE2 expression in MCs from pleura of COVID-19 patients. In vitro analysis shown that MeT5A cells expressed ACE2, TMPRSS2, ADAM17 and NRP1, plasma membrane receptors implicated in SARS-CoV-2 cell entry and infectivity. Moreover, MeT5A cells sustained SARS-CoV-2 replication and productive infection. Infected MeT5A cells produced interferons, inflammatory cytokines and metalloproteases. Overall, our data highlight the potential role of pleura MCs as promoters of the fibrotic reaction and regulators of the immune response upon SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Veronica Bordoni
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Roberta Nardacci
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy.,UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Laura Falasca
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Daniele Colombo
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Davide Mariotti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Franca Del Nonno
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Chiara Agrati
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
32
|
Larijani B, Foroughi-Heravani N, Abedi M, Tayanloo-Beik A, Rezaei-Tavirani M, Adibi H, Arjmand B. Recent Advances of COVID-19 Modeling Based on Regenerative Medicine. Front Cell Dev Biol 2021; 9:683619. [PMID: 34760882 PMCID: PMC8573217 DOI: 10.3389/fcell.2021.683619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has caused a pandemic since December 2019 that originated in Wuhan, China. Soon after that, the world health organization declared Coronavirus disease-2019 a global health concern. SARS-CoV-2 is responsible for a lethal respiratory infection as well as the involvement of other organs due to its large tropism spectrum such as neurologic, cardiovascular, endocrine, gastrointestinal, and renal systems. Since the behavior of the virus is not fully understood, a new manifestation of the infection is revealed every day. In order to be able to design more efficient drugs and vaccines to treat the infection, finding out the exact mechanism of pathogenicity would be necessary. Although there have been some big steps toward understanding the relevant process, there are still some deficiencies in this field. Accordingly, regenerative medicine (RM), can offer promising opportunities in discovering the exact mechanisms and specific treatments. For instance, since it is not always possible to catch the pathophysiology mechanisms in human beings, several modeling methods have been introduced in this field that can be studied in three main groups: stem cell-based models, organoids, and animal models. Regarding stem cell-based models, induced pluripotent stem cells are the major study subjects, which are generated by reprogramming the somatic stem cells and then directing them into different adult cell populations to study their behavior toward the infection. In organoid models, different cell lines can be guided to produce a 3D structure including liver, heart, and brain-like platforms. Among animal models, mice are the most common species in this field. However, in order for mice models to be permissive to the virus, angiotensin-converting enzyme 2 receptors, the main receptor involved in the pathogenicity of the virus, should be introduced to the host cells through different methods. Here, the current known mechanism of SARS-CoV-2 infection, different suggested models, the specific response toward different manipulation as well as challenges and shortcomings in each case have been reviewed. Finally, we have tried to provide a quick summary of the present available RM-based models for SARS-CoV-2 infection, as an essential part of developing drugs, for future therapeutic goals.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Najmeh Foroughi-Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Rijsbergen LC, van Dijk LLA, Engel MFM, de Vries RD, de Swart RL. In Vitro Modelling of Respiratory Virus Infections in Human Airway Epithelial Cells - A Systematic Review. Front Immunol 2021; 12:683002. [PMID: 34489934 PMCID: PMC8418200 DOI: 10.3389/fimmu.2021.683002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Respiratory tract infections (RTI) are a major cause of morbidity and mortality in humans. A large number of RTIs is caused by viruses, often resulting in more severe disease in infants, elderly and the immunocompromised. Upon viral infection, most individuals experience common cold-like symptoms associated with an upper RTI. However, in some cases a severe and sometimes life-threatening lower RTI may develop. Reproducible and scalable in vitro culture models that accurately reflect the human respiratory tract are needed to study interactions between respiratory viruses and the host, and to test novel therapeutic interventions. Multiple in vitro respiratory cell culture systems have been described, but the majority of these are based on immortalized cell lines. Although useful for studying certain aspects of viral infections, such monomorphic, unicellular systems fall short in creating an understanding of the processes that occur at an integrated tissue level. Novel in vitro models involving primary human airway epithelial cells and, more recently, human airway organoids, are now in use. In this review, we describe the evolution of in vitro cell culture systems and their characteristics in the context of viral RTIs, starting from advances after immortalized cell cultures to more recently developed organoid systems. Furthermore, we describe how these models are used in studying virus-host interactions, e.g. tropism and receptor studies as well as interactions with the innate immune system. Finally, we provide an outlook for future developments in this field, including co-factors that mimic the microenvironment in the respiratory tract.
Collapse
Affiliation(s)
- Laurine C. Rijsbergen
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Laura L. A. van Dijk
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Maarten F. M. Engel
- Medical Library, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| |
Collapse
|