1
|
Tollefson AE, Cline-Smith A, Spencer JF, Ying B, Reyna DM, Lipka E, James SH, Toth K. Longitudinal Monitoring of the Effects of Anti-Adenoviral Treatment Regimens in a Permissive In Vivo Model. Viruses 2024; 16:1200. [PMID: 39205174 PMCID: PMC11359180 DOI: 10.3390/v16081200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Adenovirus infections of immunocompromised patients can cause life-threatening disseminated disease. While there are presently no drugs specifically approved to treat these infections, there are several compounds that showed efficacy against adenovirus in preclinical studies. For any such compound, low toxicity is an essential requirement. As cumulative drug effects can accentuate pathology, especially in patients with other morbidities, it is important to limit antiviral exposure to what is absolutely necessary. This is achievable by monitoring the virus burden of the patients and administering antivirals to suppress virus replication to a non-pathogenic level. We modeled such a system using Syrian hamsters infected with a replication-competent adenovirus vector, in which luciferase expression is coupled to virus replication. We found that virus replication could be followed in vivo in the same animal by repeated measurement of luciferase expression. To test the utility of an interrupted treatment regimen, we used NPP-669 and valganciclovir, two antiviral compounds with high and moderate anti-adenoviral efficacy, respectively. We found that short-term treatment of adenovirus-infected hamsters at times of peak virus replication can prevent virus-associated pathology. Thus, we believe that this animal model can be used to model different treatment regimens for anti-adenoviral compounds.
Collapse
Affiliation(s)
- Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Anna Cline-Smith
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | | | | | - Scott H James
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
2
|
Langston H, Fortes Francisco A, Doidge C, Roberts CH, Khan AA, Jayawardhana S, Taylor MC, Kelly JM, Lewis MD. Dynamics of Trypanosoma cruzi infection in hamsters and novel association with progressive motor dysfunction. PLoS Negl Trop Dis 2024; 18:e0012278. [PMID: 38905323 PMCID: PMC11221660 DOI: 10.1371/journal.pntd.0012278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/03/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024] Open
Abstract
Chagas disease is a zoonosis caused by the protozoan parasite Trypanosoma cruzi. Clinical outcomes range from long-term asymptomatic carriage to cardiac, digestive, neurological and composite presentations that can be fatal in both acute and chronic stages of the disease. Studies of T. cruzi in animal models, principally mice, have informed our understanding of the biological basis of this variability and its relationship to infection and host response dynamics. Hamsters have higher translational value for many human infectious diseases, but they have not been well developed as models of Chagas disease. We transposed a real-time bioluminescence imaging system for T. cruzi infection from mice into female Syrian hamsters (Mesocricetus auratus). This enabled us to study chronic tissue pathology in the context of spatiotemporal infection dynamics. Acute infections were widely disseminated, whereas chronic infections were almost entirely restricted to the skin and subcutaneous adipose tissue. Neither cardiac nor digestive tract disease were reproducible features of the model. Skeletal muscle had only sporadic parasitism in the chronic phase, but nevertheless displayed significant inflammation and fibrosis, features also seen in mouse models. Whereas mice had normal locomotion, all chronically infected hamsters developed hindlimb muscle hypertonia and a gait dysfunction resembling spastic diplegia. With further development, this model may therefore prove valuable in studies of peripheral nervous system involvement in Chagas disease.
Collapse
Affiliation(s)
- Harry Langston
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ciaran Doidge
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Chrissy H. Roberts
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
3
|
Sanchez-Felipe L, Alpizar YA, Ma J, Coelmont L, Dallmeier K. YF17D-based vaccines - standing on the shoulders of a giant. Eur J Immunol 2024; 54:e2250133. [PMID: 38571392 DOI: 10.1002/eji.202250133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 04/05/2024]
Abstract
Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.
Collapse
Affiliation(s)
- Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Yeranddy A Alpizar
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| |
Collapse
|
4
|
Gebert JT, Scribano F, Engevik KA, Perry JL, Hyser JM. Gastrointestinal organoids in the study of viral infections. Am J Physiol Gastrointest Liver Physiol 2023; 324:G51-G59. [PMID: 36414538 PMCID: PMC9799139 DOI: 10.1152/ajpgi.00152.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Viruses are among the most prevalent enteric pathogens. Although virologists historically relied on cell lines and animal models, human intestinal organoids (HIOs) continue to grow in popularity. HIOs are nontransformed, stem cell-derived, ex vivo cell cultures that maintain the cell type diversity of the intestinal epithelium. They offer higher throughput than standard animal models while more accurately mimicking the native tissue of infection than transformed cell lines. Here, we review recent literature that highlights virological advances facilitated by HIOs. We discuss the variations and limitations of HIOs, how HIOs have allowed for the cultivation of previously uncultivatable viruses, and how they have offered insight into tropism, entry, replication kinetics, and host-pathogen interactions. In each case, we discuss exemplary viruses and archetypal studies. We discuss how the speed and flexibility of HIO-based studies contributed to our knowledge of SARS-CoV-2 and antiviral therapeutics. Finally, we discuss the current limitations of HIOs and future directions to overcome these.
Collapse
Affiliation(s)
- J Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Francesca Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Jacob L Perry
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
- Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
5
|
Xue Y, Yang D, Vogel P, Stabenow J, Zalduondo L, Kong Y, Ravi Y, Sai-Sudhakar CB, Parvathareddy J, Hayes E, Taylor S, Fitzpatrick E, Jonsson CB. Cardiopulmonary Injury in the Syrian Hamster Model of COVID-19. Viruses 2022; 14:v14071403. [PMID: 35891384 PMCID: PMC9316644 DOI: 10.3390/v14071403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/21/2022] [Accepted: 06/25/2022] [Indexed: 02/04/2023] Open
Abstract
The Syrian hamster has proved useful in the evaluation of therapeutics and vaccines for severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). To advance the model for preclinical studies, we conducted serial sacrifice of lungs, large pulmonary vessels, and hearts from male and female Syrian hamsters for days 1–4, and 8 post-infection (dpi) following infection with a high dose of SARS-CoV-2. Evaluation of microscopic lung histopathology scores suggests 4 and 8 dpi as prime indicators in the evaluation of moderate pathology with bronchial hyperplasia, alveolar involvement and bronchiolization being key assessments of lung disease and recovery, respectively. In addition, neutrophil levels, red blood cell count and hematocrit showed significant increases during early infection. We present histological evidence of severe damage to the pulmonary vasculature with extensive leukocyte transmigration and the loss of endothelial cells and tunica media. Our evidence of endothelial and inflammatory cell death in the pulmonary vessels suggests endothelialitis secondary to SARS-CoV-2 epithelial cell infection as a possible determinant of the pathological findings along with the host inflammatory response. Lastly, pathological examination of the heart revealed evidence for intracardiac platelet/fibrin aggregates in male and female hamsters on 8 dpi, which might be indicative of a hypercoagulative state in these animals.
Collapse
Affiliation(s)
- Yi Xue
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.X.); (Y.K.); (E.H.); (E.F.)
| | - Dong Yang
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
| | - Peter Vogel
- Animal Resources Center and Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Jennifer Stabenow
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
| | - Lillian Zalduondo
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
| | - Ying Kong
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.X.); (Y.K.); (E.H.); (E.F.)
| | - Yazhini Ravi
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06085, USA; (Y.R.); (C.B.S.-S.)
| | - Chittoor B. Sai-Sudhakar
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06085, USA; (Y.R.); (C.B.S.-S.)
| | - Jyothi Parvathareddy
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
| | - Ernestine Hayes
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.X.); (Y.K.); (E.H.); (E.F.)
| | - Shannon Taylor
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
| | - Elizabeth Fitzpatrick
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.X.); (Y.K.); (E.H.); (E.F.)
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Colleen B. Jonsson
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.X.); (Y.K.); (E.H.); (E.F.)
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.Y.); (J.S.); (L.Z.); (J.P.); (S.T.)
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence:
| |
Collapse
|
6
|
Eskandarian Boroujeni M, Sekrecka A, Antonczyk A, Hassani S, Sekrecki M, Nowicka H, Lopacinska N, Olya A, Kluzek K, Wesoly J, Bluyssen HAR. Dysregulated Interferon Response and Immune Hyperactivation in Severe COVID-19: Targeting STATs as a Novel Therapeutic Strategy. Front Immunol 2022; 13:888897. [PMID: 35663932 PMCID: PMC9156796 DOI: 10.3389/fimmu.2022.888897] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
A disease outbreak in December 2019, caused by a novel coronavirus SARS-CoV-2, was named COVID-19. SARS-CoV-2 infects cells from the upper and lower respiratory tract system and is transmitted by inhalation or contact with infected droplets. Common clinical symptoms include fatigue, fever, and cough, but also shortness of breath and lung abnormalities. Still, some 5% of SARS-CoV-2 infections progress to severe pneumonia and acute respiratory distress syndrome (ARDS), with pulmonary edema, acute kidney injury, and/or multiple organ failure as important consequences, which can lead to death. The innate immune system recognizes viral RNAs and triggers the expression of interferons (IFN). IFNs activate anti-viral effectors and components of the adaptive immune system by activating members of the STAT and IRF families that induce the expression of IFN-stimulated genes (ISG)s. Among other coronaviruses, such as Middle East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV, common strategies have been identified to antagonize IFN signaling. This typically coincides with hyperactive inflammatory host responses known as the “cytokine storm” that mediate severe lung damage. Likewise, SARS-CoV-2 infection combines a dysregulated IFN response with excessive production of inflammatory cytokines in the lungs. This excessive inflammatory response in the lungs is associated with the local recruitment of immune cells that create a pathogenic inflammatory loop. Together, it causes severe lung pathology, including ARDS, as well as damage to other vulnerable organs, like the heart, spleen, lymph nodes, and kidney, as well as the brain. This can rapidly progress to multiple organ exhaustion and correlates with a poor prognosis in COVID-19 patients. In this review, we focus on the crucial role of different types of IFN that underlies the progression of SARS-CoV-2 infection and leads to immune cell hyper-activation in the lungs, exuberant systemic inflammation, and multiple organ damage. Consequently, to protect from systemic inflammation, it will be critical to interfere with signaling cascades activated by IFNs and other inflammatory cytokines. Targeting members of the STAT family could therefore be proposed as a novel therapeutic strategy in patients with severe COVID-19.
Collapse
Affiliation(s)
- Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Agata Sekrecka
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Aleksandra Antonczyk
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Sanaz Hassani
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Michal Sekrecki
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Hanna Nowicka
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Natalia Lopacinska
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Arta Olya
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Katarzyna Kluzek
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Hans A R Bluyssen
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
7
|
HUANG C, WEI D, LIU Y, SHI G. Clinical and imaging findings of patients diagnosed with adenovirus-positive pneumonia during 2015-2019 in Shanghai, China. Turk J Med Sci 2022; 52:329-337. [PMID: 36161612 PMCID: PMC10381194 DOI: 10.55730/1300-0144.5319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 04/14/2022] [Accepted: 10/30/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND This study was to describe the clinical characteristics, chest CT image findings, and potential role of T cells immunity in adenovirus positive pneumonia. METHODS In this retrospective study, medical records of 53 adult Adv+ patients who were admitted to the Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, from May 2015 to August 2019 were included. The presence of adenovirus and other respiratory viruses was detected using polymerase chain reaction of throat swabs samples. Clinical features and chest computed tomography (CT) findings were compared between patients with Adv+ pneumonia and Adv+ non-pneumonia. RESULTS The top 3 most commonly occurring symptoms in Adv+ pneumonia patients were fever (66.7%), cough (63.3%), and tachypnea (16.7%). Patients with Adv+ pneumonia showed significantly higher rates of cough and fever and longer duration of hospitalization than patients with Adv+ non-pneumonia. In the Adv+ pneumonia group, consolidation (73.3%) was the most common imaging finding on chest CT scan, and the likelihood of involvement of bilateral lobes (60%) was high. Classical conspicuous consolidation with surrounding ground-glass opacity was observed in 5 (16.6%) patients with Adv+ pneumonia. Patients with Adv+ pneumonia showed a higher inhibition of T-cell immunity than did patients with Adv+ non-pneumonia, and counts of CD3+, CD4+, and CD8+ T-cells may predict the presence of pneumonia in Adv+ patients. DISCUSSION With regard to Adv+ pneumonia, the most frequent symptoms were cough and fever, and the most common CT pattern was consolidation; classical CT findings such as consolidation with surrounding ground-glass opacity could also be observed. Furthermore, our data indicated the incidence of abrogated cellular immunity in patients with Adv+ pneumonia.
Collapse
Affiliation(s)
- Chunrong HUANG
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
| | - Dong WEI
- Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of
China
| | - Yahui LIU
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
| | - Guochao SHI
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of
China
| |
Collapse
|
8
|
Bertzbach LD, Ip WH, Dobner T. Animal Models in Human Adenovirus Research. BIOLOGY 2021; 10:biology10121253. [PMID: 34943168 PMCID: PMC8698265 DOI: 10.3390/biology10121253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary Animal models are widely used to study various aspects of human diseases and disorders. Likewise, they are indispensable for preclinical testing of medicals and vaccines. Human adenovirus infections are usually self-limiting, and can cause mild respiratory symptoms with fever, eye infection or gastrointestinal symptoms, but occasional local outbreaks with severe disease courses have been reported. In addition, adenovirus infections pose a serious risk for children and patients with a weakened immune system. Human adenovirus research in animal models to study adenovirus-induced disease and tumor development started in the 1950s. Various animal species have been tested for their susceptibility to human adenovirus infection since then, and some have been shown to mimic key characteristics of the infection in humans, including persistent infection. Furthermore, some rodent species have been found to develop tumors upon human adenovirus infection. Our review summarizes the current knowledge on animal models in human adenovirus research, describing the pros and cons along with important findings and future perspectives. Abstract Human adenovirus (HAdV) infections cause a wide variety of clinical symptoms, ranging from mild upper respiratory tract disease to lethal outcomes, particularly in immunocompromised individuals. To date, neither widely available vaccines nor approved antiadenoviral compounds are available to efficiently deal with HAdV infections. Thus, there is a need to thoroughly understand HAdV-induced disease, and for the development and preclinical evaluation of HAdV therapeutics and/or vaccines, and consequently for suitable standardizable in vitro systems and animal models. Current animal models to study HAdV pathogenesis, persistence, and tumorigenesis include rodents such as Syrian hamsters, mice, and cotton rats, as well as rabbits. In addition, a few recent studies on other species, such as pigs and tree shrews, reported promising data. These models mimic (aspects of) HAdV-induced pathological changes in humans and, although they are relevant, an ideal HAdV animal model has yet to be developed. This review summarizes the available animal models of HAdV infection with comprehensive descriptions of virus-induced pathogenesis in different animal species. We also elaborate on rodent HAdV animal models and how they contributed to insights into adenovirus-induced cell transformation and cancer.
Collapse
|
9
|
Biographical Feature: William S. M. Wold, Ph.D., 1944-2021. J Virol 2021; 95:e0118421. [PMID: 34549981 DOI: 10.1128/jvi.01184-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
10
|
Guerrero-Rodríguez J, Cárdenas-Vargas A, Gutierrez-Silerio G, Sobrevilla-Navarro A, Bastidas-Ramírez B, Hernández-Ortega L, Gurrola-Díaz C, Gasca-Lozano L, Armendáriz-Borunda J, Salazar-Montes A. Delivery of Anti-IFNAR1 shRNA to Hepatic Cells Decreases IFNAR1 Gene Expression and Improves Adenoviral Transduction and Transgene Expression. Mol Biotechnol 2021; 64:413-423. [PMID: 34687024 DOI: 10.1007/s12033-021-00408-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 09/21/2021] [Indexed: 11/26/2022]
Abstract
Chronic liver injury leads to advanced fibrosis, cirrhosis, and hepatocellular carcinoma. Genetical cell treatment related to the use of adenovirus (Ads) has proven to be beneficial and efficient in the recovery of hepatic diseases. Nevertheless, they are highly immunogenic and trigger an immune response where interferons type 1 (IFN-I) play a very important role. Three shRNAs against the Interferon-1 receptor (IFNAR1) were designed and cloned in pENTR/U6 plasmid and amplified in DH5α cells. Huh7 cells were transfected with these plasmids in the presence or absence of 1 × 109 viral particles/ml of adenovirus containing the green fluorescent protein gene used as a reporter. Transfection with the shRNA plasmids partially inhibited the IFNAR1 expression. This inhibition substantially decreased antiviral response, demonstrated by the decrease of IFNAR1, IFN-α, and TNF-α gene expression, and the decrease at protein levels of IFNAR1, Protein kinase RNA-activated (PKR), and phosphorylated STAT1, allowing higher adenoviral transduction and transgene expression. Interestingly it was seen shRNA inhibited macrophage activation. These results suggest that the inhibition of the IFN-I pathway could be a strategy to minimize the immune response against Adenoviral vectors allowing higher Adenovirus transduction extending the transgene expression.
Collapse
Affiliation(s)
- J Guerrero-Rodríguez
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - A Cárdenas-Vargas
- Universidad Autónoma de Zacatecas, Jardín Juárez #147, Centro Histórico, C.P. 98000, Zacatecas, Zacatecas, Mexico
| | - G Gutierrez-Silerio
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - A Sobrevilla-Navarro
- Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555 Ejido San José Tateposco, C.P. 45425, Tonalá, Jalisco, Mexico
| | - B Bastidas-Ramírez
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - L Hernández-Ortega
- Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555 Ejido San José Tateposco, C.P. 45425, Tonalá, Jalisco, Mexico
| | - C Gurrola-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - L Gasca-Lozano
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - J Armendáriz-Borunda
- Instituto de Biología Molecular en Medicina y Terapia Génica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - A Salazar-Montes
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
11
|
Alhashimi M, Elkashif A, Sayedahmed EE, Mittal SK. Nonhuman Adenoviral Vector-Based Platforms and Their Utility in Designing Next Generation of Vaccines for Infectious Diseases. Viruses 2021; 13:1493. [PMID: 34452358 PMCID: PMC8402644 DOI: 10.3390/v13081493] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Several human adenoviral (Ad) vectors have been developed for vaccine delivery owing to their numerous advantages, including the feasibility of different vector designs, the robustness of elicited immune responses, safety, and scalability. To expand the repertoire of Ad vectors for receptor usage and circumvention of Ad vector immunity, the use of less prevalent human Ad types or nonhuman Ads were explored for vector design. Notably, many nonhuman Ad vectors have shown great promise in preclinical and clinical studies as vectors for vaccine delivery. This review describes the key features of several nonhuman Ad vector platforms and their implications in developing effective vaccines against infectious diseases.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Immunology and Infectious Disease, and Purdue University Center for Cancer Research, Department of Comparative Pathobiology, Purdue Institute for Inflammation, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2027, USA; (M.A.); (A.E.); (E.E.S.)
| |
Collapse
|
12
|
Detection and molecular characterization of enteric adenovirus in treated wastewater in the Brazilian Federal District. SN APPLIED SCIENCES 2021. [DOI: 10.1007/s42452-021-04678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractHuman enteric viruses, such as enteric adenoviruses (HAdV), are known to be involved with gastrointestinal disorders, especially acute gastroenteritis. Several studies have used HAdV as an indicator of water quality, since they are considered highly stable and widely distributed viruses in water matrices. The aim of this study was to detect and genotype HAdVs in water matrices impacted by discharges of treated effluents from wastewater treatment plants (WWTPs). Wastewater treatment plants from the sanitary system of the Brazilian Federal District were assessed in 2018 and 2019. Samples were collected upstream and downstream from discharge points for each WWTP. Viral concentration based on adsorption-elution and conventional PCR was used for molecular detection, and positive samples were sequenced for phylogenetic analysis. Pluviosity data for the period in which the samples were collected were obtained. Our results demonstrated the presence of HAdVs in 27.2% (61/224) of the samples. The positivity was significantly higher in downstream samples compared to upstream. Moreover, the HAdV positivity was higher in downstream samples collected from receiving water bodies impacted by secondary-level WWTPs in comparison with those impacted by tertiary-level WWTPs. Phylogenetic analysis demonstrated the presence of genotypes 40 and 41, with prevalence of HAdV genotype 41. Despite the predominance of HAdV-41, an increasing frequency of the HAdV-40 was associated with higher pluviosity. In conclusion, this study is the first documentation in the Brazilian Federal District dealing with the prevalence and diversity of HAdVs in several WWTP, along with their correlation with rainfall index.
Collapse
|
13
|
STAT2 signaling restricts viral dissemination but drives severe pneumonia in SARS-CoV-2 infected hamsters. Nat Commun 2020; 11:5838. [PMID: 33203860 PMCID: PMC7672082 DOI: 10.1038/s41467-020-19684-y] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
Emergence of SARS-CoV-2 causing COVID-19 has resulted in hundreds of thousands of deaths. In search for key targets of effective therapeutics, robust animal models mimicking COVID-19 in humans are urgently needed. Here, we show that Syrian hamsters, in contrast to mice, are highly permissive to SARS-CoV-2 and develop bronchopneumonia and strong inflammatory responses in the lungs with neutrophil infiltration and edema, further confirmed as consolidations visualized by micro-CT alike in clinical practice. Moreover, we identify an exuberant innate immune response as key player in pathogenesis, in which STAT2 signaling plays a dual role, driving severe lung injury on the one hand, yet restricting systemic virus dissemination on the other. Our results reveal the importance of STAT2-dependent interferon responses in the pathogenesis and virus control during SARS-CoV-2 infection and may help rationalizing new strategies for the treatment of COVID-19 patients.
Collapse
|
14
|
Zhang Z, Zhang C, Miao J, Wang Z, Wang Z, Cheng Z, Wang P, Dunmall LSC, Lemoine NR, Wang Y. A Tumor-Targeted Replicating Oncolytic Adenovirus Ad-TD-nsIL12 as a Promising Therapeutic Agent for Human Esophageal Squamous Cell Carcinoma. Cells 2020; 9:cells9112438. [PMID: 33182528 PMCID: PMC7698064 DOI: 10.3390/cells9112438] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers in China and existing therapies have been unable to significantly improve prognosis. Oncolytic adenoviruses (OAds) are novel promising anti-tumor drugs and have been evaluated in several cancers including ESCC. However, the antitumour efficacy of the first generation OAds (H101) as single agent is limited. Therefore, more effective OAds are needed. Our previous studies demonstrated that the novel oncolytic adenovirus Ad-TD-nsIL12 (human adenovirus type 5 with E1ACR2, E1B19K, E3gp19K-triple deletions)harboring human non-secretory IL-12 had significant anti-tumor effect, with no toxicity, in a Syrian hamster pancreatic cancer model. In this study, we evaluated the anti-tumor effect of Ad-TD-nsIL12 in human ESCC. The cytotoxicity of Ad-TD-nsIL12, H101 and cisplatin were investigated in two newly established patient-derived tumor cells (PDCs) and a panel of ESCC cell lines in vitro. A novel adenovirus-permissive, immune-deficient Syrian hamster model of PDCs subcutaneous xenograft was established for in vivo analysis of efficacy. The results showed that Ad-TD-nsIL12 was more cytotixic to and replicated more effectively in human ESCC cell lines than H101. Compared with cisplatin and H101, Ad-TD-nsIL12 could significantly inhibit tumor growth and tumor angiogenesis as well as enhance survival rate of animals with no side effects. These findings suggest that Ad-TD-nsIL12 has superior anti-tumor potency against human ESCC with a good safety profile.
Collapse
Affiliation(s)
- Zifang Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
| | - Chunyang Zhang
- Department of Surgical Sciences, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China;
| | - Jinxin Miao
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
- Department of Science and Technology, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhizhong Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
| | - Zhimin Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
| | - Zhenguo Cheng
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
| | - Louisa S. Chard Dunmall
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, UK;
| | - Nicholas R. Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, UK;
- Correspondence: (N.R.L.); (Y.W.); Tel.: +0044-207-8823500 (N.R.L.); +0044-207-8823596 (Y.W.)
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China; (Z.Z.); (J.M.); (Z.W.); (Z.W.); (Z.C.); (P.W.)
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, UK;
- Correspondence: (N.R.L.); (Y.W.); Tel.: +0044-207-8823500 (N.R.L.); +0044-207-8823596 (Y.W.)
| |
Collapse
|
15
|
Johansen MD, Irving A, Montagutelli X, Tate MD, Rudloff I, Nold MF, Hansbro NG, Kim RY, Donovan C, Liu G, Faiz A, Short KR, Lyons JG, McCaughan GW, Gorrell MD, Cole A, Moreno C, Couteur D, Hesselson D, Triccas J, Neely GG, Gamble JR, Simpson SJ, Saunders BM, Oliver BG, Britton WJ, Wark PA, Nold-Petry CA, Hansbro PM. Animal and translational models of SARS-CoV-2 infection and COVID-19. Mucosal Immunol 2020; 13:877-891. [PMID: 32820248 PMCID: PMC7439637 DOI: 10.1038/s41385-020-00340-z] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is causing a major once-in-a-century global pandemic. The scientific and clinical community is in a race to define and develop effective preventions and treatments. The major features of disease are described but clinical trials have been hampered by competing interests, small scale, lack of defined patient cohorts and defined readouts. What is needed now is head-to-head comparison of existing drugs, testing of safety including in the background of predisposing chronic diseases, and the development of new and targeted preventions and treatments. This is most efficiently achieved using representative animal models of primary infection including in the background of chronic disease with validation of findings in primary human cells and tissues. We explore and discuss the diverse animal, cell and tissue models that are being used and developed and collectively recapitulate many critical aspects of disease manifestation in humans to develop and test new preventions and treatments.
Collapse
Affiliation(s)
- M D Johansen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - A Irving
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, ZJU International Campus, Haining, China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - X Montagutelli
- Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - M D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - I Rudloff
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Paediatrics, Monash University, Clayton, VIC, 3168, Australia
| | - M F Nold
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Monash Newborn, Monash Children's Hospital, Clayton, VIC, Australia
| | - N G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia
| | - R Y Kim
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia
| | - C Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia
| | - G Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia
| | - A Faiz
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - K R Short
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - J G Lyons
- Centenary Institute and Dermatology, The University of Sydney and Cancer Services, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - G W McCaughan
- Centenary Institute and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - M D Gorrell
- Centenary Institute and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - A Cole
- Centenary Institute and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - C Moreno
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - D Couteur
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, and Faculty of Medicine and Health, Concord Clinical School, ANZAC Research Institute and Centre for Education and Research on Ageing, Sydney, Australia
| | - D Hesselson
- Centenary Institute and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - J Triccas
- Discipline of Infectious Diseases and Immunology, Central Clinical School, Faculty of Medicine and Health and the Charles Perkins Centre, The University of Sydney, Camperdown, Sydney, Australia
| | - G G Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - J R Gamble
- Centenary Institute and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - S J Simpson
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, and Faculty of Medicine and Health, Concord Clinical School, ANZAC Research Institute and Centre for Education and Research on Ageing, Sydney, Australia
| | - B M Saunders
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - B G Oliver
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia
- Woolcock Institute of Medical Research, Sydney, Australia
| | - W J Britton
- Centenary Institute, The University of Sydney and Department of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - P A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia
| | - C A Nold-Petry
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
| | - P M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, Australia.
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
16
|
Miao JX, Wang JY, Li HZ, Guo HR, Dunmall LSC, Zhang ZX, Cheng ZG, Gao DL, Dong JZ, Wang ZD, Wang YH. Promising xenograft animal model recapitulating the features of human pancreatic cancer. World J Gastroenterol 2020; 26:4802-4816. [PMID: 32921958 PMCID: PMC7459204 DOI: 10.3748/wjg.v26.i32.4802] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/01/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Multiple sites of metastasis and desmoplastic reactions in the stroma are key features of human pancreatic cancer (PC). There are currently no simple and reliable animal models that can mimic these features for accurate disease modeling.
AIM To create a new xenograft animal model that can faithfully recapitulate the features of human PC.
METHODS Interleukin 2 receptor subunit gamma (IL2RG) gene knockout Syrian hamster was created and characterized. A panel of human PC cell lines were transplanted into IL2RG knockout Syrian hamsters and severe immune-deficient mice subcutaneously or orthotopically. Tumor growth, local invasion, remote organ metastasis, histopathology, and molecular alterations of tumor cells and stroma were compared over time.
RESULTS The Syrian hamster with IL2RG gene knockout (named ZZU001) demonstrated an immune-deficient phenotype and function. ZZU001 hamsters faithfully recapitulated most features of human PC, in particular, they developed metastasis at multiple sites. PC tissues derived from ZZU001 hamsters displayed desmoplastic reactions in the stroma and epithelial to mesenchymal transition phenotypes, whereas PC tissues derived from immune-deficient mice did not present such features.
CONCLUSION ZZU001 hamsters engrafted with human PC cells are a superior animal model compared to immune-deficient mice. ZZU001 hamsters can be a valuable animal model for better understanding the molecular mechanism of tumorigenesis and metastasis and the evaluation of new drugs targeting human PC.
Collapse
Affiliation(s)
- Jin-Xin Miao
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Jian-Yao Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Hao-Ze Li
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Hao-Ran Guo
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Louisa S Chard Dunmall
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| | - Zhong-Xian Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Zhen-Guo Cheng
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Dong-Ling Gao
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Jian-Zeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhong-De Wang
- Department of Animal Dairy, and Veterinary Sciences, Utah State University, Logan UT 84341, United States
| | - Yao-He Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, China
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| |
Collapse
|
17
|
Li R, Ying B, Liu Y, Spencer JF, Miao J, Tollefson AE, Brien JD, Wang Y, Wold WSM, Wang Z, Toth K. Generation and characterization of an Il2rg knockout Syrian hamster model for XSCID and HAdV-C6 infection in immunocompromised patients. Dis Model Mech 2020; 13:dmm044602. [PMID: 32651192 PMCID: PMC7473636 DOI: 10.1242/dmm.044602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
Model animals are indispensable for the study of human diseases, and in general, of complex biological processes. The Syrian hamster is an important model animal for infectious diseases, behavioral science and metabolic science, for which more experimental tools are becoming available. Here, we describe the generation and characterization of an interleukin-2 receptor subunit gamma (Il2rg) knockout (KO) Syrian hamster strain. In humans, mutations in IL2RG can result in a total failure of T and natural killer (NK) lymphocyte development and nonfunctional B lymphocytes (X-linked severe combined immunodeficiency; XSCID). Therefore, we sought to develop a non-murine model to study XSCID and the infectious diseases associated with IL2RG deficiency. We demonstrated that the Il2rg KO hamsters have a lymphoid compartment that is greatly reduced in size and diversity, and is impaired in function. As a result of the defective adaptive immune response, Il2rg KO hamsters developed a more severe human adenovirus infection and cleared virus less efficiently than immune competent wild-type hamsters. Because of this enhanced virus replication, Il2rg KO hamsters developed more severe adenovirus-induced liver pathology than wild-type hamsters. This novel hamster strain will provide researchers with a new tool to investigate human XSCID and its related infections.
Collapse
Affiliation(s)
- Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Yanan Liu
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
- National Center for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - William S M Wold
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| |
Collapse
|
18
|
Ranadheera C, Valcourt EJ, Warner BM, Poliquin G, Rosenke K, Frost K, Tierney K, Saturday G, Miao J, Westover JB, Gowen BB, Booth S, Feldmann H, Wang Z, Safronetz D. Characterization of a novel STAT 2 knock-out hamster model of Crimean-Congo hemorrhagic fever virus pathogenesis. Sci Rep 2020; 10:12378. [PMID: 32704046 PMCID: PMC7378551 DOI: 10.1038/s41598-020-69054-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/30/2020] [Indexed: 01/30/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne pathogen causing a febrile illness in humans, which can progress to hemorrhagic manifestations, multi-organ failure, and death. Current mouse models of CCHFV infection reliably succumb to virus challenge but vary in their ability to reflect signs of disease similar to humans. In this study, we established a signal transducer and activator of transcription 2 (STAT2) knockout hamster model to expand the repertoire of animal models of CCHFV pathogenesis that can be used for therapeutic development. These hamsters demonstrated a systemic and lethal disease in response to infection. Hallmarks of human disease were observed including petechial rash, blood coagulation dysfunction, and various biochemistry and blood cell count abnormalities. Furthermore, we also demonstrated the utility of this model for anti-CCHFV therapeutic evaluation. The STAT2 knock-out hamster model of CCHFV infection may provide some further insights into clinical disease, viral pathogenesis, and pave the way for testing of potential drug and vaccine candidates.
Collapse
Affiliation(s)
- Charlene Ranadheera
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Bioforensics Assay Development and Diagnostics, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Emelissa J Valcourt
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Bryce M Warner
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Guillaume Poliquin
- Office of the Scientific Director, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Kyle Rosenke
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Kathy Frost
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Kevin Tierney
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA.,Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450066, People's Republic of China
| | - Jonna B Westover
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Brian B Gowen
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Stephanie Booth
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada. .,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
19
|
STAT3/5 Inhibitors Suppress Proliferation in Bladder Cancer and Enhance Oncolytic Adenovirus Therapy. Int J Mol Sci 2020; 21:ijms21031106. [PMID: 32046095 PMCID: PMC7043223 DOI: 10.3390/ijms21031106] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 02/08/2023] Open
Abstract
The JAK-STAT signalling pathway regulates cellular processes like cell division, cell death and immune regulation. Dysregulation has been identified in solid tumours and STAT3 activation is a marker for poor outcome. The aim of this study was to explore potential therapeutic strategies by targeting this pathway in bladder cancer (BC). High STAT3 expression was detected in 51.3% from 149 patient specimens with invasive bladder cancer by immunohistochemistry. Protein expression of JAK, STAT and downstream targets were confirmed in 10 cell lines. Effects of the JAK inhibitors Ruxolitinib and BSK-805, and STAT3/5 inhibitors Stattic, Nifuroxazide and SH-4-54 were analysed by cell viability assays, immunoblotting, apoptosis and cell cycle progression. Treatment with STAT3/5 but not JAK1/2 inhibitors reduced survival, levels of phosphorylated STAT3 and Cyclin-D1 and increased apoptosis. Tumour xenografts, using the chicken chorioallantoic membrane (CAM) model responded to Stattic monotherapy. Combination of Stattic with Cisplatin, Docetaxel, Gemcitabine, Paclitaxel and CDK4/6 inhibitors showed additive effects. The combination of Stattic with the oncolytic adenovirus XVir-N-31 increased viral replication and cell lysis. Our results provide evidence that inhibitors against STAT3/5 are promising as novel mono- and combination therapy in bladder cancer.
Collapse
|
20
|
Miao J, Chard LS, Wang Z, Wang Y. Syrian Hamster as an Animal Model for the Study on Infectious Diseases. Front Immunol 2019; 10:2329. [PMID: 31632404 PMCID: PMC6781508 DOI: 10.3389/fimmu.2019.02329] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/16/2019] [Indexed: 11/13/2022] Open
Abstract
Infectious diseases still remain one of the biggest challenges for human health. In order to gain a better understanding of the pathogenesis of infectious diseases and develop effective diagnostic tools, therapeutic agents, and preventive vaccines, a suitable animal model which can represent the characteristics of infectious is required. The Syrian hamster immune responses to infectious pathogens are similar to humans and as such, this model is advantageous for studying pathogenesis of infection including post-bacterial, viral and parasitic pathogens, along with assessing the efficacy and interactions of medications and vaccines for those pathogens. This review summarizes the current status of Syrian hamster models and their use for understanding the underlying mechanisms of pathogen infection, in addition to their use as a drug discovery platform and provides a strong rationale for the selection of Syrian hamster as animal models in biomedical research. The challenges of using Syrian hamster as an alternative animal model for the research of infectious diseases are also addressed.
Collapse
Affiliation(s)
- Jinxin Miao
- Department of Science and Technology, Henan University of Chinese Medicine, Zhengzhou, China
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S. Chard
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhimin Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
21
|
Diminished stimulator of interferon genes production with cigarette smoke-exposure contributes to weakened anti-adenovirus vectors response and destruction of lung in chronic obstructive pulmonary disease model. Exp Cell Res 2019; 384:111545. [PMID: 31470016 DOI: 10.1016/j.yexcr.2019.111545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 12/14/2022]
Abstract
Cigarette smoke (CS) is the primary risk factor for chronic obstructive pulmonary disease (COPD) and dampens antiviral response, which increases viral infections and leads to COPD acute exacerbation (AECOPD). Adenovirus, a nonenveloped DNA virus, is linked with AECOPD, whose DNAs trigger innate immune response via interacting with pattern recognition receptors (PRRs). Stimulator of interferon genes (STING), as a cytosolic DNA sensor, participates in adenovirus-induced interferon β (IFNβ)-dependent antiviral response. STING is involved in various pulmonary diseases, but role of STING in pathogenesis of AECOPD is not well documented. In the present study, we explored relationship between STING and AECOPD induced by recombinant adenovirus vectors (rAdVs) and CS in wild type (WT) and STING-/- mice; and also characterized the inhibition of STING- IFNβ pathway in pulmonary epithelium exposed to cigarette smoke extract (CSE). We found that CS or CSE exposure alone dramatically inhibited STING expression, but not significantly effected IFNβ production. Moreover, CS or CSE-exposed significantly suppressed activation of STING-IFNβ pathway induced by rAdVs and suppressed clearance of rAdVs DNA. Inflammation, fibrosis and emphysema of lung tissues were exaggerated when treated with CS plus rAdVs, which further deteriorate in absences of STING. In A549 cells with knockdown of STING, we also observed enhancing apoptosis related to emphysema, especially CSE and adenovirus vectors in combination. Therefore, STING may play a protective role in preventing the progress of COPD.
Collapse
|
22
|
Wold WSM, Tollefson AE, Ying B, Spencer JF, Toth K. Drug development against human adenoviruses and its advancement by Syrian hamster models. FEMS Microbiol Rev 2019; 43:380-388. [PMID: 30916746 DOI: 10.1093/femsre/fuz008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/25/2019] [Indexed: 02/02/2023] Open
Abstract
The symptoms of human adenovirus infections are generally mild and self-limiting. However, these infections have been gaining importance in recent years because of a growing number of immunocompromised patients. Solid organ and hematopoietic stem cell transplant patients are subjected to severe immunosuppressive regimes and cannot efficaciously eliminate virus infections. In these patients, adenovirus infections can develop into deadly multi-organ disseminated disease. Presently, in the absence of approved therapies, physicians rely on drugs developed for other purposes to treat adenovirus infections. As there is a need for anti-adenoviral therapies, researchers have been developing new agents and repurposing existing ones to treat adenovirus infections. There are several small molecule drugs that are being tested for their efficacy against human adenoviruses; some of these have reached clinical trials, while others are still in the preclinical phase. Besides these compounds, research on immunotherapy against adenoviral infection has made significant progress, promising another modality for treatment. The availability of an animal model confirmed the activity of some drugs already in clinical use while proving that others are inactive. This led to the identification of several lead compounds that await further development. In the present article, we review the current status of anti-adenoviral therapies and their advancement by in vivo studies in the Syrian hamster model.
Collapse
Affiliation(s)
- William S M Wold
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Ann E Tollefson
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Baoling Ying
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Jacqueline F Spencer
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Karoly Toth
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| |
Collapse
|
23
|
Dehler CE, Lester K, Della Pelle G, Jouneau L, Houel A, Collins C, Dovgan T, Machat R, Zou J, Boudinot P, Martin SAM, Collet B. Viral Resistance and IFN Signaling in STAT2 Knockout Fish Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:465-475. [PMID: 31142600 PMCID: PMC6612602 DOI: 10.4049/jimmunol.1801376] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/30/2019] [Indexed: 01/17/2023]
Abstract
IFN belong to a group of cytokines specialized in the immunity to viruses. Upon viral infection, type I IFN is produced and alters the transcriptome of responding cells through induction of a set of IFN stimulated genes (ISGs) with regulatory or antiviral function, resulting in a cellular antiviral state. Fish genomes have both type I IFN and type II IFN (IFN-γ), but no type III (λ) IFN has been identified. Their receptors are not simple counterparts of the mammalian type I/II IFN receptors, because alternative chains are used in type I IFN receptors. The mechanisms of the downstream signaling remain partly undefined. In mammals, members of the signal transducer and activator of family of transcription factors are responsible for the transmission of the signal from cytokine receptors, and STAT2 is required for type I but not type II IFN signaling. In fish, its role in IFN signaling in fish remains unclear. We isolated a Chinook salmon (Oncorhynchus tshawytscha) cell line, GS2, with a stat2 gene knocked out by CRISPR/Cas9 genome editing. In this cell line, the induction of ISGs by stimulation with a recombinant type I IFN is completely obliterated as evidenced by comparative RNA-seq analysis of the transcriptome of GS2 and its parental counterpart, EC. Despite a complete absence of ISGs induction, the GS2 cell line has a remarkable ability to resist to viral infections. Therefore, other STAT2-independent pathways may be induced by the viral infection, illustrating the robustness and redundancy of the innate antiviral defenses in fish.
Collapse
Affiliation(s)
| | - Katherine Lester
- Marine Scotland, Marine Laboratory, AB11 9DB Aberdeen, United Kingdom; and
| | - Giulia Della Pelle
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| | - Luc Jouneau
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| | - Armel Houel
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| | - Catherine Collins
- Marine Scotland, Marine Laboratory, AB11 9DB Aberdeen, United Kingdom; and
| | - Tatiana Dovgan
- University of Aberdeen, AB24 2TZ Aberdeen, United Kingdom
- Marine Scotland, Marine Laboratory, AB11 9DB Aberdeen, United Kingdom; and
| | - Radek Machat
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| | - Jun Zou
- University of Aberdeen, AB24 2TZ Aberdeen, United Kingdom
| | - Pierre Boudinot
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| | | | - Bertrand Collet
- Marine Scotland, Marine Laboratory, AB11 9DB Aberdeen, United Kingdom; and
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78352 Jouy-en-Josas cedex, France
| |
Collapse
|
24
|
Overcoming the limitations of locally administered oncolytic virotherapy. BMC Biomed Eng 2019; 1:17. [PMID: 32903299 PMCID: PMC7422506 DOI: 10.1186/s42490-019-0016-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
Adenovirus (Ad) has been most extensively evaluated gene transfer vector in clinical trials due to facile production in high viral titer, highly efficient transduction, and proven safety record. Similarly, an oncolytic Ad, which replicates selectively in cancer cells through genetic modifications, is actively being evaluated in various phases of clinical trials as a promising next generation therapeutic against cancer. Most of these trials with oncolytic Ads to date have employed intratumoral injection as the standard administration route. Although these locally administered oncolytic Ads have shown promising outcomes, the therapeutic efficacy is not yet optimal due to poor intratumoral virion retention, nonspecific shedding of virion to normal organs, variable infection efficacy due to heterogeneity of tumor cells, adverse antiviral immune response, and short biological activity of oncolytic viruses in situ. These inherent problems associated with locally administered Ad also holds true for other oncolytic viral vectors. Thus, this review will aim to discuss various nanomaterial-based delivery strategies to improve the intratumoral administration efficacy of oncolytic Ad as well as other types of oncolytic viruses.
Collapse
|
25
|
He Z, Chen X, Fu M, Tang J, Li X, Cao H, Wang Y, Zheng SJ. Inhibition of fowl adenovirus serotype 4 replication in Leghorn male hepatoma cells by SP600125 via blocking JNK MAPK pathway. Vet Microbiol 2019; 228:45-52. [DOI: 10.1016/j.vetmic.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022]
|
26
|
Atkins C, Miao J, Kalveram B, Juelich T, Smith JK, Perez D, Zhang L, Westover JLB, Van Wettere AJ, Gowen BB, Wang Z, Freiberg AN. Natural History and Pathogenesis of Wild-Type Marburg Virus Infection in STAT2 Knockout Hamsters. J Infect Dis 2018; 218:S438-S447. [PMID: 30192975 PMCID: PMC6249581 DOI: 10.1093/infdis/jiy457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Marburg virus (MARV; family Filoviridae) causes sporadic outbreaks of Marburg hemorrhagic fever in sub-Saharan Africa with case fatality rates reaching 90%. Wild-type filoviruses, including MARV and the closely related Ebola virus, are unable to suppress the type I interferon response in rodents, and therefore require adaptation of the viruses to cause disease in immunocompetent animals. In the current study, we demonstrate that STAT2 knockout Syrian hamsters are susceptible to infection with different wild-type MARV variants. MARV Musoke causes a robust and systemic infection resulting in lethal disease. Histopathological findings share features similar to those observed in human patients and other animal models of filovirus infection. Reverse-transcription polymerase chain reaction analysis of host transcripts shows a dysregulation of the innate immune response. Our results demonstrate that the STAT2 knockout hamster represents a novel small animal model of severe MARV infection and disease without the requirement for virus adaptation.
Collapse
Affiliation(s)
- Colm Atkins
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Birte Kalveram
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Terry Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jennifer K Smith
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - David Perez
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Lihong Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jonna L B Westover
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Arnaud J Van Wettere
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Brian B Gowen
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston
| |
Collapse
|
27
|
Abstract
Human mastadenovirus (HAdVs) can cause a broad spectrum of diseases in both children and adults, including acute respiratory infection, gastroenteritis, epidemic keratoconjunctivitis. Populations susceptible to adenovirus infection include children, immunocompromised patients and military recruits. To date, seven species (A-G) including more than 79 genotypes have been characterized, of which HAdV-B3, B4, B7 and the recently reemerged types 14 and 55 often lead to severe pneumonia. The live oral enteric-coated adenovirus type 4 and 7 vaccine, which was approved for use in US military personnel of 17 through 50 years of age, had been shown to be safe and highly effective in numerous clinical trials and by ongoing surveillance of febrile respiratory illness. However, there is currently no vaccine approved for general use in children and adults in any part of the world. This review article will summarize the epidemiological data available for adenovirus and the effectiveness of the adenovirus vaccine in the US military. It will also provide a brief overview of innovative vaccine strategies, animal models for vaccine evaluation, and issues regarding vaccine production.
Collapse
Affiliation(s)
- Shiying Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510182, China
| | - Xingui Tian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510182, China
| |
Collapse
|
28
|
STAT2-Dependent Immune Responses Ensure Host Survival despite the Presence of a Potent Viral Antagonist. J Virol 2018; 92:JVI.00296-18. [PMID: 29743368 PMCID: PMC6026732 DOI: 10.1128/jvi.00296-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/04/2018] [Indexed: 01/12/2023] Open
Abstract
A pathogen encounter induces interferons, which signal via Janus kinases and STAT transcription factors to establish an antiviral state. However, the host and pathogens are situated in a continuous arms race which shapes host evolution toward optimized immune responses and the pathogens toward enhanced immune-evasive properties. Mouse cytomegalovirus (MCMV) counteracts interferon responses by pM27-mediated degradation of STAT2, which directly affects the signaling of type I as well as type III interferons. Using MCMV mutants lacking M27 and mice lacking STAT2, we studied the opposing relationship between antiviral activities and viral antagonism in a natural host-pathogen pair in vitro and in vivo In contrast to wild-type (wt) MCMV, ΔM27 mutant MCMV was efficiently cleared from all organs within a few days in BALB/c, C57BL/6, and 129 mice, highlighting the general importance of STAT2 antagonism for MCMV replication. Despite this effective and relevant STAT2 antagonism, wt and STAT2-deficient mice exhibited fundamentally different susceptibilities to MCMV infections. MCMV replication was increased in all assessed organs (e.g., liver, spleen, lungs, and salivary glands) of STAT2-deficient mice, resulting in mortality during the first week after infection. Taken together, the results of our study reveal the importance of cytomegaloviral interferon antagonism for viral replication as well as a pivotal role of the remaining STAT2 activity for host survival. This mutual influence establishes a stable evolutionary standoff situation with fatal consequences when the equilibrium is disturbed.IMPORTANCE The host limits viral replication by the use of interferons (IFNs), which signal via STAT proteins. Several viruses evolved antagonists targeting STATs to antagonize IFNs (e.g., cytomegaloviruses, Zika virus, dengue virus, and several paramyxoviruses). We analyzed infections caused by MCMV expressing or lacking the STAT2 antagonist pM27 in STAT2-deficient and control mice to evaluate its importance for the host and the virus in vitro and in vivo The inability to counteract STAT2 directly translates into exaggerated IFN susceptibility in vitro and pronounced attenuation in vivo Thus, the antiviral activity mediated by IFNs via STAT2-dependent signaling drove the development of a potent MCMV-encoded STAT2 antagonist which became indispensable for efficient virus replication and spread to organs required for dissemination. Despite this clear impact of viral STAT2 antagonism, the host critically required the remaining STAT2 activity to prevent overt disease and mortality upon MCMV infection. Our findings highlight a remarkably delicate balance between host and virus.
Collapse
|
29
|
Radke JR, Cook JL. Human adenovirus infections: update and consideration of mechanisms of viral persistence. Curr Opin Infect Dis 2018; 31:251-256. [PMID: 29601326 PMCID: PMC6367924 DOI: 10.1097/qco.0000000000000451] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW To provide an update on recent studies of human adenoviral (HAdV) infections and to explore the mechanisms of viral persistence and the role of persistent infection in disseminated disease in immunocompromised patients. RECENT FINDINGS Human adenoviruses continue to be a problem in ophthalmology clinics and to cause periodic, limited, global outbreaks of respiratory disease. Ad14p1 remains in worldwide circulation and continues to result in miniepidemics of severe respiratory infections. New variants of Ad4 and Ad7 have emerged in both the United States and Asia. The severity of Ad4 infections in outbreaks appears to depend more on preexisting conditions in patients than on genetically determined, viral virulence factors, in contrast to limited evidence of Ad7 mutations that may convey increased viral pathogenesis. Reactivation of persistent adenovirus infection appears to be the primary source of disseminated infections in immunocompromised patients. New studies suggest that establishment of persistent infection and reactivation are related to variations in interferon-mediated control of viral replication. SUMMARY Innate immune responses can create a state of adenoviral persistence, and repression of these host defenses can result in reactivation and dissemination of infection. A better definition of the molecular mechanisms of immune-mediated control of viral replication might lead to new strategies for treatment of HAdV reactivation and dissemination.
Collapse
Affiliation(s)
- Jay R Radke
- Boise VA Hospital, Idaho Veterans Research and Education Foundation, Boise, Idaho
| | - James L Cook
- Loyola University Chicago - Stritch School of Medicine and Edward Hines, Jr. Veterans Administration Hospital, Chicago, Illinois, USA
| |
Collapse
|
30
|
Miao J, Ying B, Li R, Tollefson AE, Spencer JF, Wold WSM, Song SH, Kong IK, Toth K, Wang Y, Wang Z. Characterization of an N-Terminal Non-Core Domain of RAG1 Gene Disrupted Syrian Hamster Model Generated by CRISPR Cas9. Viruses 2018; 10:E243. [PMID: 29734775 PMCID: PMC5977236 DOI: 10.3390/v10050243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
The accumulating evidence demonstrates that Syrian hamsters have advantages as models for various diseases. To develop a Syrian hamster (Mesocricetus auratus) model of human immunodeficiency caused by RAG1 gene mutations, we employed the CRISPR/Cas9 system and introduced an 86-nucleotide frameshift deletion in the hamster RAG1 gene encoding part of the N-terminal non-core domain of RAG1. Histological and immunohistochemical analyses demonstrated that these hamsters (referred herein as RAG1-86nt hamsters) had atrophic spleen and thymus, and developed significantly less white pulp and were almost completely devoid of splenic lymphoid follicles. The RAG1-nt86 hamsters had barely detectable CD3⁺ and CD4⁺ T cells. The expression of B and T lymphocyte-specific genes (CD3γ and CD4 for T cell-specific) and (CD22 and FCMR for B cell-specific) was dramatically reduced, whereas the expression of macrophage-specific (CD68) and natural killer (NK) cell-specific (CD94 and KLRG1) marker genes was increased in the spleen of RAG1-nt86 hamsters compared to wildtype hamsters. Interestingly, despite the impaired development of B and T lymphocytes, the RAG1-86nt hamsters still developed neutralizing antibodies against human adenovirus type C6 (HAdV-C6) upon intranasal infection and were capable of clearing the infectious viruses, albeit with slower kinetics. Therefore, the RAG1-86nt hamster reported herein (similar to the hypomorphic RAG1 mutations in humans that cause Omenn syndrome), may provide a useful model for studying the pathogenesis of the specific RAG1-mutation-induced human immunodeficiency, the host immune response to adenovirus infection and other pathogens as well as for evaluation of cell and gene therapies for treatment of this subset of RAG1 mutation patients.
Collapse
Affiliation(s)
- Jinxin Miao
- Department of Pathology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA.
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA.
| | - Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - William S M Wold
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Seok-Hwan Song
- Department of Animal Science, Division of Applied Life Science (BK21Plus), Graduate School of Gyeongsang National University, Jinju 52828, Korea.
| | - Il-Keun Kong
- Department of Animal Science, Division of Applied Life Science (BK21Plus), Graduate School of Gyeongsang National University, Jinju 52828, Korea.
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea.
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Yaohe Wang
- Department of Pathology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA.
- Auratus Bio, LLC., Canton, SD 57104, USA.
| |
Collapse
|
31
|
Morinaga T, Nguyễn TTT, Zhong B, Hanazono M, Shingyoji M, Sekine I, Tada Y, Tatsumi K, Shimada H, Hiroshima K, Tagawa M. An image cytometric technique is a concise method to detect adenoviruses and host cell proteins and to monitor the infection and cellular responses induced. Virol J 2017; 14:219. [PMID: 29126418 PMCID: PMC5681831 DOI: 10.1186/s12985-017-0888-0] [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: 06/04/2017] [Accepted: 10/31/2017] [Indexed: 11/30/2022] Open
Abstract
Background Genetically modified adenoviruses (Ad) with preferential replications in tumor cells have been examined for a possible clinical applicability as an anti-cancer agent. A simple method to detect viral and cellular proteins is valuable to monitor the viral infections and to predict the Ad-mediated cytotoxicity. Methods We used type 5 Ad in which the expression of E1A gene was activated by 5′-regulatory sequences of genes that were augmented in the expression in human tumors. The Ad were further modified to have the fiber-knob region replaced with that derived from type 35 Ad. We infected human mesothelioma cells with the fiber-replaced Ad, and sequentially examined cytotoxic processes together with an expression level of the viral E1A, hexon, and cellular cleaved caspase-3 with image cytometric and Western blot analyses. Results The replication-competent Ad produced cytotoxicity on mesothelioma cells. The infected cells expressed E1A and hexon 24 h after the infection and then showed cleavage of caspase-3, all of which were detected with image cytometry and Western blot analysis. Image cytometry furthermore demonstrated that increased Ad doses did not enhance an expression level of E1A and hexon in an individual cell and that caspase-3-cleaved cells were found more frequently in hexon-positive cells than in E1A-positive cells. Image cytometry thus detected these molecular changes in a sensitive manner and at a single cell level. We also showed that an image cytometric technique detected expression changes of other host cell proteins, cyclin-E and phosphorylated histone H3 at a single cell level. Conclusions Image cytometry is a concise procedure to detect expression changes of Ad and host cell proteins at a single cell level, and is useful to analyze molecular events after the infection. Electronic supplementary material The online version of this article (10.1186/s12985-017-0888-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takao Morinaga
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, 260-8717, Japan
| | - Thảo Thi Thanh Nguyễn
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, 260-8717, Japan.,Department of Molecular Biology and Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Boya Zhong
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, 260-8717, Japan.,Department of Molecular Biology and Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Michiko Hanazono
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, 260-8717, Japan
| | | | - Ikuo Sekine
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuji Tada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideaki Shimada
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Kenzo Hiroshima
- Department of Pathology, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo, Japan
| | - Masatoshi Tagawa
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, 260-8717, Japan. .,Department of Molecular Biology and Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
32
|
Ying B, Spencer JF, Tollefson AE, Wold WSM, Toth K. Male Syrian hamsters are more susceptible to intravenous infection with species C human adenoviruses than are females. Virology 2017; 514:66-78. [PMID: 29132049 DOI: 10.1016/j.virol.2017.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/11/2017] [Accepted: 10/19/2017] [Indexed: 12/25/2022]
Abstract
Recently, increasing attention has been focused on the influence of sex on the course of infectious diseases. Thus far, the best-documented examples point toward an immune-mediated mechanism: the generally stronger immune response in females can result in a faster clearance of the pathogen or, conversely, a more severe immune-mediated pathology. Here, we report that human species C adenoviruses replicate more and cause more pathology in male Syrian hamsters than in females. We also show that this sex disparity is not caused by a stronger immune response to the infection by the female hamsters. Rather, the liver of male hamsters is more susceptible to adenovirus infection: after intravenous injection, more hepatocytes become infected in male animals than in females. We hypothesize that Kupffer cells (hepatic tissue macrophages) of female animals are more active in sequestering circulating virions, and thus protect hepatocytes more efficiently than those of males.
Collapse
Affiliation(s)
- Baoling Ying
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, St. Louis, MO, USA
| | - Jacqueline F Spencer
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, St. Louis, MO, USA
| | - Ann E Tollefson
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, St. Louis, MO, USA
| | - William S M Wold
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, St. Louis, MO, USA.
| | - Karoly Toth
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, St. Louis, MO, USA.
| |
Collapse
|
33
|
St Claire MC, Ragland DR, Bollinger L, Jahrling PB. Animal Models of Ebolavirus Infection. Comp Med 2017; 67:253-262. [PMID: 28662754 PMCID: PMC5482517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/29/2016] [Accepted: 01/26/2017] [Indexed: 06/07/2023]
Abstract
Ebola virus is a highly pathogenic member of the family Filoviridae that causes a severe hemorrhagic disease in humans and NHP. The 2013-2016 West African outbreak has increased interest in the development and refinement of animal models of Ebola virus disease. These models are used to test countermeasures and vaccines, gain scientific insights into the mechanisms of disease progression and transmission, and study key correlates of immunology. Ebola virus is classified as a BSL4 pathogen and Category A agent, for which the United States government requires preparedness in case of bioterrorism. Rodents, such as Syrian golden hamsters (Mesocricetus auratus), mice (Mus musculus), and guinea pigs (Cavia porcellus), are the most common research species. However, NHP, especially macaques, are favored for Ebola virus disease research due to similarities with humans regarding the pathogenesis, clinical presentation, laboratory findings, and causes of fatality. To satisfy the regulatory requirements for approval of countermeasures against high-consequence pathogens, the FDA instituted the Animal Rule, which permits efficacy studies in animal models in place of human clinical data when such studies are not feasible or ethical. This review provides a comprehensive summary of various animal models and their use in Ebola virus disease research.
Collapse
Affiliation(s)
- Marisa C St Claire
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland;,
| | - Dan R Ragland
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Laura Bollinger
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Peter B Jahrling
- Integrated Research Facility, Division of Clinical Research, Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| |
Collapse
|
34
|
Tollefson AE, Ying B, Spencer JF, Sagartz JE, Wold WSM, Toth K. Pathology in Permissive Syrian Hamsters after Infection with Species C Human Adenovirus (HAdV-C) Is the Result of Virus Replication: HAdV-C6 Replicates More and Causes More Pathology than HAdV-C5. J Virol 2017; 91:e00284-17. [PMID: 28250128 PMCID: PMC5411597 DOI: 10.1128/jvi.00284-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 02/06/2023] Open
Abstract
Syrian hamsters are permissive for the replication of species C human adenoviruses (HAdV-C). The virus replicates to high titers in the liver of these animals after intravenous infection, while respiratory infection results in virus replication in the lung. Here we show that two types belonging to species C, HAdV-C5 and HAdV-C6, replicate to significantly different extents and cause pathology with significantly different severities, with HAdV-C6 replicating better and inducing more severe and more widespread lesions. The virus burdens in the livers of HAdV-C6-infected hamsters are higher than the virus burdens in HAdV-C5-infected ones because more of the permissive hepatocytes get infected. Furthermore, when hamsters are infected intravenously with HAdV-C6, live, infectious virus can be isolated from the lung and the kidney, which is not seen with HAdV-C5. Similarly to mouse models, in hamsters, HAdV-C6 is sequestered by macrophages to a lesser degree than HAdV-C5. Depletion of Kupffer cells from the liver greatly increases the replication of HAdV-C5 in the liver, while it has only a modest effect on the replication of HAdV-C6. Elimination of Kupffer cells also dramatically increases the pathology induced by HAdV-C5. These findings indicate that in hamsters, pathology resulting from intravenous infection with adenoviruses is caused mostly by replication in hepatocytes and not by the abortive infection of Kupffer cells and the following cytokine storm.IMPORTANCE Immunocompromised human patients can develop severe, often lethal adenovirus infections. Respiratory adenovirus infection among military recruits is a serious problem, in some cases requiring hospitalization of the patient. Furthermore, adenovirus-based vectors are frequently used as experimental viral therapeutic agents. Thus, it is imperative that we investigate the pathogenesis of adenoviruses in a permissive animal model. Syrian hamsters are susceptible to infection with certain human adenoviruses, and the pathology accompanying these infections is similar to what is observed with adenovirus-infected human patients. We demonstrate that replication in permissive cells in a susceptible host animal is a major part of the mechanism by which systemic adenovirus infection induces pathology, as opposed to the chiefly immune-mediated pathology observed in nonsusceptible hosts. These findings support the use of compounds inhibiting adenovirus replication as a means to block adenovirus-induced pathology.
Collapse
Affiliation(s)
- Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - John E Sagartz
- Department of Comparative Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - William S M Wold
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
35
|
Siddharthan V, Van Wettere AJ, Li R, Miao J, Wang Z, Morrey JD, Julander JG. Zika virus infection of adult and fetal STAT2 knock-out hamsters. Virology 2017; 507:89-95. [PMID: 28431283 DOI: 10.1016/j.virol.2017.04.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 02/02/2023]
Abstract
Zika virus (ZIKV) infection was investigated in adult and fetal STAT2 knock-out (KO) hamsters. Subcutaneous injection of ZIKV of adults resulted in morbidity, mortality, and infection of the uterus, placenta, brain, spinal cord, and testicles, thus providing an opportunity to evaluate congenital ZIKV infection in a second rodent species besides mice. ZIKV-infected cells with morphologies of Sertoli cells and spermatogonia were observed in the testes, which may have implications for sexual transmission and male sterility. Neonates exposed as fetuses to ZIKV at 8 days post-coitus were not smaller than controls. Nevertheless, infectious virus and ZIKV RNA was detected in some, but not all, placentas and fetal brains of KO hamsters. STAT2 KO hamsters may be useful for addressing sexual transmission, pathogenesis, routes of fetal infection, and neurological disease outcomes, and may also be used in antiviral or vaccine studies to identify intervention strategies.
Collapse
Affiliation(s)
- Venkatraman Siddharthan
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-9825, United States.
| | - Arnaud J Van Wettere
- Utah Veterinary Diagnostics Laboratory, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States.
| | - Rong Li
- Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States
| | - Jinxin Miao
- Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States
| | - Zhongde Wang
- Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States.
| | - John D Morrey
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-9825, United States.
| | - Justin G Julander
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, Utah State University, 5600 Old Main Hill, Logan, UT 84322-9825, United States.
| |
Collapse
|
36
|
Shen CF, Wang SM, Ho TS, Liu CC. Clinical features of community acquired adenovirus pneumonia during the 2011 community outbreak in Southern Taiwan: role of host immune response. BMC Infect Dis 2017; 17:196. [PMID: 28270104 PMCID: PMC5341368 DOI: 10.1186/s12879-017-2272-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/17/2017] [Indexed: 11/11/2022] Open
Abstract
Background Human adenovirus 7 (HAdV-7) was responsible for a significant number of fatalities during the 2011 community outbreak in Taiwan. The mechanisms underlying the pathogenesis of severe adenovirus infections in non-immunocompromised individuals remain unclear. Adenovirus pneumonia was associated with pleural effusion in a number of patients from the 2011 outbreak suggesting that similar to bacterial pneumonia, patients diagnosed with adenovirus pneumonia who have pleural effusion are more severely and systemically infected, and may have a more protracted disease course. We hypothesized that the host immunological response determines the severity of adenoviral infection. Methods This retrospective case series study included patients diagnosed with severe lower respiratory tract infections at the National Cheng Kung University Hospital in southern Taiwan between December 2010 and October 2011. The main inclusion criteria were 1) presence of multifocal patchy infiltrates, lobar consolidation or reticular interstitial opacities in chest X-rays, and 2) presence of adenovirus isolated from respiratory specimens. All patients had adenovirus isolated from respiratory specimens, and were negative for other viruses. Pleural effusion was confirmed in all patients using chest echography. Clinical features and laboratory data were compared in patients with (n = 12) and without (n = 15) parapneumonic effusion. Results Presence of parapneumonic effusion was significantly associated with a longer febrile duration, more complicated clinical management, and a greater risk of extrapulmonary involvement, notably hepatitis. Patients without pleural effusion had significantly higher numbers of WBCs, platelets, and absolute segment cell counts (ASCs) compared to patients with pleural effusion (all p < 0.05). Patients without pleural effusion had significantly higher counts of CD4+, CD8+, and CD20+ T cells (all p < 0.05) compared to patients with pleural effusion. Conclusion Our data indicated that presence of parapneumonic effusion in adenoviral pneumonia was associated with longer febrile duration, more complicated clinical management, a greater risk of hepatitis, and suppression of host cellular immunity. Further prospective, large-scale studies are needed to validate our results.
Collapse
Affiliation(s)
- Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, North Dist., Tainan, 70403, Taiwan
| | - Shih-Min Wang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzong-Shiann Ho
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, North Dist., Tainan, 70403, Taiwan. .,Center of Infectious Disease and Signaling Research, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center for Infection Control, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
37
|
MITA/STING and Its Alternative Splicing Isoform MRP Restrict Hepatitis B Virus Replication. PLoS One 2017; 12:e0169701. [PMID: 28056087 PMCID: PMC5215812 DOI: 10.1371/journal.pone.0169701] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023] Open
Abstract
An efficient clearance of hepatitis B virus (HBV) requires the coordinated work of both the innate and adaptive immune responses. MITA/STING, an adapter protein of the innate immune signaling pathways, plays a key role in regulating innate and adaptive immune responses to DNA virus infection. Previously, we identified an alternatively spliced isoform of MITA/STING, called MITA-related protein (MRP), and found that MRP could specifically block MITA-mediated interferon (IFN) induction while retaining the ability to activate NF-κB. Here, we asked whether MITA/STING and MRP were able to control the HBV replication. Both MITA/STING and MRP significantly inhibited HBV replication in vitro. MITA overexpression stimulated IRF3-IFN pathway; while MRP overexpression activated NF-κB pathway, suggesting these two isoforms may inhibit HBV replication through different ways. Using a hydrodynamic injection (HI) mouse model, we found that HBV replication was reduced following MITA/STING and MRP expression vectors in mice and was enhanced by the knockout of MITA/STING (MITA/STING-/-). The HBV specific humoral and CD8+ T cell responses were impaired in MITA/STING deficient mice, suggesting the participation of MITA/STING in the initiation of host adaptive immune responses. In summary, our data suggest that MITA/STING and MRP contribute to HBV control via modulation of the innate and adaptive responses.
Collapse
|
38
|
Rezza G, Ippolito G. Syrian Hamsters as a Small Animal Model for Emerging Infectious Diseases: Advances in Immunologic Methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 972:87-101. [PMID: 27722960 PMCID: PMC7121384 DOI: 10.1007/5584_2016_135] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The use of small animal models for the study of infectious disease is critical for understanding disease progression and for developing prophylactic and therapeutic treatment options. For many diseases, Syrian golden hamsters have emerged as an ideal animal model due to their low cost, small size, ease of handling, and ability to accurately reflect disease progression in humans. Despite the increasing use and popularity of hamsters, there remains a lack of available reagents for studying hamster immune responses. Without suitable reagents for assessing immune responses, researchers are left to examine clinical signs and disease pathology. This becomes an issue for the development of vaccine and treatment options where characterizing the type of immune response generated is critical for understanding protection from disease. Despite the relative lack of reagents for use in hamsters, significant advances have been made recently with several hamster specific immunologic methods being developed. Here we discuss the progress of this development, with focus on classical methods used as well as more recent molecular methods. We outline what methods are currently available for use in hamsters and what is readily used as well as what limitations still exist and future perspectives of reagent and assay development for hamsters. This will provide valuable information to researchers who are deciding whether to use hamsters as an animal model.
Collapse
|
39
|
Banadyga L, Dolan MA, Ebihara H. Rodent-Adapted Filoviruses and the Molecular Basis of Pathogenesis. J Mol Biol 2016; 428:3449-66. [PMID: 27189922 PMCID: PMC5010511 DOI: 10.1016/j.jmb.2016.05.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/04/2016] [Accepted: 05/06/2016] [Indexed: 11/29/2022]
Abstract
Ebola, Marburg, and Ravn viruses, all filoviruses, are the causative agents of severe hemorrhagic fever. Much of what we understand about the pathogenesis of filovirus disease is derived from work with animal models, including nonhuman primates, which are considered the "gold standard" filovirus model since they faithfully recapitulate the clinical hallmarks of filovirus disease. However, rodent models, including the mouse, guinea pig, and hamster, also exist for Ebola, Marburg, and Ravn viruses, and although they may not reproduce all the clinical signs of filovirus disease, thanks to their relative ease of use and low cost, they are often the first choice for initial descriptions of virus pathogenesis and evaluation of antiviral prophylactics and therapeutics. Since filoviruses do not cause significant disease in adult, immunocompetent rodents, these models rely on "rodent-adapted" viruses that have been passaged several times through their host until virulence and lethality are achieved. In the process of adaptation, the viruses acquire numerous nucleotide/amino acid mutations that contribute to virulence in their rodent host. Interestingly, virus protein 24 (VP24) and nucleoprotein (NP) appear to be major virulence factors for ebolaviruses in rodents, whereas VP40 appears to be the major virulence factor for marburgviruses. By characterizing these mutations and understanding the molecular mechanisms that lead to the acquisition of virulence, we can gain better insight into the pathogenic processes that underlie filovirus disease in humans. These processes, and the viral and/or cellular proteins that contribute to them, will make attractive targets for the development of novel therapeutics and counter-measures.
Collapse
Affiliation(s)
- Logan Banadyga
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Michael A Dolan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hideki Ebihara
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA.
| |
Collapse
|
40
|
Diminished Innate Antiviral Response to Adenovirus Vectors in cGAS/STING-Deficient Mice Minimally Impacts Adaptive Immunity. J Virol 2016; 90:5915-27. [PMID: 27076643 DOI: 10.1128/jvi.00500-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/09/2016] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Infection by adenovirus, a nonenveloped DNA virus, induces antiviral innate and adaptive immune responses. Studies of transformed human and murine cell lines using short hairpin RNA (shRNA) knockdown strategies identified cyclic guanine adenine synthase (cGAS) as a pattern recognition receptor (PRR) that contributes to the antiadenovirus response. Here we demonstrate how the cGAS/STING cascade influences the antiviral innate and adaptive immune responses in a murine knockout model. Using knockout bone marrow-derived dendritic cells (BMDCs) and bone marrow-derived macrophages (BMMOs), we determined that cGAS and STING are essential to the induction of the antiadenovirus response in these antigen-presenting cells (APCs) in vitro We next determined how the cGAS/STING cascade impacts the antiviral response following systemic administration of a recombinant adenovirus type 5 vector (rAd5V). Infection of cGAS(-/-) and STING(-/-) mice results in a compromised early antiviral innate response compared to that in wild-type (WT) controls: significantly lower levels of beta interferon (IFN-β) secretion, low levels of proinflammatory chemokine induction, and reduced levels of antiviral transcript induction in hepatic tissue. At 24 h postinfection, levels of viral DNA and reporter gene expression in the liver were similar in all strains. At 28 days postinfection, clearance of infected hepatocytes in cGAS or STING knockout mice was comparable to that in WT C57BL/6 mice. Levels of neutralizing anti-Ad5V antibody were modestly reduced in infected cGAS mice. These data support a dominant role for the cGAS/STING cascade in the early innate antiviral inflammatory response to adenovirus vectors. However, loss of the cGAS/STING pathway did not affect viral clearance, and cGAS deficiency had a modest influence on the magnitude of the antiviral humoral immune response to adenovirus infections. IMPORTANCE The detection of viral infection by host sentinel immune cells contributes to the activation of a complex and varied antiviral innate and adaptive immune response, which limits virus replication, spread, and susceptibility to infection. In this study, we have characterized how the cGAS/STING DNA-sensing cascade contributes to early detection of adenovirus infections. cGAS influences APC activation and early innate antiviral inflammatory immune responses, but adaptive immune pathways associated with virus clearance and anti-Ad antibody production were minimally influenced by the loss of the cGAS PRR signaling cascade.
Collapse
|
41
|
Julander JG. Animal models of yellow fever and their application in clinical research. Curr Opin Virol 2016; 18:64-9. [PMID: 27093699 DOI: 10.1016/j.coviro.2016.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/30/2016] [Indexed: 11/18/2022]
Abstract
Yellow fever virus (YFV) is an arbovirus that causes significant human morbidity and mortality. This virus has been studied intensively over the past century, although there are still no treatment options for those who become infected. Periodic and unpredictable yellow fever (YF) outbreaks in Africa and South America continue to occur and underscore the ongoing need to further understand this viral disease and to develop additional countermeasures to prevent or treat cases of illness. The use of animal models of YF is critical to accomplishing this goal. There are several animal models of YF that replicate various aspects of clinical disease and have provided insight into pathogenic mechanisms of the virus. These typically include mice, hamsters and non-human primates (NHP). The utilities and shortcomings of the available animal models of YF are discussed. Information on recent discoveries that have been made in the field of YFV research is also included as well as important future directions in further ameliorating the morbidity and mortality that occur as a result of YFV infection. It is anticipated that these model systems will help facilitate further improvements in the understanding of this virus and in furthering countermeasures to prevent or treat infections.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, United States.
| |
Collapse
|
42
|
The Persistent Mystery of Adenovirus Persistence. Trends Microbiol 2016; 24:323-324. [PMID: 26916790 DOI: 10.1016/j.tim.2016.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 11/22/2022]
|
43
|
Zheng Y, Stamminger T, Hearing P. E2F/Rb Family Proteins Mediate Interferon Induced Repression of Adenovirus Immediate Early Transcription to Promote Persistent Viral Infection. PLoS Pathog 2016; 12:e1005415. [PMID: 26809031 PMCID: PMC4726734 DOI: 10.1371/journal.ppat.1005415] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/04/2016] [Indexed: 01/06/2023] Open
Abstract
Interferons (IFNs) are cytokines that have pleiotropic effects and play important roles in innate and adaptive immunity. IFNs have broad antiviral properties and function by different mechanisms. IFNs fail to inhibit wild-type Adenovirus (Ad) replication in established cancer cell lines. In this study, we analyzed the effects of IFNs on Ad replication in normal human cells. Our data demonstrate that both IFNα and IFNγ blocked wild-type Ad5 replication in primary human bronchial epithelial cells (NHBEC) and TERT-immortalized normal human diploid fibroblasts (HDF-TERT). IFNs inhibited the replication of divergent adenoviruses. The inhibition of Ad5 replication by IFNα and IFNγ is the consequence of repression of transcription of the E1A immediate early gene product. Both IFNα and IFNγ impede the association of the transactivator GABP with the E1A enhancer region during the early phase of infection. The repression of E1A expression by IFNs requires a conserved E2F binding site in the E1A enhancer, and IFNs increased the enrichment of the E2F-associated pocket proteins, Rb and p107, at the E1A enhancer in vivo. PD0332991 (Pabociclib), a specific CDK4/6 inhibitor, dephosphoryles pocket proteins to promote their interaction with E2Fs and inhibited wild-type Ad5 replication dependent on the conserved E2F binding site. Consistent with this result, expression of the small E1A oncoprotein, which abrogates E2F/pocket protein interactions, rescued Ad replication in the presence of IFNα or IFNγ. Finally, we established a persistent Ad infection model in vitro and demonstrated that IFNγ suppresses productive Ad replication in a manner dependent on the E2F binding site in the E1A enhancer. This is the first study that probes the molecular basis of persistent adenovirus infection and reveals a novel mechanism by which adenoviruses utilize IFN signaling to suppress lytic virus replication and to promote persistent infection. Interferons play important roles in both innate and adaptive immunity, and have broad antiviral properties. We demonstrate that type I (IFNα) and type II (IFNγ) IFNs inhibit the replication of divergent adenoviruses via an evolutionally conserved E2F binding site. IFNs augment the association of the tumor suppressors Rb and p107 with the E1A enhancer region in vivo to repress viral immediate early transcription. By comparing the properties of wild type and E2F site mutant viruses, we show that the IFN–E2F/Rb axis is critical for restriction of adenovirus replication to promote persistent viral infection. Relief of E2F/Rb repression counteracts IFN signaling whereas enforcement of E2F/Rb interaction mimics IFN signaling. These results reveal a novel mechanism by which adenoviruses utilize IFN signaling to suppress lytic virus replication and promote persistent infection.
Collapse
Affiliation(s)
- Yueting Zheng
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Patrick Hearing
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
Toth K, Lee SR, Ying B, Spencer JF, Tollefson AE, Sagartz JE, Kong IK, Wang Z, Wold WSM. Correction: STAT2 Knockout Syrian Hamsters Support Enhanced Replication and Pathogenicity of Human Adenovirus, Revealing an Important Role of Type I Interferon Response in Viral Control. PLoS Pathog 2016; 12:e1005392. [PMID: 26745633 PMCID: PMC4706453 DOI: 10.1371/journal.ppat.1005392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
[This corrects the article DOI: 10.1371/journal.ppat.1005084.].
Collapse
|
45
|
Rhein BA, Powers LS, Rogers K, Anantpadma M, Singh BK, Sakurai Y, Bair T, Miller-Hunt C, Sinn P, Davey RA, Monick MM, Maury W. Interferon-γ Inhibits Ebola Virus Infection. PLoS Pathog 2015; 11:e1005263. [PMID: 26562011 PMCID: PMC4643030 DOI: 10.1371/journal.ppat.1005263] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022] Open
Abstract
Ebola virus outbreaks, such as the 2014 Makona epidemic in West Africa, are episodic and deadly. Filovirus antivirals are currently not clinically available. Our findings suggest interferon gamma, an FDA-approved drug, may serve as a novel and effective prophylactic or treatment option. Using mouse-adapted Ebola virus, we found that murine interferon gamma administered 24 hours before or after infection robustly protects lethally-challenged mice and reduces morbidity and serum viral titers. Furthermore, we demonstrated that interferon gamma profoundly inhibits Ebola virus infection of macrophages, an early cellular target of infection. As early as six hours following in vitro infection, Ebola virus RNA levels in interferon gamma-treated macrophages were lower than in infected, untreated cells. Addition of the protein synthesis inhibitor, cycloheximide, to interferon gamma-treated macrophages did not further reduce viral RNA levels, suggesting that interferon gamma blocks life cycle events that require protein synthesis such as virus replication. Microarray studies with interferon gamma-treated human macrophages identified more than 160 interferon-stimulated genes. Ectopic expression of a select group of these genes inhibited Ebola virus infection. These studies provide new potential avenues for antiviral targeting as these genes that have not previously appreciated to inhibit negative strand RNA viruses and specifically Ebola virus infection. As treatment of interferon gamma robustly protects mice from lethal Ebola virus infection, we propose that interferon gamma should be further evaluated for its efficacy as a prophylactic and/or therapeutic strategy against filoviruses. Use of this FDA-approved drug could rapidly be deployed during future outbreaks. Filovirus outbreaks occur sporadically, but with increasing frequency. With no current approved filovirus therapeutics, the 2014 Makona Ebola virus epidemic in Guinea, Sierra Leone and Liberia emphasizes the need for effective treatments against this highly pathogenic family of viruses. The use of this FDA-approved drug to inhibit Ebola virus infection would allow rapid implementation of a novel antiviral therapy for future crises. Interferon gamma elicits an antiviral state in antigen-presenting cells and stimulates cellular immune responses. We demonstrate that interferon gamma profoundly inhibits Ebola virus infection of macrophages, which are early cellular targets of Ebola virus. We also identify novel interferon gamma-stimulated genes in human macrophage populations that have not been previously appreciated to inhibit filoviruses or other negative strand RNA viruses. Finally and most importantly, we show that interferon gamma given 24 hours prior to or after virus infection protects mice from lethal Ebola virus challenge, suggesting that this drug may serve as an effective prophylactic and/or therapeutic strategy against this deadly virus.
Collapse
Affiliation(s)
- Bethany A. Rhein
- Department of Microbiology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Linda S. Powers
- Department of Internal Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Kai Rogers
- Department of Microbiology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Brajesh K. Singh
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Yasuteru Sakurai
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Thomas Bair
- Iowa Institute for Human Genetics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Catherine Miller-Hunt
- Department of Microbiology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Patrick Sinn
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Robert A. Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Martha M. Monick
- Department of Internal Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Wendy Maury
- Department of Microbiology, The University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
46
|
Low-Level Expression of the E1B 20-Kilodalton Protein by Adenovirus 14p1 Enhances Viral Immunopathogenesis. J Virol 2015; 90:497-505. [PMID: 26491152 DOI: 10.1128/jvi.01790-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/14/2015] [Indexed: 02/07/2023] Open
Abstract
Adenovirus 14p1 (Ad14p1) is an emergent variant of Ad serotype 14 (Ad14) that has caused increased severity of respiratory illnesses during globally distributed outbreaks, including cases of acute respiratory distress syndrome and death. We found that human cell infection with Ad14p1 results in markedly decreased expression of the E1B 20-kilodalton (20K) protein compared to that with infection with wild-type (wt) Ad14. This reduced Ad14p1 E1B 20K expression caused a loss-of-function phenotype of Ad-infected cell corpses that, in contrast to cells infected with wt Ad14, either failed to repress or increased NF-κB-dependent, proinflammatory cytokine responses of responder human alveolar macrophages. A small-animal model of Ad14-induced lung infection was used to test the translational relevance of these in vitro observations. Intratracheal infection of Syrian hamsters with Ad14p1 caused a marked, patchy bronchopneumonia, whereas hamster infection with wt Ad14 caused minimal peribronchial inflammation. These results suggest that this difference in E1B 20K gene expression during Ad14p1 infection and its modulating effect on the interactions between Ad14-infected cells and the host innate immune response could explain the increased immunopathogenic potential and associated increase in clinical illness in some people infected with the Ad14p1 outbreak strain.IMPORTANCE We previously reported that Ad-infected human cells exhibit E1B 19K-dependent repression of virally induced, NF-κB-dependent macrophage cytokine responses (J. R. Radke, F. Grigera, D. S. Ucker, and J. L. Cook, J Virol 88:2658-2669, 2014, http://dx.doi.org/10.1128/JVI.02372-13). The more virulent, emergent strain of Ad14, Ad14p1, causes increased cytopathology in vitro, which suggested a possible E1B 20K defect. Whether there is a linkage between these observations was unknown. We show that there is markedly reduced expression of E1B 20K in Ad14p1-infected human cells and that this causes an increased proinflammatory cytokine response of human alveolar macrophages and more severe inflammatory lung disease in infected hamsters. This is the first evidence of a clinical relevance of differential expression of the small Ad E1B gene product. The results suggest that there is a low, critical threshold of E1B 19/20K expression that is needed for viral replication and infection transmission but that a higher level of E1B 19/20K expression is required for the usual repression and control of the Ad-triggered host innate immune response.
Collapse
|