1
|
Lee YZ, Zhang YN, Newby ML, Ward G, Gomes KB, Auclair S, DesRoberts C, Allen JD, Ward AB, Stanfield RL, He L, Crispin M, Wilson IA, Zhu J. Rational design of next-generation filovirus vaccines with glycoprotein stabilization, nanoparticle display, and glycan modification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.641072. [PMID: 40060701 PMCID: PMC11888476 DOI: 10.1101/2025.03.02.641072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Filoviruses pose a significant threat to human health with frequent outbreaks and high mortality. Although two vector-based vaccines are available for Ebola virus, a broadly protective filovirus vaccine remains elusive. In this study, we evaluate a general strategy for stabilizing glycoprotein (GP) structures of Ebola, Sudan, and Bundibugyo ebolaviruses and Ravn marburgvirus. A 3.2 Å-resolution crystal structure provides atomic details for the redesigned Ebola virus GP, and cryo-electron microscopy reveals how a pan-ebolavirus neutralizing antibody targets a conserved site on the Sudan virus GP (3.13 Å-resolution), in addition to a low-resolution model of antibody-bound Ravn virus GP. A self-assembling protein nanoparticle (SApNP), I3-01v9, is redesigned at the N-terminus to allow the optimal surface display of filovirus GP trimers. Following detailed in vitro characterization, the lymph node dynamics of Sudan virus GP and GP-presenting SApNPs are investigated in a mouse model. Compared with soluble GP trimer, SApNPs show ~112 times longer retention in lymph node follicles, up-to-28 times greater presentation on follicular dendritic cell dendrites, and up-to-3 times stronger germinal center reactions. Functional antibody responses induced by filovirus GP trimers and SApNPs bearing wildtype and modified glycans are assessed in mice. Our study provides a foundation for next-generation filovirus vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maddy L. Newby
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Connor DesRoberts
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Joel D. Allen
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robyn L. Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Max Crispin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Uvax Bio, LLC, Newark, DE 19702, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
2
|
Wang LL, Seravalli J, Eaton B, Liu Y, Holbrook MR, Lan WJ, Xiang SH. Identification of Filovirus Entry Inhibitors from Marine Fungus-Derived Indole Alkaloids. Mar Drugs 2025; 23:23. [PMID: 39852525 PMCID: PMC11766795 DOI: 10.3390/md23010023] [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: 12/04/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Filoviruses, mainly consisting of the two genera of Ebolavirus and Marburgvirus, are enveloped negative-strand RNA viruses that can infect humans to cause severe hemorrhagic fevers and outbreaks with high mortality rates. However, we still do not have effective medicines for treating these diseases. To search for effective drugs, we have identified three marine indole alkaloids that exhibit potent activities against filovirus infection. Thus, it is suggested that marine indole alkaloids can be a valuable compound source for filovirus drug screening and development. Since marine indole alkaloids comprise a large diverse group of secondary metabolites, their biological properties would be helpful for pharmaceutical drug development to treat various filovirus infections.
Collapse
Affiliation(s)
- Leah Liu Wang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Javier Seravalli
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Brett Eaton
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Yi Liu
- Holland Computing Center, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Michael R. Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shi-Hua Xiang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
3
|
Mohd OB, Sawaqed S, Kundu M, Ghannam RA, Mohd AB, AlSamhori JF, Musallam OK, Altiti A, Hasan H, Khaity A. The Development of Ebola Virus Outbreaks: A Review of Epidemiological Trends, Clinical Features, and Treatment Advances. Cureus 2024; 16:e74078. [PMID: 39712786 PMCID: PMC11660724 DOI: 10.7759/cureus.74078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
The Ebola virus, a filovirus that causes human Ebola virus disease (EVD), has caused multiple epidemics in the African continent for about 50 years. Wild animals were the source from which the virus was transmitted to humans, and it spread among people through direct contact. The majority of Ebola outbreaks occurred in African nations, particularly in Sudan, the Democratic Republic of the Congo (DRC), Uganda, and Gabon. Although EVD is a lethal disease, it has posed a challenge to human efforts to comprehend its etiology, epidemiology, pathophysiology, pathogenesis, and the best methods for treatment and prevention. This review aims to present the history of Ebola epidemics in Africa, each subtype that caused an outbreak, the development of therapies and vaccines, and the significance of travel regulations.
Collapse
Affiliation(s)
- Omar B Mohd
- Faculty of Medicine, Hashemite University, Zarqa, JOR
| | - Seri Sawaqed
- Faculty of Medicine, Hashemite University, Zarqa, JOR
| | - Mrinmoy Kundu
- College of Medicine, Institute of Medical Sciences and SUM Hospital, Bhubaneswar, IND
| | | | - Ahmed B Mohd
- Faculty of Medicine, Hashemite University, Zarqa, JOR
| | | | | | | | | | | |
Collapse
|
4
|
Hewson R. Understanding Viral Haemorrhagic Fevers: Virus Diversity, Vector Ecology, and Public Health Strategies. Pathogens 2024; 13:909. [PMID: 39452780 PMCID: PMC11510013 DOI: 10.3390/pathogens13100909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Viral haemorrhagic fevers encompass a diverse group of severe, often life-threatening illnesses caused by viruses from multiple families, including Arenaviridae, Filoviridae, Flaviviridae, Hantaviridae, Nairoviridae, Peribunyaviridae, and Phenuiviridae. Characterised by fever and haemorrhagic symptoms, these diseases challenge public health systems by overwhelming healthcare facilities, complicating diagnostic processes, and requiring extensive resources for containment and treatment, especially in resource-limited settings. This discussion explores the intricate relationships between VHFs and their transmission vectors-both animal and arthropod-and examines the impact of ecological and geographic factors on disease spread. The primary transmission of VHFs typically occurs through direct contact with infected animals or via bites from haematophagous arthropods, facilitating zoonotic and, at times, human-to-human transmission. With an emphasis on the role of diverse wildlife, domesticated animals, and vectors such as mosquitoes and ticks in the epidemiology of VHFs, there is a recognised need for robust surveillance and strategic public health responses to manage outbreaks. This review discusses the necessity of interdisciplinary approaches that integrate virology, ecology, and public health to enhance diagnostic capabilities, develop vaccines and antivirals, and improve outbreak interventions. Exploring the ecological and biological dynamics of VHFs will help bolster a deeper understanding of these emerging viruses and underpin preparation for future outbreaks. The importance of enhanced global cooperation, continuous research, and collaboration to mitigate the public health threats posed by these complex infections is a central theme, serving as a foundational strategy to reinforce worldwide preparedness and response efforts. Future directions include addressing gaps in vaccine development and tailoring public health strategies to the unique challenges of managing VHFs, such as the rapid mutation rates of viruses, the need for cold chain logistics for vaccine distribution, and socio-economic barriers to healthcare access, in order to ensure readiness for and effective response to emerging threats worldwide.
Collapse
Affiliation(s)
- Roger Hewson
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
- Virus Reference & Research (Special Pathogens), WHO—Collaborating Centre, Salisbury SP4 0JG, UK
- UK—Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| |
Collapse
|
5
|
Hackbart M, López CB. Characterization of non-standard viral genomes during arenavirus infections identifies prominent S RNA intergenic region deletions. mBio 2024; 15:e0161224. [PMID: 39258905 PMCID: PMC11481572 DOI: 10.1128/mbio.01612-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Arenaviruses, a family of negative-sense RNA viruses spread by rodents, are a leading cause of severe hemorrhagic fever in humans. Due to a paucity of antivirals and vaccines for arenaviruses, there is a need to identify new mechanisms for interfering with arenavirus replication. In several negative-sense RNA viruses, natural viral interference results from the production of non-standard viral genomes (nsVGs) that activate the innate immune system and/or compete for essential viral products. Although it is well established that arenaviruses produce strong interfering activities, it is unknown if they produce interfering nsVGs. Here, we show that arenaviruses produce deletions within the intergenic region of their small (S) RNA genome, and these deletions inhibit viral glycoprotein production during minigenome replication. S RNA deletions are more abundant when arenaviruses are grown in high-interfering conditions and are associated with reduced viral replication. Overall, we found that arenaviruses produce internal deletions within the S RNA intergenic region that are capable of decreasing glycoprotein production. These natural arenavirus interfering molecules provide a new target for the generation of therapeutics against arenaviruses.IMPORTANCEArenaviruses are hemorrhagic fever-causing pathogens that infect millions of people a year. There are currently no approved antivirals that target arenaviruses, and understanding natural mechanisms that inhibit arenavirus replication is crucial for the development of effective therapeutics. Here, we identified multiple deletions within arenavirus genomes that remove major replicative elements of the viral genomes. We show that deletions that remove the intergenic region of the viral genome can prevent viral protein production. These deletions were found in all arenaviruses tested in this study representing a mechanism that could be harnessed for the development of antivirals that broadly target the arenavirus family.
Collapse
Affiliation(s)
- Matthew Hackbart
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carolina B. López
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Creppy JR, Delache B, Lemaitre J, Horvat B, Vecellio L, Ducancel F. Administration of airborne pathogens in non-human primates. Inhal Toxicol 2024; 36:475-500. [PMID: 39388247 DOI: 10.1080/08958378.2024.2412685] [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: 10/16/2023] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Airborne pathogen scan penetrate in human respiratory tract and can cause illness. The use of animal models to predict aerosol deposition and study respiratory disease pathophysiology is therefore important for research and a prerequisite to test and study the mechanism of action of treatment. NHPs are relevant animal species for inhalation studies because of their similarities with humans in terms of anatomical structure, respiratory parameters and immune system. MATERIALS AND METHODS The aim of this review is to provide an overview of the state of the art of pathogen aerosol studies performed in non-human primates (NHPs). Herein, we present and discuss the deposition of aerosolized bacteria and viruses. In this review, we present important advantages of using NHPs as model for inhalation studies. RESULTS We demonstrate that deposition in the respiratory tract is not only a function of aerosol size but also the technique of administration influences the biological activity and site of aerosol deposition. Finally, we observe an influence of a region of pathogen deposition in the respiratory tract on the development of the pathophysiological effect in NHPs. CONCLUSION The wide range of methods used for the delivery of pathogento NHP respiratory airways is associated with varying doses and deposition profiles in the airways.
Collapse
Affiliation(s)
- Justina R Creppy
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Benoit Delache
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université de Lyon, Lyon, France
| | - Laurent Vecellio
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Frédéric Ducancel
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
7
|
Huong NTC, Hai NP, Van Khanh C, Kamel MG, Vinh Chau NV, Truong NT, Vinh NT, Elsheikh R, Makram AM, Elsheikh A, Canh HN, Iqtadar S, Hirayama K, Le Hoa PT, Huy NT. New biomarkers for liver involvement by dengue infection in adult Vietnamese patients: a case-control study. BMC Infect Dis 2024; 24:800. [PMID: 39118006 PMCID: PMC11308448 DOI: 10.1186/s12879-024-09527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/17/2024] [Indexed: 08/10/2024] Open
Abstract
Liver injury with marked elevation of aspartate aminotransferase enzyme (AST) is commonly observed in dengue infection. To understand the pathogenesis of this liver damage, we compared the plasma levels of hepatic specific, centrilobular predominant enzymes (glutamate dehydrogenase, GLDH; glutathione S transferase-α, αGST), periportal enriched 4-hydroxyphenylpyruvate dioxygenase (HPPD), periportal predominant arginase-1 (ARG-1), and other non-specific biomarkers (paraoxonase-1, PON-1) in patients with different outcomes of dengue infection. This hospital-based study enrolled 87 adult dengue patients, stratified into three groups based on plasma AST levels (< 80, 80-400, > 400 U/L) in a 1:1:1 ratio (n = 40, n = 40, n = 40, respectively. The new liver enzymes in the blood samples from the 4th to 6th days of their illness were measured by commercial enzyme-linked immunosorbent assay (ELISA) or colorimetric kits. Based on the diagnosis at discharge days, our patients were classified as 40 (46%) dengue without warning signs (D), 35 (40.2%) dengue with warning signs (DWS), and 11 (12.6%) severe dengue (SD) with either shock (two patients) or AST level over 1000 U/L (nine patients), using the 2009 WHO classification. The group of high AST (> 400 U/L) also had higher ALT, GLDH, ARG-1, and HPPD than the other groups, while the high (> 400 U/L) and moderate (80-400 U/L) AST groups had higher ALT, αGST, ARG-1, and HPPD than the low AST group (< 80 U/L). There was a good correlation between AST, alanine aminotransferase enzyme (ALT), and the new liver biomarkers such as GLDH, αGST, ARG-1, and HPPD. Our findings suggest that dengue-induced liver damage initiates predominantly in the centrilobular area toward the portal area during the dengue progression. Moreover, these new biomarkers should be investigated further to explain the pathogenesis of dengue and to validate their prognostic utility.
Collapse
Affiliation(s)
- Nguyen Thi Cam Huong
- Department of Infectious Diseases, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Phuong Hai
- Department of Infectious Diseases, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Department of Infectious Diseases, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Chau Van Khanh
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
- Institute of Malariology, Parasitology, and Entomology Quy Nhon, Quy Nhon, Vietnam
| | - Mohamed Gomaa Kamel
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
- Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Colchester, UK
| | | | | | | | - Randa Elsheikh
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Abdelrahman M Makram
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.
- Department of Anesthesia and Intensive Care Medicine, October 6 University, Giza, Egypt.
| | - Aya Elsheikh
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
- Faculty of Medicine, Mansoura Manchester University, Mansoura, Egypt
| | - Hiep Nguyen Canh
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
- Center of Pathology and Cytopathology, Bach Mai Hospital, Hanoi, Vietnam
| | - Somia Iqtadar
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Kenji Hirayama
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Pham Thi Le Hoa
- Department of Infectious Diseases, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam.
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam.
| | - Nguyen Tien Huy
- School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
- Online Research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam.
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam.
| |
Collapse
|
8
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [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: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
9
|
Xia Y, Jia B, Chen Y, Wang S, Xu X. Clinical value of coagulation parameters in predicting the severity of severe fever with thrombocytopenia syndrome. Front Microbiol 2024; 15:1335664. [PMID: 38633697 PMCID: PMC11021696 DOI: 10.3389/fmicb.2024.1335664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a novel bunyavirus infection with a high lethality rate. The purpose of this study was to investigate the changes in coagulation parameters in patients with SFTS, aiming to provide clinical evidence for early diagnosis, treatment, and disease analysis. Methods A total of 40 patients with SFTS attended from April 1, 2020 to May 21, 2022 in Nanjing Drum Tower Hospital were selected and grouped according to the duration of the disease, mild and severe disease, cure and death, with 50 healthy physical examiners as controls, and the risk of severe and death disease was predicted using ROC curves. Results Comparison between the healthy, mild and severe groups revealed that PT, INR, APTT, TT, D-D and vWF levels were higher than those in the healthy control group, and FII, FIX, FX, FXI, FXII, PC and PS levels were lower than those in the healthy control group, the differences were statistically significant (p < 0.05). Comparing the results of SFTS patients with different course times, the results of Fib, FV, FVII, FVIII, FIX, FX, FXI were statistically significant (p < 0.05). Among the survived and deceased patients, the PT, INR, DD and PS results of the deceased patients were higher than those of the survived patients, and the FVIII, FIX, FXI, FXII and PC were lower than those of the survived patients. The area under the ROC curve showed that D-D had higher predictive ability for the risk of severe disease (AUROC 0.93, sensitivity and specificity at a Cut-off value of 1.50 mg/L were 90.0 and 86.5%, respectively) and the risk of death occurring (AUROC 0.84, sensitivity and specificity at a Cut-off value of 3.39 mg/L were 87.5 and 80.0%, respectively). Discussion The monitoring of the coagulation parameters in patients with SFTS is great significance for identifying the severity and death of the patient's condition, and it is of great clinical value to provide early attention, timely intervention and maximum reduction of the mortality rate for patients at risk of severe disease.
Collapse
Affiliation(s)
- Yanyan Xia
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bei Jia
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuxin Chen
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Sen Wang
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejing Xu
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Ali Z, Cardoza JV, Basak S, Narsaria U, Bhattacharjee S, G UM, Isaac SP, Franca TCC, LaPlante SR, George SS. A Multi-epitope Vaccine Candidate Against Bolivian Hemorrhagic fever Caused by Machupo Virus. Appl Biochem Biotechnol 2024; 196:2137-2160. [PMID: 37479961 DOI: 10.1007/s12010-023-04604-9] [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] [Accepted: 06/19/2023] [Indexed: 07/23/2023]
Abstract
Bolivian hemorrhagic fever (BHF) caused by Machupo virus (MACV) is a New World arenavirus having a reported mortality rate of 25-35%. The BHF starts with fever, followed by headache, and nausea which rapidly progresses to severe hemorrhagic phase within 7 days of disease onset. One of the key promoters for MACV viral entry into the cell followed by viral propagation is performed by the viral glycoprotein (GPC). GPC is post-transcriptionally cleaved into GP1, GP2 and a signal peptide. These proteins all take part in the viral infection in host body. Therefore, GPC protein is an ideal target for developing therapeutics against MACV infection. In this study, GPC protein was considered to design a multi-epitope, multivalent vaccine containing antigenic and immunogenic CTL and HTL epitopes. Different structural validations and physicochemical properties were analysed to validate the vaccine. Docking and molecular dynamics simulations were conducted to understand the interactions of the vaccine with various immune receptors. Finally, the vaccine was codon optimised in silico and along with which immune simulation studies was performed in order to evaluate the vaccine's effectiveness in triggering an efficacious immune response against MACV.
Collapse
Affiliation(s)
- Zeeshan Ali
- Krupanidhi College of Physiotherapy, Bangalore, Karnataka, 560035, India
| | | | | | | | | | | | - Samuel Paul Isaac
- Krupanidhi College of Physiotherapy, Bangalore, Karnataka, 560035, India
| | - Tanos C C Franca
- Military Institute of Engineering, Rio de Janerio, Brazil
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université de Québec, Laval, Québec, H7V 1B7, Canada
- University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Steven R LaPlante
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université de Québec, Laval, Québec, H7V 1B7, Canada
| | - Sudhan S George
- Krupanidhi College of Physiotherapy, Bangalore, Karnataka, 560035, India.
| |
Collapse
|
11
|
Cruz-Holguín VJ, González-García LD, Velázquez-Cervantes MA, Arévalo-Romero H, De Jesús-González LA, Helguera-Repetto AC, León-Reyes G, Salazar MI, Cedillo-Barrón L, León-Juárez M. Collateral Damage in the Placenta during Viral Infection in Pregnancy: A Possible Mechanism for Vertical Transmission and an Adverse Pregnancy Outcome. Diseases 2024; 12:59. [PMID: 38534983 PMCID: PMC10969698 DOI: 10.3390/diseases12030059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 11/11/2024] Open
Abstract
In mammals, the placenta is a connection between a mother and a new developing organism. This tissue has a protective function against some microorganisms, transports nutrients, and exchanges gases and excretory substances between the mother and the fetus. Placental tissue is mainly composed of chorionic villi functional units called trophoblasts (cytotrophoblasts, the syncytiotrophoblast, and extravillous trophoblasts). However, some viruses have developed mechanisms that help them invade the placenta, causing various conditions such as necrosis, poor perfusion, and membrane rupture which, in turn, can impact the development of the fetus and put the mother's health at risk. In this study, we collected the most relevant information about viral infection during pregnancy which can affect both the mother and the fetus, leading to an increase in the probability of vertical transmission. Knowing these mechanisms could be relevant for new research in the maternal-fetal context and may provide options for new therapeutic targets and biomarkers in fetal prognosis.
Collapse
Affiliation(s)
- Victor Javier Cruz-Holguín
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Luis Didier González-García
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
| | - Manuel Adrián Velázquez-Cervantes
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| | - Haruki Arévalo-Romero
- Laboratorio de Inmunologia y Microbiologia Molecular, Division Academica Multidisciplinaria de Jalpa de Méndez, Jalpa de Mendez 86205, Mexico;
| | | | | | - Guadalupe León-Reyes
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Ma. Isabel Salazar
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
- Laboratorio Nacional de Vacunología y Virus Tropicales (LNVyVT), Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Moisés León-Juárez
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| |
Collapse
|
12
|
Gupta A, Yadav K, Yadav A, Ahmad R, Srivastava A, Kumar D, Khan MA, Dwivedi UN. Mannose-specific plant and microbial lectins as antiviral agents: A review. Glycoconj J 2024; 41:1-33. [PMID: 38244136 DOI: 10.1007/s10719-023-10142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024]
Abstract
Lectins are non-immunological carbohydrate-binding proteins classified on the basis of their structure, origin, and sugar specificity. The binding specificity of such proteins with the surface glycan moiety determines their activity and clinical applications. Thus, lectins hold great potential as diagnostic and drug discovery agents and as novel biopharmaceutical products. In recent years, significant advancements have been made in understanding plant and microbial lectins as therapeutic agents against various viral diseases. Among them, mannose-specific lectins have being proven as promising antiviral agents against a variety of viruses, such as HIV, Influenza, Herpes, Ebola, Hepatitis, Severe Acute Respiratory Syndrome Coronavirus-1 (SARS-CoV-1), Middle Eastern Respiratory Syndrome Coronavirus (MERS-CoV) and most recent Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The binding of mannose-binding lectins (MBLs) from plants and microbes to high-mannose containing N-glycans (which may be simple or complex) of glycoproteins found on the surface of viruses has been found to be highly specific and mainly responsible for their antiviral activity. MBLs target various steps in the viral life cycle, including viral attachment, entry and replication. The present review discusses the brief classification and structure of lectins along with antiviral activity of various mannose-specific lectins from plants and microbial sources and their diagnostic and therapeutic applications against viral diseases.
Collapse
Affiliation(s)
- Ankita Gupta
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Kusum Yadav
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India.
| | - Anurag Yadav
- Department of Microbiology, C.P. College of Agriculture, Sardarkrushinagar Dantiwada Agriculture University, District-Banaskantha, Gujarat, India
| | - Rumana Ahmad
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India.
| | - Aditi Srivastava
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Dileep Kumar
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
- Department of Biotechnology, Khwaja Moinuddin Chishti Language University, Lucknow, Uttar Pradesh, India
| | - Mohammad Amir Khan
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - U N Dwivedi
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
| |
Collapse
|
13
|
Guito JC, Arnold CE, Schuh AJ, Amman BR, Sealy TK, Spengler JR, Harmon JR, Coleman-McCray JD, Sanchez-Lockhart M, Palacios GF, Towner JS, Prescott JB. Peripheral immune responses to filoviruses in a reservoir versus spillover hosts reveal transcriptional correlates of disease. Front Immunol 2024; 14:1306501. [PMID: 38259437 PMCID: PMC10800976 DOI: 10.3389/fimmu.2023.1306501] [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: 10/03/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARV-infected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity.
Collapse
Affiliation(s)
- Jonathan C. Guito
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Catherine E. Arnold
- Biological Defense Research Directorate, Naval Medical Research Center, Frederick, MD, United States
- RD-CBR, Research and Development Directorate, Chemical and Biological Technologies Directorate, Research Center of Excellence, Defense Threat Reduction Agency, Fort Belvoir, VA, United States
| | - Amy J. Schuh
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Brian R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Tara K. Sealy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joann D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Mariano Sanchez-Lockhart
- Center for Genome Sciences, Molecular Biology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Gustavo F. Palacios
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jonathan S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joseph B. Prescott
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
14
|
Hackbart M, López CB. S RNA Intergenic Deletions Drive Viral Interference during Arenavirus Infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.564889. [PMID: 37961573 PMCID: PMC10635013 DOI: 10.1101/2023.10.31.564889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Arenaviruses, a family of negative-sense RNA viruses spread by rodents, are a leading cause of severe hemorrhagic fever in humans. Due to a paucity of antivirals and vaccines for arenaviruses, there is a need to identify new mechanisms for interfering with arenavirus replication. In several negative-sense RNA viruses, natural viral interference results from the production of non-standard viral genomes (nsVGs) that activate the innate immune system and/or compete for essential viral products. Although it is well established that arenaviruses produce strong interfering activities, it is unknown if they produce interfering nsVGs. Here we show that arenaviruses produce deletions within the intergenic region of their Small (S) RNA genome, which prevents the production of viral mRNA and protein. These deletions are more abundant when arenaviruses are grown in high-interfering conditions and are associated with inhibited viral replication. Overall, we found that arenaviruses produce internal deletions within the S RNA intergenic region that are produced by arenaviruses and can block viral replication. These natural arenavirus interfering molecules provide a new target for the generation of antivirals as well as an alternative strategy for producing attenuated arenaviruses for vaccines.
Collapse
Affiliation(s)
- Matthew Hackbart
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St. MO
| | - Carolina B. López
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St. MO
| |
Collapse
|
15
|
Kurosu T, Okuzaki D, Sakai Y, Kadi MA, Phanthanawiboon S, Ami Y, Shimojima M, Yoshikawa T, Fukushi S, Nagata N, Suzuki T, Kamimura D, Murakami M, Ebihara H, Saijo M. Dengue virus infection induces selective expansion of Vγ4 and Vγ6TCR γδ T cells in the small intestine and a cytokine storm driving vascular leakage in mice. PLoS Negl Trop Dis 2023; 17:e0011743. [PMID: 37939119 PMCID: PMC10659169 DOI: 10.1371/journal.pntd.0011743] [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: 07/13/2023] [Revised: 11/20/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
Dengue is a major health problem in tropical and subtropical regions. Some patients develop a severe form of dengue, called dengue hemorrhagic fever, which can be fatal. Severe dengue is associated with a transient increase in vascular permeability. A cytokine storm is thought to be the cause of the vascular leakage. Although there are various research reports on the pathogenic mechanism, the complete pathological process remains poorly understood. We previously reported that dengue virus (DENV) type 3 P12/08 strain caused a lethal systemic infection and severe vascular leakage in interferon (IFN)-α/β and γ receptor knockout mice (IFN-α/β/γRKO mice), and that blockade of TNF-α signaling protected mice. Here, we performed transcriptome analysis of liver and small intestine samples collected chronologically from P12/08-infected IFN-α/β/γRKO mice in the presence/absence of blockade of TNF-α signaling and evaluated the cytokine and effector-level events. Blockade of TNF-α signaling mainly protected the small intestine but not the liver. Infection induced the selective expansion of IL-17A-producing Vγ4 and Vγ6 T cell receptor (TCR) γδ T cells in the small intestine, and IL-17A, together with TNF-α, played a critical role in the transition to severe disease via the induction of inflammatory cytokines such as TNF-α, IL-1β, and particularly the excess production of IL-6. Infection also induced the infiltration of neutrophils, as well as neutrophil collagenase/matrix metalloprotease 8 production. Blockade of IL-17A signaling reduced mortality and suppressed the expression of most of these cytokines, including TNF-α, indicating that IL-17A and TNF-α synergistically enhance cytokine expression. Blockade of IL-17A prevented nuclear translocation of NF-κB p65 in stroma-like cells and epithelial cells in the small intestine but only partially prevented recruitment of immune cells to the small intestine. This study provides an overall picture of the pathogenesis of infection in individual mice at the cytokine and effector levels.
Collapse
Affiliation(s)
- Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mohamad Al Kadi
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, Japan
| | | | - Yasusi Ami
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Kamimura
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Team of Quantumimmunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
- Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Hideki Ebihara
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
16
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
17
|
Gupta V, Acharya S, Keerti A. Common Coagulopathies Associated With COVID-19 Patients. Cureus 2023; 15:e38067. [PMID: 37234147 PMCID: PMC10208414 DOI: 10.7759/cureus.38067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) outbreak, which first appeared in the Chinese province of Hubei city of Wuhan, has been spreading internationally since December 2019. The World Health Organization (WHO) declared the coronavirus illness from 2019 to be a pandemic on March 11, 2020. Patients hospitalised with severe coronavirus or comorbid conditions (like cardiovascular disease and obesity) are linked to a worse prognosis. The rise in D-dimer and its relationship to prognosis are the most often documented aberrations in coagulation/fibrinolysis in COVID-19. However, the D-dimer assessment's utility is not limitless. Since the coagulation/fibrinolytic state might occasionally change over a short period of time, routine exams are also advantageous in understanding the relevance of the inquiry. Both thrombotic and hemorrhagic diseases should be taken into consideration, despite the fact that the pathophysiology of disseminated intravascular coagulation (DIC) linked with coronavirus disease 19 differs significantly from that of septic disseminated intravascular coagulation. Coagulation as well as fibrinolysis indicators are used to make the diagnosis of COVID-19 thrombosis, which encompasses both macro- and micro-thrombosis. Compared to bacterial-sepsis-associated coagulopathy/DIC, COVID-19 has a lower prevalence of prolonged prothrombin time, activated partial thromboplastin time, and decreased antithrombin activity. However, the causes of coagulopathy remain poorly understood. Hypoxia, endothelial injury, dysregulated immunological responses mediated by inflammatory cytokines, and lymphocyte cell death are thought to be implicated. While blood loss tends to be rare, it is uncertain if COVID-19 suffers from thrombosis or whether the current recommendations for regular venous thromboembolic dose are appropriate. It is important to decide on the COVID-19 therapy phases. Antiviral therapy, cytokine storm therapy, and thrombosis therapy are the steps. Future advancements are predicted, such as a therapy that combines heparin and nafamostat.
Collapse
Affiliation(s)
- Vinish Gupta
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Sourya Acharya
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Akshunna Keerti
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| |
Collapse
|
18
|
Taki E, Ghanavati R, Navidifar T, Dashtbin S, Heidary M, Moghadamnia M. Ebanga™: The most recent FDA-approved drug for treating Ebola. Front Pharmacol 2023; 14:1083429. [PMID: 36969842 PMCID: PMC10032372 DOI: 10.3389/fphar.2023.1083429] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/31/2023] [Indexed: 03/29/2023] Open
Abstract
Ebolavirus (EBOV) is a virulent pathogen that causes Ebola virus disease (EVD), which is a life-threatening human condition with a fatality rate of up to 90%. Since the first outbreak in Africa in 1976, several outbreaks and epidemics of EBOV have occurred across the globe. While EVD is recognized as a serious threat to human health and outbreaks occur almost every year, the treatment options for the disease are limited. In designing therapeutic strategies against EBOV infection, viral structural proteins, such as glycoprotein (GP), could be an excellent target for neutralizing the virus. According to the latest research, GP-specific antibodies are the most efficient post-exposure treatments for EVD. Ansuvimab-zykl, i.e., mAb114 (Ebanga™), is a recent FDA-approved human immunoglobulin monoclonal antibody targeting EBOV GP. This review provides a brief overview of the pharmacological effects and safety profile of ansuvimab in clinical trials and provides insights into the precise mechanism of this new drug for treating EVD.
Collapse
Affiliation(s)
- Elahe Taki
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Marjan Moghadamnia
- Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Huang M, Wang T, Huang Y, Wang Y, Wu S, Wang F, Tang G, Wei W, Liu W, Hou H. The clinical and immunological characteristics in fatal severe fever with thrombocytopenia syndrome virus (SFTSV) infection. Clin Immunol 2023; 248:109262. [PMID: 36796470 DOI: 10.1016/j.clim.2023.109262] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
OBJECTIVE This study aimed to make a comprehensive evaluation of peripheral immune profiles for further understanding the immunopathogenesis of severe fever with thrombocytopenia syndrome (SFTS). METHODS Forty-seven patients with SFTS virus infection were included, of which 24 were deceased. The percentages, absolute numbers, phenotype of lymphocyte subsets were detected by flow cytometry. RESULTS In patients with SFTS, the numbers of CD3+T, CD4+T, CD8+T and NKT cells were decreased compared with healthy controls (HCs), accompanied with highly active and exhausted phenotypes for T cells, and overproliferating plasmablasts. High inflammatory status, dysregulation of coagulation and host immune response were more obvious in deceased patients than that of survivors. Higher levels of PCT, IL-6, IL-10, TNF-α, APTT, TT and the occurrence of hemophagocytic lymphohistiocytosis were poor prognostic indicators of SFTS. CONCLUSIONS The evaluation of immunological markers in combination with laboratory tests has critical value for selecting prognostic markers and potential treatment target.
Collapse
Affiliation(s)
- Ming Huang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Huang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiji Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoxing Tang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wei
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Franzoni G, Pedrera M, Sánchez-Cordón PJ. African Swine Fever Virus Infection and Cytokine Response In Vivo: An Update. Viruses 2023; 15:233. [PMID: 36680273 PMCID: PMC9864779 DOI: 10.3390/v15010233] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023] Open
Abstract
African swine fever (ASF) is a hemorrhagic viral disease of domestic pigs and wild suids (all Sus scrofa) caused by the ASF virus (ASFV). The disease is spreading worldwide without control, threatening pig production due to the absence of licensed vaccine or commercially available treatments. A thorough understanding of the immunopathogenic mechanisms behind ASFV infection is required to better fight the disease. Cytokines are small, non-structural proteins, which play a crucial role in many aspects of the immune responses to viruses, including ASFV. Infection with virulent ASFV isolates often results in exacerbated immune responses, with increased levels of serum pro-inflammatory interleukins (IL-1α, IL-1β, IL-6), TNF and chemokines (CCL2, CCL5, CXCL10). Increased levels of IL-1, IL-6 and TNF are often detected in several tissues during acute ASFV infections and associated with lymphoid depletion, hemorrhages and oedemas. IL-1Ra is frequently released during ASFV infection to block further IL-1 activity, with its implication in ASFV immunopathology having been suggested. Increased levels of IFN-α and of the anti-inflammatory IL-10 seem to be negatively correlated with animal survival, whereas some correlation between virus-specific IFN-γ-producing cells and protection has been suggested in different studies where different vaccine candidates were tested, although future works should elucidate whether IFN-γ release by specific cell types is related to protection or disease development.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy
| | - Miriam Pedrera
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos, 28130 Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos, 28130 Madrid, Spain
| |
Collapse
|
21
|
Zong L, Yang F, Liu S, Gao Y, Xia F, Zheng M, Xu Y. CD8 + T cells mediate antiviral response in severe fever with thrombocytopenia syndrome. FASEB J 2023; 37:e22722. [PMID: 36571509 DOI: 10.1096/fj.202201343rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/27/2022]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), which is caused by a novel Bunyavirus, has gradually become a threatening infectious disease in rural areas of Asia. Studies have identified a severe cytokine storm and impaired humoral immune response in SFTS. However, the cellular immune response to SFTS virus (SFTSV) infection remains largely unknown. Here we report that SFTS patients had a cytokine storm accompanied by high levels of chemokines. CD8+ T cells in peripheral blood mononuclear cells of SFTS patients exhibited a more activated phenotype and enhanced the antiviral responses. They increased the expression of CD69 and CD25, secreted a higher level of IFN-γ and granzyme, and had a stronger proliferative ability than in healthy controls. In convalescent SFTS patients, the expression of CD69 and CD25 on CD8+ T cells was reduced. In addition, we found the ratio and cellularity of CD14+ CD16+ intermediate monocytes were increased in peripheral blood of SFTS patients. Both the expression of C-X-C motif chemokine ligand 10 (CXCL10) on CD14+ CD16+ intermediate monocytes and the expression of C-X-C motif chemokine receptor 3 (CXCR3) on CD8+ T cells increased dramatically in SFTS patients. Our studies reveal a potential pathway that CD8+ T cells rapidly activate and are mostly recruited by intermediate monocytes through CXCL10 in SFTSV infection. Our results may be of clinical relevance for further treatment and discharge instructions in SFTSV infections.
Collapse
Affiliation(s)
- Lu Zong
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Siyu Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yufeng Gao
- Department of Infectious Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fang Xia
- Department of Clinical Laboratory, People's Hospital of Hanshan County, Maanshan, China
| | - Meijuan Zheng
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
22
|
Exotic viral hepatitis: A review on epidemiology, pathogenesis, and treatment. J Hepatol 2022; 77:1431-1443. [PMID: 35817222 DOI: 10.1016/j.jhep.2022.06.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/14/2022] [Accepted: 06/19/2022] [Indexed: 12/04/2022]
Abstract
Certain "exotic" viruses are known to cause clinical diseases with potential liver involvement. These include viruses, beyond regular hepatotropic viruses (hepatitis A, -B(D), -C, -E, cytomegalovirus, Epstein-Barr virus), that can be found in (sub)tropical areas and can cause "exotic viral hepatitis". Transmission routes typically involve arthropods (Crimean Congo haemorrhagic fever, dengue, Rift Valley fever, yellow fever). However, some of these viruses are transmitted by the aerosolised excreta of rodents (Hantavirus, Lassa fever), or via direct contact or contact with bodily fluids (Ebola). Although some exotic viruses are associated with high fatality rates, such as Ebola for example, the clinical presentation of most exotic viruses can range from mild flu-like symptoms, in most cases, right through to being potentially fatal. A smaller percentage of people develop severe disease with haemorrhagic fever, possibly with (fulminant) hepatitis. Liver involvement is often caused by direct tropism for hepatocytes and Kupffer cells, resulting in virus-mediated and/or immune-mediated necrosis. In all exotic hepatitis viruses, PCR is the most sensitive diagnostic method. The determination of IgM/IgG antibodies is a reasonable alternative, but cross-reactivity can be a problem in the case of flaviviruses. Licenced vaccines are available for yellow fever and Ebola, and they are currently under development for dengue. Therapy for exotic viral hepatitis is predominantly supportive. To ensure that preventive measures can be introduced to control possible outbreaks, the timely detection of these viruses is very important.
Collapse
|
23
|
Nyakarahuka L, Whitmer S, Kyondo J, Mulei S, Cossaboom CM, Telford CT, Tumusiime A, Akurut GG, Namanya D, Kamugisha K, Baluku J, Lutwama J, Balinandi S, Shoemaker T, Klena JD. Crimean-Congo Hemorrhagic Fever Outbreak in Refugee Settlement during COVID-19 Pandemic, Uganda, April 2021. Emerg Infect Dis 2022; 28:2326-2329. [PMID: 36198315 PMCID: PMC9622249 DOI: 10.3201/eid2811.220365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) was detected in 2 refugees living in a refugee settlement in Kikuube district, Uganda. Investigations revealed a CCHF IgG seroprevalence of 71.3% (37/52) in goats within the refugee settlement. This finding highlights the need for a multisectoral approach to controlling CCHF in humans and animals in Uganda.
Collapse
|
24
|
Flórez-Álvarez L, de Souza EE, Botosso VF, de Oliveira DBL, Ho PL, Taborda CP, Palmisano G, Capurro ML, Pinho JRR, Ferreira HL, Minoprio P, Arruda E, de Souza Ferreira LC, Wrenger C, Durigon EL. Hemorrhagic fever viruses: Pathogenesis, therapeutics, and emerging and re-emerging potential. Front Microbiol 2022; 13:1040093. [PMID: 36386719 PMCID: PMC9640979 DOI: 10.3389/fmicb.2022.1040093] [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: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 01/29/2023] Open
Abstract
Hemorrhagic fever viruses (HFVs) pose a threat to global public health owing to the emergence and re-emergence of highly fatal diseases. Viral hemorrhagic fevers (VHFs) caused by these viruses are mostly characterized by an acute febrile syndrome with coagulation abnormalities and generalized hemorrhage that may lead to life-threatening organ dysfunction. Currently, the events underlying the viral pathogenicity associated with multiple organ dysfunction syndrome still underexplored. In this minireview, we address the current knowledge of the mechanisms underlying VHFs pathogenesis and discuss the available development of preventive and therapeutic options to treat these infections. Furthermore, we discuss the potential of HFVs to cause worldwide emergencies along with factors that favor their spread beyond their original niches.
Collapse
Affiliation(s)
| | | | | | | | - Paulo Lee Ho
- Virology Laboratory, Butantan Institute, São Paulo, Brazil
| | | | - Giuseppe Palmisano
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - João Renato Rebello Pinho
- Albert Einstein Institute for Teaching and Research (IIEP), Hospital Israelita Albert Einstein, São Paulo, Brazil,Hospital das Clínicas da Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Helena Lage Ferreira
- Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | - Eurico Arruda
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Luís Carlos de Souza Ferreira
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil
| | - Carsten Wrenger
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| |
Collapse
|
25
|
Toba S, Sato A, Kawai M, Taoda Y, Unoh Y, Kusakabe S, Nobori H, Uehara S, Uemura K, Taniguchi K, Kobayashi M, Noshi T, Yoshida R, Naito A, Shishido T, Maruyama J, Paessler S, Carr MJ, Hall WW, Yoshimatsu K, Arikawa J, Matsuno K, Sakoda Y, Sasaki M, Orba Y, Sawa H, Kida H. Identification of cap-dependent endonuclease inhibitors with broad-spectrum activity against bunyaviruses. Proc Natl Acad Sci U S A 2022; 119:e2206104119. [PMID: 36037386 PMCID: PMC9457168 DOI: 10.1073/pnas.2206104119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/07/2022] [Indexed: 11/18/2022] Open
Abstract
Viral hemorrhagic fevers caused by members of the order Bunyavirales comprise endemic and emerging human infections that are significant public health concerns. Despite the disease severity, there are few therapeutic options available, and therefore effective antiviral drugs are urgently needed to reduce disease burdens. Bunyaviruses, like influenza viruses (IFVs), possess a cap-dependent endonuclease (CEN) that mediates the critical cap-snatching step of viral RNA transcription. We screened compounds from our CEN inhibitor (CENi) library and identified specific structural compounds that are 100 to 1,000 times more active in vitro than ribavirin against bunyaviruses, including Lassa virus, lymphocytic choriomeningitis virus (LCMV), and Junin virus. To investigate their inhibitory mechanism of action, drug-resistant viruses were selected in culture. Whole-genome sequencing revealed that amino acid substitutions in the CEN region of drug-resistant viruses were located in similar positions as those of the CEN α3-helix loop of IFVs derived under drug selection. Thus, our studies suggest that CENi compounds inhibit both bunyavirus and IFV replication in a mechanistically similar manner. Structural analysis revealed that the side chain of the carboxyl group at the seventh position of the main structure of the compound was essential for the high antiviral activity against bunyaviruses. In LCMV-infected mice, the compounds significantly decreased blood viral load, suppressed symptoms such as thrombocytopenia and hepatic dysfunction, and improved survival rates. These data suggest a potential broad-spectrum clinical utility of CENis for the treatment of both severe influenza and hemorrhagic diseases caused by bunyaviruses.
Collapse
Affiliation(s)
- Shinsuke Toba
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Akihiko Sato
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | | | | | - Yuto Unoh
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Shinji Kusakabe
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | | | | | - Kentaro Uemura
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Keiichi Taniguchi
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | | | | | | | | | | | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Michael J. Carr
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin, Ireland
| | - William W. Hall
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin, Ireland
- Global Virus Network, Baltimore, MD 21201
| | - Kumiko Yoshimatsu
- Laboratory of Animal Experimentation, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
| | - Jiro Arikawa
- Office for Biosafety Auditor, Nagasaki University, Nagasaki 852-8521, Japan
| | - Keita Matsuno
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Yoshihiro Sakoda
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Global Virus Network, Baltimore, MD 21201
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Hiroshi Kida
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| |
Collapse
|
26
|
Yuan Y, Wang G, Chen X, Ye XL, Li XK, Li R, Jiang WL, Zeng HL, Du J, Zhang XA, Li H, Fang LQ, Lu QB, Liu W. Thrombocytopenia and increased risk of adverse outcome in COVID-19 patients. PeerJ 2022; 10:e13608. [PMID: 35791362 PMCID: PMC9250762 DOI: 10.7717/peerj.13608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 05/27/2022] [Indexed: 01/17/2023] Open
Abstract
Background Thrombocytopenia was common in the coronavirus disease 2019 (COVID-19) patients during the infection, while the role of thrombocytopenia in COVID-19 pathogenesis and its relationship with systemic host response remained obscure. The study aimed to systematically evaluate the relationship between thrombocytopenia in COVID-19 patients and clinical, haematological and biochemical markers of the disease as well as adverse outcomes. Methods To assess the relationship between abnormal platelet levels and disease progression, a multi-center retrospective cohort study was conducted. COVID-19 patients with thrombocytopenia and a sub-cohort of matched patients without thrombocytopenia were compared for their clinical manifestations, haematological disorders, biochemical parameters, inflammatory markers and clinical outcome. Results Thrombocytopenia was present in 127 of 2,209 analyzed patients on admission. Compared with the control group, thrombocytopenia patients developed significantly higher frequency of respiratory failure (41.9% vs. 22.6%, P = 0.020), intensive care unit entrance (25.6% vs. 11.5%, P = 0.012), disseminated intravascular coagulation (45.2% vs. 10.6%, P < 0.001), more altered platelet morphology indexes and coagulation perturbation, higher levels of inflammatory markers. In addition, a significantly increased all-cause mortality (hazard ratio 3.08, 95% confidence interval 2.26-4.18, P < 0.001) was also observed in the patients with thrombocytopenia. Late development of thrombocytopenia beyond 14 days post-symptom was observed in 61 patients, from whom a comparable mortality rate yet longer duration to death was observed compared to those with early thrombocytopenia. Conclusions Our finding from this study adds to previous evidence that thrombocytopenia is associated with adverse outcome of the disease and recommend that platelet count and indices be included alongside other haematological, biochemical and inflammatory markers in COVID-19 patients' assessment during the hospital stay.
Collapse
Affiliation(s)
- Yang Yuan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Gang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xi Chen
- Department of Thoracic and Vascular Surgery, Wuhan First Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Lei Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiao-Kun Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Rui Li
- Department of Healthcare, School of Health Sciences, Wuhan University, Wuhan, Hubei, Wuhan, China,Global Health Institute, Wuhan University, Wuhan, Hubei, Wuhan, China
| | - Wan-Li Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao-Long Zeng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Du
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Xiao-Ai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Li-Qun Fang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China,Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
27
|
Recent advances in treatment Crimean-Congo hemorrhagic fever virus: A concise overview. Microb Pathog 2022; 169:105657. [PMID: 35753597 DOI: 10.1016/j.micpath.2022.105657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
The Crimean Congo Hemorrhagic Fever Virus (CCHFV) is widespread in Africa, Asia, and Europe, among other places. The disease was initially discovered in the Crimean cities of the Soviet Union and the Congo, and it was given the name Crimean Congo because it induces hemorrhagic fever. According to studies, when the virus enters the body, it settles in immune cells such as macrophages and dendritic cells, causing them to malfunction and secrete inflammatory cytokines such as TNF-alpha, IL1, and IL6, resulting in cytokine storms that induces shock via endothelial activation and vascular leakage, while on the other hand, clots and disseminated intravascular coagulation (DIC) formation causes massive defects in various organs such as the liver and kidneys, as well as fatal bleeding. Disease prevention and treatment are crucial since no other effective vaccination against the disease has yet been developed. Immunotherapy is utilized as a consequence. One of the most effective treatments, when combined with compensatory therapies such as blood and platelet replacement, water, electrolytes, Fresh Frozen Plasma (FFP) replacement, and other compensatory therapies, is one of the most effective treatments. Studies; show that immunotherapy using IVIG and neutralizing and non-neutralizing monoclonal antibodies; cytokine therapy, and anti-inflammatory therapy using corticosteroids are effective ways to treat the disease.
Collapse
|
28
|
Ng WM, Sahin M, Krumm SA, Seow J, Zeltina A, Harlos K, Paesen GC, Pinschewer DD, Doores KJ, Bowden TA. Contrasting Modes of New World Arenavirus Neutralization by Immunization-Elicited Monoclonal Antibodies. mBio 2022; 13:e0265021. [PMID: 35315691 PMCID: PMC9040744 DOI: 10.1128/mbio.02650-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/28/2022] [Indexed: 12/31/2022] Open
Abstract
Transmission of the New World hemorrhagic fever arenaviruses Junín virus (JUNV) and Machupo virus (MACV) to humans is facilitated, in part, by the interaction between the arenavirus GP1 glycoprotein and the human transferrin receptor 1 (hTfR1). We utilize a mouse model of live-attenuated immunization with envelope exchange viruses to isolate neutralizing monoclonal antibodies (NAbs) specific to JUNV GP1 and MACV GP1. Structures of two NAbs, termed JUN1 and MAC1, demonstrate that they neutralize through disruption of hTfR1 recognition. JUN1 utilizes a binding mode common to all characterized infection- and vaccine-elicited JUNV-specific NAbs, which involves mimicking hTfR1 binding through the insertion of a tyrosine into the receptor-binding site. In contrast, MAC1 undergoes a tyrosine-mediated mode of antigen recognition distinct from that used by the reported anti-JUNV NAbs and the only other characterized anti-MACV NAb. These data reveal the varied modes of GP1-specific recognition among New World arenaviruses by the antibody-mediated immune response. IMPORTANCE The GP1 subcomponent of the New World arenavirus GP is a primary target of the neutralizing antibody response, which has been shown to be effective in the prevention and treatment of infection. Here, we characterize the structural basis of the antibody-mediated immune response that arises from immunization of mice against Junín virus and Machupo virus, two rodent-borne zoonotic New World arenaviruses. We isolate a panel of GP1-specific monoclonal antibodies that recognize overlapping epitopes and exhibit neutralizing behavior, in vitro. Structural characterization of two of these antibodies indicates that antibody recognition likely interferes with GP1-mediated recognition of the transferrin receptor 1. These data provide molecular-level detail for a key region of vulnerability on the New World arenavirus surface and a blueprint for therapeutic antibody development.
Collapse
Affiliation(s)
- Weng M. Ng
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Mehmet Sahin
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Stefanie A. Krumm
- Kings College London, Department of Infectious Diseases, Guy’s Hospital, London, United Kingdom
| | - Jeffrey Seow
- Kings College London, Department of Infectious Diseases, Guy’s Hospital, London, United Kingdom
| | - Antra Zeltina
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Karl Harlos
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Daniel D. Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Katie J. Doores
- Kings College London, Department of Infectious Diseases, Guy’s Hospital, London, United Kingdom
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Meuren LM, Prestes EB, Papa MP, de Carvalho LRP, Mustafá YM, da Costa LS, Da Poian AT, Bozza MT, Arruda LB. Infection of Endothelial Cells by Dengue Virus Induces ROS Production by Different Sources Affecting Virus Replication, Cellular Activation, Death and Vascular Permeability. Front Immunol 2022; 13:810376. [PMID: 35185902 PMCID: PMC8847576 DOI: 10.3389/fimmu.2022.810376] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 01/20/2023] Open
Abstract
Exacerbated inflammatory response and altered vascular function are hallmarks of dengue disease. Reactive oxygen species (ROS) production has been associated to endothelial barrier disturbance and microvascular alteration in distinct pathological conditions. Increased ROS has been reported in in vitro models of dengue virus (DENV) infection, but its impact for endothelial cell physiology had not been fully investigated. Our group had previously demonstrated that infection of human brain microvascular endothelial cells (HBMEC) with DENV results in the activation of RNA sensors and production of proinflammatory cytokines, which culminate in cell death and endothelial permeability. Here, we evaluated the role of mitochondrial function and NADPH oxidase (NOX) activation for ROS generation in HBMEC infected by DENV and investigated whether altered cellular physiology could be a consequence of virus-induced oxidative stress. DENV-infected HBMECs showed a decrease in the maximal respiratory capacity and altered membrane potential, indicating functional mitochondrial alteration, what might be related to mtROS production. Indeed, mtROS was detected at later time points after infection. Specific inhibition of mtROS diminished virus replication, cell death, and endothelial permeability, but did not affect cytokine production. On the other hand, inhibition of NOX-associated ROS production decreased virus replication and cell death, as well as the secretion of inflammatory cytokines, including IL-6, IL-8, and CCL5. These results demonstrated that DENV replication in endothelial cells induces ROS production by different pathways, which impacts biological functions that might be relevant for dengue pathogenesis. Those data also indicate oxidative stress events as relevant therapeutical targets to avoid vascular permeability, inflammation, and neuroinvasion during DENV infection.
Collapse
Affiliation(s)
- Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Beatriz Prestes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Premazzi Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States
| | | | - Yasmin Mucunã Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro Silva da Costa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Torres Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Barros Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Abdel-Bakky MS, Amin E, Ewees MG, Mahmoud NI, Mohammed HA, Altowayan WM, Abdellatif AAH. Coagulation System Activation for Targeting of COVID-19: Insights into Anticoagulants, Vaccine-Loaded Nanoparticles, and Hypercoagulability in COVID-19 Vaccines. Viruses 2022; 14:228. [PMID: 35215822 PMCID: PMC8876839 DOI: 10.3390/v14020228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as COVID-19, is currently developing into a rapidly disseminating and an overwhelming worldwide pandemic. In severe COVID-19 cases, hypercoagulability and inflammation are two crucial complications responsible for poor prognosis and mortality. In addition, coagulation system activation and inflammation overlap and produce life-threatening complications, including coagulopathy and cytokine storm, which are associated with overproduction of cytokines and activation of the immune system; they might be a lead cause of organ damage. However, patients with severe COVID-19 who received anticoagulant therapy had lower mortality, especially with elevated D-dimer or fibrin degradation products (FDP). In this regard, the discovery of natural products with anticoagulant potential may help mitigate the numerous side effects of the available synthetic drugs. This review sheds light on blood coagulation and its impact on the complication associated with COVID-19. Furthermore, the sources of natural anticoagulants, the role of nanoparticle formulation in this outbreak, and the prevalence of thrombosis with thrombocytopenia syndrome (TTS) after COVID-19 vaccines are also reviewed. These combined data provide many research ideas related to the possibility of using these anticoagulant agents as a treatment to relieve acute symptoms of COVID-19 infection.
Collapse
Affiliation(s)
- Mohamed S. Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Elham Amin
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Mohamed G. Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Nesreen I. Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Waleed M. Altowayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Ahmed A. H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Qasssim 52471, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
31
|
Siering O, Cattaneo R, Pfaller CK. C Proteins: Controllers of Orderly Paramyxovirus Replication and of the Innate Immune Response. Viruses 2022; 14:v14010137. [PMID: 35062341 PMCID: PMC8778822 DOI: 10.3390/v14010137] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 01/07/2023] Open
Abstract
Particles of many paramyxoviruses include small amounts of proteins with a molecular weight of about 20 kDa. These proteins, termed “C”, are basic, have low amino acid homology and some secondary structure conservation. C proteins are encoded in alternative reading frames of the phosphoprotein gene. Some viruses express nested sets of C proteins that exert their functions in different locations: In the nucleus, they interfere with cellular transcription factors that elicit innate immune responses; in the cytoplasm, they associate with viral ribonucleocapsids and control polymerase processivity and orderly replication, thereby minimizing the activation of innate immunity. In addition, certain C proteins can directly bind to, and interfere with the function of, several cytoplasmic proteins required for interferon induction, interferon signaling and inflammation. Some C proteins are also required for efficient virus particle assembly and budding. C-deficient viruses can be grown in certain transformed cell lines but are not pathogenic in natural hosts. C proteins affect the same host functions as other phosphoprotein gene-encoded proteins named V but use different strategies for this purpose. Multiple independent systems to counteract host defenses may ensure efficient immune evasion and facilitate virus adaptation to new hosts and tissue environments.
Collapse
Affiliation(s)
- Oliver Siering
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, 63225 Langen, Germany;
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55906, USA
- Correspondence: (R.C.); (C.K.P.)
| | - Christian K. Pfaller
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, 63225 Langen, Germany;
- Correspondence: (R.C.); (C.K.P.)
| |
Collapse
|
32
|
Ghosh S, Saha A, Samanta S, Saha RP. Genome structure and genetic diversity in the Ebola virus. Curr Opin Pharmacol 2021; 60:83-90. [PMID: 34364102 DOI: 10.1016/j.coph.2021.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022]
Abstract
Ebola is a deadly pathogen responsible for Ebola virus disease, first came to prominence in the year 1976. This rapidly evolving virus imposed a serious threat to the human population in the last few decades and also continues to be a probable threat to our race. A better understanding of the virus in terms of its genomic structure is very much needed to develop an effective antiviral therapy against this deadly pathogen. Complete knowledge of its genomic structure and variations will help us and the entire scientific community to design effective therapy in terms of either vaccine development or the development of proper antiviral medicine.
Collapse
Affiliation(s)
- Sanmitra Ghosh
- Department of Microbiology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Saikat Samanta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Rudra P Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India.
| |
Collapse
|
33
|
Venous and arterial thromboembolic disease in COVID-19. J Thromb Thrombolysis 2021; 52:1007-1009. [PMID: 34255267 PMCID: PMC8276541 DOI: 10.1007/s11239-021-02524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 11/12/2022]
|
34
|
Richardson E, García-Bernal D, Calabretta E, Jara R, Palomo M, Baron RM, Yanik G, Fareed J, Vlodavsky I, Iacobelli M, Díaz-Ricart M, Richardson PG, Carlo-Stella C, Moraleda JM. Defibrotide: potential for treating endothelial dysfunction related to viral and post-infectious syndromes. Expert Opin Ther Targets 2021; 25:423-433. [PMID: 34167431 DOI: 10.1080/14728222.2021.1944101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Defibrotide (DF) is a polyribonucleotide with antithrombotic, pro-fibrinolytic, and anti-inflammatory effects on endothelium. These effects and the established safety of DF present DF as a strong candidate to treat viral and post-infectious syndromes involving endothelial dysfunction. AREAS COVERED We discuss DF and other therapeutic agents that have the potential to target endothelial components of pathogenesis in viral and post-infectious syndromes. We introduce defibrotide (DF), describe its mechanisms of action, and explore its established pleiotropic effects on the endothelium. We describe the established pathophysiology of Coronavirus Disease 2019 (COVID-19) and highlight the processes specific to COVID-19 potentially modulated by DF. We also present influenza A and viral hemorrhagic fevers, especially those caused by hantavirus, Ebola virus, and dengue virus, as viral syndromes in which DF might serve therapeutic benefit. Finally, we offer our opinion on novel treatment strategies targeting endothelial dysfunction in viral infections and their severe manifestations. EXPERT OPINION Given the critical role of endothelial dysfunction in numerous infectious syndromes, in particular COVID-19, therapeutic pharmacology for these conditions should increasingly prioritize endothelial stabilization. Several agents with endothelial protective properties should be further studied as treatments for severe viral infections and vasculitides, especially where other therapeutic modalities have failed.
Collapse
Affiliation(s)
- Edward Richardson
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut, USA.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David García-Bernal
- Department of Medicine, Stem Cell Transplant and Cell Therapy Unit, IMIB-Arrixaca, Virgen De La Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Eleonora Calabretta
- Department of Biomedical Sciences, Humanitas University, Rozzano-Milano, Italy.,Department of Oncology and Hematology, IRCCS - Humanitas Research Hospital, Rozzano-Milano, Italy
| | - Rubén Jara
- Intensive Care Unit, Virgen De La Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Marta Palomo
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Barcelona Endothelium Team, Barcelona, Spain
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory Yanik
- Department of Pediatrics, Blood and Marrow Transplantation Program, University of Michigan, Ann Arbor, MI, USA.,Department of Internal Medicine, Blood and Marrow Transplantation Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Jawed Fareed
- Department of Molecular Pharmacology and Therapeutics, Hemostasis and Thrombosis Research Laboratories, Loyola University Medical Center, Chicago, Illinois, USA
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | - Maribel Díaz-Ricart
- Barcelona Endothelium Team, Barcelona, Spain.,Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain.,IDIBAPS, Barcelona, Spain
| | - Paul G Richardson
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Hematologic Malignancy, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Carmelo Carlo-Stella
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut, USA.,Department of Biomedical Sciences, Humanitas University, Rozzano-Milano, Italy.,Department of Oncology and Hematology, IRCCS - Humanitas Research Hospital, Rozzano-Milano, Italy.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jose M Moraleda
- Department of Medicine, Stem Cell Transplant and Cell Therapy Unit, IMIB-Arrixaca, Virgen De La Arrixaca University Hospital, University of Murcia, Murcia, Spain
| |
Collapse
|
35
|
Sarute N, Cheng H, Yan Z, Salas-Briceno K, Richner J, Rong L, Ross SR. Signal-regulatory protein alpha is an anti-viral entry factor targeting viruses using endocytic pathways. PLoS Pathog 2021; 17:e1009662. [PMID: 34097709 PMCID: PMC8211255 DOI: 10.1371/journal.ppat.1009662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/17/2021] [Accepted: 05/20/2021] [Indexed: 01/17/2023] Open
Abstract
Signal-regulatory protein alpha (SIRPA) is a well-known inhibitor of phagocytosis when it complexes with CD47 expressed on target cells. Here we show that SIRPA decreased in vitro infection by a number of pathogenic viruses, including New World and Old World arenaviruses, Zika virus, vesicular stomatitis virus and pseudoviruses bearing the Machupo virus, Ebola virus and SARS-CoV-2 glycoproteins, but not HSV-1, MLV or mNoV. Moreover, mice with targeted mutation of the Sirpa gene that renders it non-functional were more susceptible to infection with the New World arenaviruses Junín virus vaccine strain Candid 1 and Tacaribe virus, but not MLV or mNoV. All SIRPA-inhibited viruses have in common the requirement for trafficking to a low pH endosomal compartment. This was clearly demonstrated with SARS-CoV-2 pseudovirus, which was only inhibited by SIRPA in cells in which it required trafficking to the endosome. Similar to its role in phagocytosis inhibition, SIRPA decreased virus internalization but not binding to cell surface receptors. We also found that increasing SIRPA levels via treatment with IL-4 led to even greater anti-viral activity. These data suggest that enhancing SIRPA’s activity could be a target for anti-viral therapies. Viruses enter cells via different routes. Many RNA viruses require trafficking to a low pH compartment to accomplish entry. Similarly, phagocytosis of dead cells by macrophages results in their degradation in an acidic compartment. Here we show that SIRPA, which is a major inhibitor of phagocytosis, also inhibits infection by a variety of viruses that enter via acidic compartments, including many human pathogens such as Zika, Ebola and SARS-CoV-2. These findings suggest that phagocytosis and virus endocytosis share a common mechanistic pathway, and could lead to new approaches to the development of anti-viral therapeutics.
Collapse
Affiliation(s)
- Nicolás Sarute
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Han Cheng
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Zhonghao Yan
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Karen Salas-Briceno
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Justin Richner
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Lijun Rong
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Susan R. Ross
- University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
36
|
Fang XZ, Wang YX, Xu JQ, He YJ, Peng ZK, Shang Y. Immunothrombosis in Acute Respiratory Dysfunction of COVID-19. Front Immunol 2021; 12:651545. [PMID: 34149692 PMCID: PMC8207198 DOI: 10.3389/fimmu.2021.651545] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023] Open
Abstract
COVID-19 is an acute, complex disorder that was caused by a new β-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Based on current reports, it was surprising that the characteristics of many patients with COVID-19, who fulfil the Berlin criteria for acute respiratory distress syndrome (ARDS), are not always like those of patients with typical ARDS and can change over time. While the mechanisms of COVID-19–related respiratory dysfunction in COVID-19 have not yet been fully elucidated, pulmonary microvascular thrombosis is speculated to be involved. Considering that thrombosis is highly related to other inflammatory lung diseases, immunothrombosis, a two-way process that links coagulation and inflammation, seems to be involved in the pathophysiology of COVID-19, including respiratory dysfunction. Thus, the current manuscript will describe the proinflammatory milieu in COVID-19, summarize current evidence of thrombosis in COVID-19, and discuss possible interactions between these two.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Qain Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Kang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Wang Q, Xin Q, Shang W, Wan W, Xiao G, Zhang LK. Activation of the STAT3 Signaling Pathway by the RNA-Dependent RNA Polymerase Protein of Arenavirus. Viruses 2021; 13:v13060976. [PMID: 34070281 PMCID: PMC8225222 DOI: 10.3390/v13060976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Arenaviruses cause chronic and asymptomatic infections in their natural host, rodents, and several arenaviruses cause severe hemorrhagic fever that has a high mortality in infected humans, seriously threatening public health. There are currently no FDA-licensed drugs available against arenaviruses; therefore, it is important to develop novel antiviral strategies to combat them, which would be facilitated by a detailed understanding of the interactions between the viruses and their hosts. To this end, we performed a transcriptomic analysis on cells infected with arenavirus lymphocytic choriomeningitis virus (LCMV), a neglected human pathogen with clinical significance, and found that the signal transducer and activator of transcription 3 (STAT3) signaling pathway was activated. A further investigation indicated that STAT3 could be activated by the RNA-dependent RNA polymerase L protein (Lp) of LCMV. Our functional analysis found that STAT3 cannot affect LCMV multiplication in A549 cells. We also found that STAT3 was activated by the Lp of Mopeia virus and Junin virus, suggesting that this activation may be conserved across certain arenaviruses. Our study explored the interactions between arenaviruses and STAT3, which may help us to better understand the molecular and cell biology of arenaviruses.
Collapse
Affiliation(s)
- Qingxing Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qilin Xin
- UMR754, Viral Infections and Comparative Pathology, 50 Avenue Tony Garnier, CEDEX 07, 69366 Lyon, France;
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
| | - Weiwei Wan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (G.X.); (L.-K.Z.)
| | - Lei-Ke Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (G.X.); (L.-K.Z.)
| |
Collapse
|
38
|
He L, Chaudhary A, Lin X, Sou C, Alkutkar T, Kumar S, Ngo T, Kosviner E, Ozorowski G, Stanfield RL, Ward AB, Wilson IA, Zhu J. Single-component multilayered self-assembling nanoparticles presenting rationally designed glycoprotein trimers as Ebola virus vaccines. Nat Commun 2021; 12:2633. [PMID: 33976149 DOI: 10.1101/2020.08.22.262634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/06/2021] [Indexed: 05/27/2023] Open
Abstract
Ebola virus (EBOV) glycoprotein (GP) can be recognized by neutralizing antibodies (NAbs) and is the main target for vaccine design. Here, we first investigate the contribution of the stalk and heptad repeat 1-C (HR1C) regions to GP metastability. Specific stalk and HR1C modifications in a mucin-deleted form (GPΔmuc) increase trimer yield, whereas alterations of HR1C exert a more complex effect on thermostability. Crystal structures are determined to validate two rationally designed GPΔmuc trimers in their unliganded state. We then display a modified GPΔmuc trimer on reengineered protein nanoparticles that encapsulate a layer of locking domains (LD) and a cluster of helper T-cell epitopes. In mice and rabbits, GP trimers and nanoparticles elicit cross-ebolavirus NAbs, as well as non-NAbs that enhance pseudovirus infection. Repertoire sequencing reveals quantitative profiles of vaccine-induced B-cell responses. This study demonstrates a promising vaccine strategy for filoviruses, such as EBOV, based on GP stabilization and nanoparticle display.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antigens, Viral/administration & dosage
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/ultrastructure
- B-Lymphocytes/immunology
- Crystallography, X-Ray
- Disease Models, Animal
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/genetics
- Ebola Vaccines/immunology
- Ebolavirus/genetics
- Ebolavirus/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/ultrastructure
- Female
- Glycoproteins/administration & dosage
- Glycoproteins/genetics
- Glycoproteins/immunology
- Glycoproteins/ultrastructure
- Hemorrhagic Fever, Ebola/blood
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/therapy
- Hemorrhagic Fever, Ebola/virology
- Humans
- Mice
- Nanoparticles/chemistry
- Protein Domains/genetics
- Protein Domains/immunology
- Protein Engineering
- Protein Multimerization/genetics
- Protein Multimerization/immunology
- Protein Stability
- Rabbits
- T-Lymphocytes, Helper-Inducer/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Viral Proteins/administration & dosage
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Proteins/ultrastructure
Collapse
Affiliation(s)
- Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Anshul Chaudhary
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Xiaohe Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Cindy Sou
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tanwee Alkutkar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sonu Kumar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Timothy Ngo
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ezra Kosviner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
39
|
He L, Chaudhary A, Lin X, Sou C, Alkutkar T, Kumar S, Ngo T, Kosviner E, Ozorowski G, Stanfield RL, Ward AB, Wilson IA, Zhu J. Single-component multilayered self-assembling nanoparticles presenting rationally designed glycoprotein trimers as Ebola virus vaccines. Nat Commun 2021; 12:2633. [PMID: 33976149 PMCID: PMC8113551 DOI: 10.1038/s41467-021-22867-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
Ebola virus (EBOV) glycoprotein (GP) can be recognized by neutralizing antibodies (NAbs) and is the main target for vaccine design. Here, we first investigate the contribution of the stalk and heptad repeat 1-C (HR1C) regions to GP metastability. Specific stalk and HR1C modifications in a mucin-deleted form (GPΔmuc) increase trimer yield, whereas alterations of HR1C exert a more complex effect on thermostability. Crystal structures are determined to validate two rationally designed GPΔmuc trimers in their unliganded state. We then display a modified GPΔmuc trimer on reengineered protein nanoparticles that encapsulate a layer of locking domains (LD) and a cluster of helper T-cell epitopes. In mice and rabbits, GP trimers and nanoparticles elicit cross-ebolavirus NAbs, as well as non-NAbs that enhance pseudovirus infection. Repertoire sequencing reveals quantitative profiles of vaccine-induced B-cell responses. This study demonstrates a promising vaccine strategy for filoviruses, such as EBOV, based on GP stabilization and nanoparticle display. Ebola virus glycoprotein (GP) is a major target for vaccine design. Here, the authors identify mutations to improve GP stability and yield, design two multilayered nanoparticle carriers, and demonstrate good immunogenicity of the modified GP on nanoparticles in mice and rabbits.
Collapse
Affiliation(s)
- Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Anshul Chaudhary
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Xiaohe Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Cindy Sou
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tanwee Alkutkar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sonu Kumar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Timothy Ngo
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ezra Kosviner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. .,Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. .,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
40
|
Lai JH, Wu DW, Wu CH, Hung LF, Huang CY, Ka SM, Chen A, Chang ZF, Ho LJ. Mitochondrial CMPK2 mediates immunomodulatory and antiviral activities through IFN-dependent and IFN-independent pathways. iScience 2021; 24:102498. [PMID: 34142025 PMCID: PMC8188380 DOI: 10.1016/j.isci.2021.102498] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/31/2021] [Accepted: 04/28/2021] [Indexed: 01/22/2023] Open
Abstract
Mitochondria regulate the immune response after dengue virus (DENV) infection. Microarray analysis of genes identified the upregulation of mitochondrial cytidine/uridine monophosphate kinase 2 (CMPK2) by DENV infection. We used small interfering RNA-mediated knockdown (KD) and CRISPR-Cas9 knockout (KO) approaches, to investigate the role of CMPK2 in mouse and human cells. The results showed that CMPK2 was critical in DENV-induced antiviral cytokine release and mitochondrial oxidative stress and mitochondrial DNA release to the cytosol. The DENV-induced activation of Toll-like receptor (TLR)-9, inflammasome pathway, and cell migration was suppressed by CMPK2 depletion; however, viral production increased under CMPK2 deficiency. Examining mouse bone marrow-derived dendritic cells from interferon-alpha (IFN-α) receptor-KO mice and signal transducer and activator of transcription 1 (STAT1)-KO mice, we confirmed that CMPK2-mediated antiviral activity occurred in IFN-dependent and IFN-independent manners. In sum, CMPK2 is a critical factor in DENV-induced immune responses to determine innate immunity.
Collapse
Affiliation(s)
- Jenn-Haung Lai
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, R.O.C
| | - De-Wei Wu
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, R.O.C
| | - Chien-Hsiang Wu
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, R.O.C
| | - Li-Feng Hung
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| | - Chuan-Yueh Huang
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| | - Shuk-Man Ka
- Graduate Institute of Aerospace and Undersea Medicine, Department of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ann Chen
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Zee-Fen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| |
Collapse
|
41
|
Wang LL, Palermo N, Estrada L, Thompson C, Patten JJ, Anantpadma M, Davey RA, Xiang SH. Identification of filovirus entry inhibitors targeting the endosomal receptor NPC1 binding site. Antiviral Res 2021; 189:105059. [PMID: 33705865 PMCID: PMC8088776 DOI: 10.1016/j.antiviral.2021.105059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/27/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022]
Abstract
Filoviruses, mainly consisting of Ebola viruses (EBOV) and Marburg viruses (MARV), are enveloped negative-strand RNA viruses which can infect humans to cause severe hemorrhagic fevers and outbreaks with high mortality rates. The filovirus infection is mediated by the interaction of viral envelope glycoprotein (GP) and the human endosomal receptor Niemann-Pick C1 (NPC1). Blocking this interaction will prevent the infection. Therefore, we utilized an In silico screening approach to conduct virtual compound screening against the NPC1 receptor-binding site (RBS). Twenty-six top-hit compounds were purchased and evaluated by in vitro cell based inhibition assays against pseudotyped or replication-competent filoviruses. Two classes (A and U) of compounds were identified to have potent inhibitory activity against both Ebola and Marburg viruses. The IC50 values are in the lower level of micromolar concentrations. One compound (compd-A) was found to have a sub-micromolar IC50 value (0.86 μM) against pseudotyped Marburg virus. The cytotoxicity assay (MTT) indicates that compd-A has a moderate cytotoxicity level but the compd-U has much less toxicity and the CC50 value was about 100 μM. Structure-activity relationship (SAR) study has found some analogs of compd-A and -U have reduced the toxicity and enhanced the inhibitory activity. In conclusion, this work has identified several qualified lead-compounds for further drug development against filovirus infection.
Collapse
Affiliation(s)
- Leah Liu Wang
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Nicholas Palermo
- Computational Chemistry Core Facility, VCR Cores, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Leslie Estrada
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Colton Thompson
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - J J Patten
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Manu Anantpadma
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Robert A Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Shi-Hua Xiang
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
42
|
Arruda LB, Haider N, Olayemi A, Simons D, Ehichioya D, Yinka-Ogunleye A, Ansumana R, Thomason MJ, Asogun D, Ihekweazu C, Fichet-Calvet E, Kock RA. The niche of One Health approaches in Lassa fever surveillance and control. Ann Clin Microbiol Antimicrob 2021; 20:29. [PMID: 33894784 PMCID: PMC8067790 DOI: 10.1186/s12941-021-00431-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
Lassa fever (LF), a zoonotic illness, represents a public health burden in West African countries where the Lassa virus (LASV) circulates among rodents. Human exposure hinges significantly on LASV ecology, which is in turn shaped by various parameters such as weather seasonality and even virus and rodent-host genetics. Furthermore, human behaviour, despite playing a key role in the zoonotic nature of the disease, critically affects either the spread or control of human-to-human transmission. Previous estimations on LF burden date from the 80s and it is unclear how the population expansion and the improvement on diagnostics and surveillance methods have affected such predictions. Although recent data have contributed to the awareness of epidemics, the real impact of LF in West African communities will only be possible with the intensification of interdisciplinary efforts in research and public health approaches. This review discusses the causes and consequences of LF from a One Health perspective, and how the application of this concept can improve the surveillance and control of this disease in West Africa.
Collapse
Affiliation(s)
- Liã Bárbara Arruda
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK.
| | - Najmul Haider
- The Royal Veterinary College, University of London, Hatfield, UK
| | - Ayodeji Olayemi
- Natural History Museum, Obafemi Awolowo University, Ile Ife, Nigeria
| | - David Simons
- The Royal Veterinary College, University of London, Hatfield, UK
| | - Deborah Ehichioya
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Nigeria.,Department of Microbiology, Ambrose Alli University, Ekpoma, Nigeria
| | | | - Rashid Ansumana
- School of Community Health Sciences, Njala University, Bo, Sierra Leone
| | - Margaret J Thomason
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| | - Danny Asogun
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | | | | | - Richard A Kock
- The Royal Veterinary College, University of London, Hatfield, UK
| |
Collapse
|
43
|
Odendaal L, Davis AS, Venter EH. Insights into the Pathogenesis of Viral Haemorrhagic Fever Based on Virus Tropism and Tissue Lesions of Natural Rift Valley Fever. Viruses 2021; 13:v13040709. [PMID: 33923863 PMCID: PMC8073615 DOI: 10.3390/v13040709] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV) infects humans and a wide range of ungulates and historically has caused devastating epidemics in Africa and the Arabian Peninsula. Lesions of naturally infected cases of Rift Valley fever (RVF) have only been described in detail in sheep with a few reports concerning cattle and humans. The most frequently observed lesion in both ruminants and humans is randomly distributed necrosis, particularly in the liver. Lesions supportive of vascular endothelial injury are also present and include mild hydropericardium, hydrothorax and ascites; marked pulmonary congestion and oedema; lymph node congestion and oedema; and haemorrhages in many tissues. Although a complete understanding of RVF pathogenesis is still lacking, antigen-presenting cells in the skin are likely the early targets of the virus. Following suppression of type I IFN production and necrosis of dermal cells, RVFV spreads systemically, resulting in infection and necrosis of other cells in a variety of organs. Failure of both the innate and adaptive immune responses to control infection is exacerbated by apoptosis of lymphocytes. An excessive pro-inflammatory cytokine and chemokine response leads to microcirculatory dysfunction. Additionally, impairment of the coagulation system results in widespread haemorrhages. Fatal outcomes result from multiorgan failure, oedema in many organs (including the lungs and brain), hypotension, and circulatory shock. Here, we summarize current understanding of RVF cellular tropism as informed by lesions caused by natural infections. We specifically examine how extant knowledge informs current understanding regarding pathogenesis of the haemorrhagic fever form of RVF, identifying opportunities for future research.
Collapse
Affiliation(s)
- Lieza Odendaal
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0002, South Africa
- Correspondence: (L.O.); (A.S.D.)
| | - A Sally Davis
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0002, South Africa
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
- Correspondence: (L.O.); (A.S.D.)
| | - Estelle H Venter
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria 0002, South Africa;
- College of Public Health Medical and Veterinary Sciences, Discipline Veterinary Science, James Cook University, Townsville, QLD 4811, Australia
| |
Collapse
|
44
|
Kolawole D, Raji H, Okeke MI. Phylogenetic and Mutational Analysis of Lassa Virus Strains Isolated in Nigeria: Proposal for an In Silico Study. JMIR Res Protoc 2021; 10:e23015. [PMID: 33769296 PMCID: PMC8088840 DOI: 10.2196/23015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 12/01/2022] Open
Abstract
Background In 2018, the total number of Lassa fever cases in Nigeria was significantly higher than that observed in previous years. Hence, studies had attempted to determine the underlying cause. However, reports using phylogenetic methods to analyze this finding ruled out the emergence of potentially more transmissible Lassa virus strains or an increase in human-to-human viral transmission as the cause underlying the increase in cases. Two years later, the situation seems even worse as the number of confirmed cases has reached an all-time high according to situational reports released by the Nigerian Center for Disease Control. Objective Considering the increasing trend of Lassa fever cases and related mortality, the major objective of this study is to map mutations within the genomes of Lassa virus isolates from 2018 and 2019 using the reference sequence available at the National Center for Biotechnology Information as a benchmark and compare them to the genomes of viruses isolated during 1969-2017. This study would also attempt to identify a viral marker gene for easier identification and grouping. Finally, the time-scaled evolution of Lassa virus in Nigeria will be reconstructed. Methods After collecting the sequence data of Lassa virus isolates, Bayesian phylogenetic trees, a sequence identity matrix, and a single nucleotide polymorphism matrix will be generated using BEAST (version 2.6.2), Base-By-Base, and DIVEIN (a web-based tool for variant calling), respectively. Results Mining and alignment of Lassa virus genome sequences have been completed, while mutational analysis and the reconstruction of time-scaled maximum clade credibility trees, congruence tests for inferred segments, and gene phylogeny analysis are ongoing. Conclusions The findings of this study would further the current knowledge of the evolutionary history of the Lassa virus in Nigeria and would document the mutations in Nigerian isolates from 1969 to 2019. International Registered Report Identifier (IRRID) DERR1-10.2196/23015
Collapse
Affiliation(s)
- Daniel Kolawole
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| | - Hayatu Raji
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| | - Malachy Ifeanyi Okeke
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| |
Collapse
|
45
|
Iba T, Levy JH, Connors JM, Warkentin TE, Thachil J, Levi M. Managing thrombosis and cardiovascular complications of COVID-19: answering the questions in COVID-19-associated coagulopathy. Expert Rev Respir Med 2021; 15:1003-1011. [PMID: 33667146 DOI: 10.1080/17476348.2021.1899815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The first patients with Coronavirus disease 2019 (COVID-19) emerged at the end of 2019. This novel viral infection demonstrated unique features that include prothrombotic clinical presentations. However, one year after the first occurrence, there remain many unanswered questions. We tried to address some of the important queries in this review. AREAS COVERED We raised the following critical questions. 'Why is COVID-19 so hypercoagulable?', 'Why are most coagulation test results relatively normal?', 'Why is COVID-19-associated coagulopathy more thrombotic than most other infectious diseases?', 'Why is arterial thrombus formed frequently?', 'Is anticoagulant therapy for COVID-19 effective?', and 'Are there racial disparities in thrombosis in COVID-19?' EXPERT OPINION There are commonalities and differences in the pathogeneses and clinical features between COVID-19 and other infectious diseases. Correct understanding will help discussing appropriate anticoagulation prophylaxis or treatment for thromboembolism.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Jean Marie Connors
- Hematology Division Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Theodore E Warkentin
- Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Canada
| | - Jecko Thachil
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Marcel Levi
- Department of Medicine, University College London Hospitals NHS Foundation Trust, and Cardio-metabolic Programme-NIHR UCLH/UCL BRC London, UK
| |
Collapse
|
46
|
Pathogen Dose in Animal Models of Hemorrhagic Fever Virus Infections and the Potential Impact on Studies of the Immune Response. Pathogens 2021; 10:pathogens10030275. [PMID: 33804381 PMCID: PMC7999429 DOI: 10.3390/pathogens10030275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Viral hemorrhagic fever viruses come from a wide range of virus families and are a significant cause of morbidity and mortality worldwide each year. Animal models of infection with a number of these viruses have contributed to our knowledge of their pathogenesis and have been crucial for the development of therapeutics and vaccines that have been approved for human use. Most of these models use artificially high doses of virus, ensuring lethality in pre-clinical drug development studies. However, this can have a significant effect on the immune response generated. Here I discuss how the dose of antigen or pathogen is a critical determinant of immune responses and suggest that the current study of viruses in animal models should take this into account when developing and studying animal models of disease. This can have implications for determination of immune correlates of protection against disease as well as informing relevant vaccination and therapeutic strategies.
Collapse
|
47
|
Fragkou PC, Moschopoulos CD, Karofylakis E, Kelesidis T, Tsiodras S. Update in Viral Infections in the Intensive Care Unit. Front Med (Lausanne) 2021; 8:575580. [PMID: 33708775 PMCID: PMC7940368 DOI: 10.3389/fmed.2021.575580] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 02/02/2021] [Indexed: 12/15/2022] Open
Abstract
The advent of highly sensitive molecular diagnostic techniques has improved our ability to detect viral pathogens leading to severe and often fatal infections that require admission to the Intensive Care Unit (ICU). Viral infections in the ICU have pleomorphic clinical presentations including pneumonia, acute respiratory distress syndrome, respiratory failure, central or peripheral nervous system manifestations, and viral-induced shock. Besides de novo infections, certain viruses fall into latency and can be reactivated in both immunosuppressed and immunocompetent critically ill patients. Depending on the viral strain, transmission occurs either directly through contact with infectious materials and large droplets, or indirectly through suspended air particles (airborne transmission of droplet nuclei). Many viruses can efficiently spread within hospital environment leading to in-hospital outbreaks, sometimes with high rates of mortality and morbidity, thus infection control measures are of paramount importance. Despite the advances in detecting viral pathogens, limited progress has been made in antiviral treatments, contributing to unexpectedly high rates of unfavorable outcomes. Herein, we review the most updated data on epidemiology, common clinical features, diagnosis, pathogenesis, treatment and prevention of severe community- and hospital-acquired viral infections in the ICU settings.
Collapse
Affiliation(s)
- Paraskevi C. Fragkou
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| | - Charalampos D. Moschopoulos
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| | - Emmanouil Karofylakis
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| | - Theodoros Kelesidis
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| |
Collapse
|
48
|
Schultz MJ, Deen J, von Seidlein L, Sivakorn C, Kumar A, Adhikari NKJ, Wills B, Kekulé AS, Day NPJ, Cooper B, White NJ, Salam AP, Horby P, Olliaro P, Dondorp AM. Remote-Controlled and Pulse Pressure-Guided Fluid Treatment for Adult Patients with Viral Hemorrhagic Fevers. Am J Trop Med Hyg 2021; 104:1172-1175. [PMID: 33591934 PMCID: PMC8045617 DOI: 10.4269/ajtmh.20-1515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/17/2020] [Indexed: 11/08/2022] Open
Abstract
Circulatory shock, caused by severe intravascular volume depletion resulting from gastrointestinal losses and profound capillary leak, is a common clinical feature of viral hemorrhagic fevers, including Ebola virus disease, Marburg hemorrhagic fever, and Lassa fever. These conditions are associated with high case fatality rates, and they carry a significant risk of infection for treating personnel. Optimized fluid therapy is the cornerstone of management of these diseases, but there are few data on the extent of fluid losses and the severity of the capillary leak in patients with VHFs, and no specific guidelines for fluid resuscitation and hemodynamic monitoring exist. We propose an innovative approach for monitoring VHF patients, in particular suited for low-resource settings, facilitating optimizing fluid therapy through remote-controlled and pulse pressure–guided fluid resuscitation. This strategy would increase the capacity for adequate supportive care, while decreasing the risk for virus transmission to health personnel.
Collapse
Affiliation(s)
- Marcus J Schultz
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,2Department of Intensive Care, Amsterdam University Medical Centres, Amsterdam, The Netherlands.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jacqueline Deen
- 4Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines, Manila, Philippines
| | - Lorenz von Seidlein
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Chaisith Sivakorn
- 5Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Alex Kumar
- 6School of Population Health and Environmental Sciences, King's College London and Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Neill K J Adhikari
- 7Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| | - Bridget Wills
- 3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,8Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Alexander S Kekulé
- 9Institute for Medical Microbiology, Martin Luther University, Halle-Wittenberg, Germany
| | - Nicholas P J Day
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ben Cooper
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J White
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alex P Salam
- 3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,10United Kingdom Public Health Rapid Support Team, London, United Kingdom
| | - Peter Horby
- 3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,11ISARIC Global Support Centre, International Severe Acute Respiratory and Emerging Infection Consortium, Oxford, United Kingdom
| | - Piero Olliaro
- 3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,11ISARIC Global Support Centre, International Severe Acute Respiratory and Emerging Infection Consortium, Oxford, United Kingdom
| | - Arjen M Dondorp
- 1Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,2Department of Intensive Care, Amsterdam University Medical Centres, Amsterdam, The Netherlands.,3Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Alfsnes K, Lagerqvist N, Vene S, Bohlin J, Verner-Carlsson J, Ekqvist D, Bråve A, Holmes EC, Shi W, Pettersson JHO. Retrospective meta-transcriptomic identification of severe dengue in a traveller returning from Africa to Sweden, 1990. One Health 2021; 12:100217. [PMID: 33553563 PMCID: PMC7851179 DOI: 10.1016/j.onehlt.2021.100217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
Pathogens associated with haemorrhagic fever commonly have zoonotic origins. The first documented imported case of likely viral severe haemorrhagic fever in Sweden occurred in 1990. Despite extensive study, no aetiological agent was identified. Following retrospective investigation with total RNA-sequencing of samples collected between 7 and 36 days from onset of symptoms we identified dengue virus 3 (DENV-3) and a human pegivirus (HPgV). We conclude that the patient likely suffered from haemorrhagic symptoms due to an atypical severe and undiagnosed dengue infection.
Collapse
Affiliation(s)
- Kristian Alfsnes
- Infectious Disease Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Nina Lagerqvist
- Public Health Agency of Sweden, Nobels väg 18, SE-171 82 Solna, Sweden
| | - Sirkka Vene
- Public Health Agency of Sweden, Nobels väg 18, SE-171 82 Solna, Sweden
| | - Jon Bohlin
- Infectious Disease Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | | | - David Ekqvist
- Department of Infectious Diseases, University Hospital Linköping, Sweden
| | - Andreas Bråve
- Public Health Agency of Sweden, Nobels väg 18, SE-171 82 Solna, Sweden
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, the University of Sydney, Sydney, New South Wales 2006, Australia
| | - Weifeng Shi
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, China
| | - John H-O Pettersson
- Public Health Agency of Sweden, Nobels väg 18, SE-171 82 Solna, Sweden.,Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, the University of Sydney, Sydney, New South Wales 2006, Australia.,Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| |
Collapse
|
50
|
Furuse Y. [Epidemiology of Viral Hemorrhagic Fever in Africa]. Uirusu 2021; 71:11-18. [PMID: 35526990 DOI: 10.2222/jsv.71.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A variety of viral hemorrhagic fevers such as Ebola virus disease exist in Africa and impose a great threat in public health due to their high fatality. It is considered to be difficult to eradicate the etiological agents of viral hemorrhagic fever because they have non-human natural hosts. Therefore, the importance of public health measures remains high in addition to the urgent need for the development of medicines for treatment and prevention. Furthermore, public health measures directly lead to the accumulation of epidemiological knowledge about the diseases. As an infectious disease consultant for the World Health Organization, I have been involved with public health activities including the development of clinical guidelines, the establishment of laboratory diagnostic systems, the training for infection, prevention and control, the planning of budget for outbreak response, and the analysis of epidemiological data. On the last point, I reported the situation of Ebola virus disease outbreak in Liberia, 2014-2015 and Lassa fever outbreak in Nigeria, 2018-2019 describing the risk factors, morbidity, and mortality of the diseases.
Collapse
Affiliation(s)
- Yuki Furuse
- Institute for Frontier Life and Medical Sciences, Kyoto University
- Hakubi Center for Advanced Research, Kyoto University
| |
Collapse
|