1
|
Valayer S, Alexandre M, Prague M, Beavogui AH, Doumbia S, Kieh M, Greenwood B, Leigh B, Poupelin M, Schwimmer C, Sow SO, Berry IM, Kuhn JH, Fusco D, Cauwelaert ND, Watson-Jones D, Thiébaut R, Lévy Y, Yazdanpanah Y, Richert L, Lhomme E. Evaluation of waning of IgG antibody responses after rVSVΔG-ZEBOV-GP and Ad26.ZEBOV, MVA-BN-Filo Ebola virus disease vaccines: a modelling study from the PREVAC randomized trial. Emerg Microbes Infect 2025; 14:0. [PMID: 39559990 PMCID: PMC11632942 DOI: 10.1080/22221751.2024.2432353] [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: 07/16/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
rVSVΔG-ZEBOV-GP and Ad26.ZEBOV, MVA-BN-Filo are WHO-prequalified vaccination regimens against Ebola virus disease (EVD). Challenges associated with measuring long-term clinical protection warrant the evaluation of immune response kinetics after vaccination. Data from a large phase 2 randomized double-blind clinical trial (PREVAC) were used to evaluate waning of anti-Ebola virus (EBOV) glycoprotein (GP1,2) antibody concentrations after rVSVΔG-ZEBOV-GP or Ad26.ZEBOV, MVA-BN-Filo vaccination with linear mixed-effect regression models. After a post-vaccination peak, each vaccination strategy was associated with a decrease of anti-EBOV GP1,2 antibody concentrations with distinct kinetics, highlighting a less-rapid decline in antibody levels after vaccination by rVSVΔG-ZEBOV-GP. One year after administration of the vaccine, antibody concentrations were higher in children compared to adults for both vaccines, although with different effect sizes: 1.74-fold higher concentrations (95% confidence interval [CI] [1.48; 2.02]) for children 12-17 years old to 3.10-fold higher concentrations (95% CI [2.58; 3.69]) for those 1-4 years old compared to adults for Ad26.ZEBOV, MVA-BN-Filo versus 1.36-fold (95% CI [1.12; 1.61]) to 1.41-fold (95% CI [1.21; 1.62]) higher than these values for adults, with relatively small changes from one age category of children to another, for rVSVΔG-ZEBOV-GP. Antibody concentrations also differed according to geographical location, pre-vaccination antibody concentration, and sex. In combination with knowledge on memory response, characterization of the major determinants of immune response durability of both vaccinations may guide future EVD control protocols.Trial registration: ClinicalTrials.gov identifier: NCT02876328.
Collapse
Affiliation(s)
- Simon Valayer
- Infection, Antimicrobials, Modelling, Evolution (IAME), Université Sorbonne Paris Nord, Université Sorbonne Paris Cité, and Institut national de la santé et de la recherche médicale (Inserm), Paris, France
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
| | - Marie Alexandre
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
| | - Mélanie Prague
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
| | - Abdoul Habib Beavogui
- Centre National de Formation et de Recherche en Santé Rurale (CNFRSR) de Mafèrinyah, Ministère de la Santé et de l’Hygiène Publique, Mafèrinyah, Guinea
| | - Seydou Doumbia
- University Clinical Research Center, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Mark Kieh
- Partnership for Research on Ebola Virus in Liberia (PREVAIL), Monrovia, Liberia
| | - Brian Greenwood
- London School of Hygiene & Tropical Medicine (LSHTM), London, UK
| | - Bailah Leigh
- College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Marie Poupelin
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
| | - Christine Schwimmer
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
| | - Samba O. Sow
- Centre pour le Développement des Vaccins, Ministère de la Santé et du Développement Social du Mali, Bamako, Mali
| | - Irina Maljkovic Berry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, USA
| | - Daniela Fusco
- French Agency for Research on AIDS and Viral Hepatitis (ANRS), Emerging Infectious Diseases, PariSanté Campus, Paris, France
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Rodolphe Thiébaut
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- Inserm, Paris Cedex 13, France
- Service d’Immunologie Clinique et Maladies Infectieuses, Hôpital Henri-Mondor, Créteil, France
| | - Yazdan Yazdanpanah
- Infection, Antimicrobials, Modelling, Evolution (IAME), Université Sorbonne Paris Nord, Université Sorbonne Paris Cité, and Institut national de la santé et de la recherche médicale (Inserm), Paris, France
- Inserm, Paris Cedex 13, France
- Service de Maladies Infectieuses et Tropicales, Hôpital Bichat, Paris, France
| | - Laura Richert
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Edouard Lhomme
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - the PREVAC Study Team
- Edouard Lhomme SISTM Team – Inserm U1219 Bordeaux Population Health, 146 Rue Léo Saignat, 33000Bordeaux, France
| |
Collapse
|
2
|
Lopez S, Komarova NL. An optimal network that promotes the spread of an advantageous variant in an SIR epidemic. J Theor Biol 2025; 605:112095. [PMID: 40107346 DOI: 10.1016/j.jtbi.2025.112095] [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: 11/14/2024] [Revised: 02/01/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
In the course of epidemics, the pathogen may mutate to acquire a higher fitness. At the same time, such a mutant is automatically in an unfavorable position because the resident virus has a head start in accessing the pool of susceptible individuals. We considered a class of tunable small-world networks, where a parameter, p (the rewiring probability), characterizes the prevalence of non-local connections, and we asked, whether the underlying network can influence the fate of a mutant virus. Under an SIR model, we considered two measures of mutant success: the expected height of the peak of mutant infected individuals, and the total number of recovered from mutant individuals at the end of the epidemic. Using these measures, we have found the existence of an optimal (for an advantageous mutant virus) rewiring probability that promotes a larger infected maximum and a larger total recovered population corresponding to the advantageous pathogen strain. This optimal rewiring probability decreases as mean degree and the infectivity of the wild type are increased, and it increases with the mutant advantage. The non-monotonic behavior of the advantageous mutant as a function of rewiring probability may shed light into some of the complex patterns in the size of mutant peaks experienced by different countries during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Samuel Lopez
- Department of Mathematics, University of California Irvine, Irvine, CA, 92617, United States
| | - Natalia L Komarova
- Department of Mathematics, University of California San Diego, La Jolla, CA, 92093, United States.
| |
Collapse
|
3
|
Sigal A, Neher RA, Lessells RJ. The consequences of SARS-CoV-2 within-host persistence. Nat Rev Microbiol 2025; 23:288-302. [PMID: 39587352 DOI: 10.1038/s41579-024-01125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
SARS-CoV-2 causes an acute respiratory tract infection that resolves in most people in less than a month. Yet some people with severely weakened immune systems fail to clear the virus, leading to persistent infections with high viral titres in the respiratory tract. In a subset of cases, persistent SARS-CoV-2 replication results in an accelerated accumulation of adaptive mutations that confer escape from neutralizing antibodies and enhance cellular infection. This may lead to the evolution of extensively mutated SARS-CoV-2 variants and introduce an element of chance into the timing of variant evolution, as variant formation may depend on evolution in a single person. Whether long COVID is also caused by persistence of replicating SARS-CoV-2 is controversial. One line of evidence is detection of SARS-CoV-2 RNA and proteins in different body compartments long after SARS-CoV-2 infection has cleared from the upper respiratory tract. However, thus far, no replication competent virus has been cultured from individuals with long COVID who are immunocompetent. In this Review, we consider mechanisms of viral persistence, intra-host evolution in persistent infections, the connection of persistent infections with SARS-CoV-2 variants and the possible role of SARS-CoV-2 persistence in long COVID. Understanding persistent infections may therefore resolve much of what is still unclear in COVID-19 pathophysiology, with possible implications for other emerging viruses.
Collapse
Affiliation(s)
- Alex Sigal
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Richard A Neher
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Richard J Lessells
- KwaZulu-Natal Research Innovation & Sequencing Platform, School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
4
|
Mani V, Chu WT, Yang HJ, Morris CP, Laux J, Byrum R, Cooper K, Liu DX, Wang H, Johnson C, Hadley K, Bernbaum JG, Hart R, Anthony SM, Marketon AE, Bernbaum-Cutler R, Pahar B, Worwa G, Kuhn JH, Crozier I, Calcagno C, Gale E. Reactive oxygen species-related oxidative changes are associated with splenic lymphocyte depletion in Ebola virus infection. NPJ IMAGING 2025; 3:16. [PMID: 40291761 PMCID: PMC12021656 DOI: 10.1038/s44303-025-00079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
The dysregulated production of reactive oxygen species (ROS) during viral infections may lead to immune cell death and ineffective host responses. ROS dynamics have been under-investigated in severe Ebola virus disease (EVD), a condition in which hyperinflammation and excessive immune cell death are well described but poorly understood. Through ex vivo immunohistochemistry and in vivo ROS-sensitive magnetic resonance imaging (MRI) we demonstrate significant ROS-related oxidative changes in the spleens of domestic ferrets exposed to Ebola virus (EBOV). By immunohistochemistry or MRI, detection of splenic ROS was inversely correlated with the number of CD4+/CD8+ T lymphocytes and apoptotic CD8+ lymphocytes, but detection was positively correlated with the frequency of apoptotic CD4+ cells and the number and frequency of apoptotic B lymphocytes. These results suggest that ROS-induced apoptosis may contribute to the loss of splenic CD4+ T lymphocytes in EBOV-exposed ferrets and warrant further investigation of the role of ROS in severe EVD.
Collapse
Affiliation(s)
- Venkatesh Mani
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Winston T. Chu
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Hee-Jeong Yang
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - C. Paul Morris
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Joseph Laux
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Russell Byrum
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - David X. Liu
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Hui Wang
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Cristal Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Kyra Hadley
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - John G. Bernbaum
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Scott M. Anthony
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Anthony E. Marketon
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Rebecca Bernbaum-Cutler
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Bapi Pahar
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Gabriella Worwa
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Claudia Calcagno
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Eric Gale
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA USA
- Harvard Medical School, Boston, MA USA
| |
Collapse
|
5
|
Muwonge H, Nasimiyu C, Bakamutumaho B, Elyanu P, Joloba ML, Situma S, Schieffelin J, Gunn B, Bai S, Breiman RF, Ssewanyana I, Nabadda S, Lutwama J, Tegen Y, Muruta A, Kirenga B, Olaro C, Aceng JR, Bosa HK, Njenga MK. Severe long-term clinical sequelae among Sudan ebolavirus disease survivors 2 years post-infection. RESEARCH SQUARE 2025:rs.3.rs-6325522. [PMID: 40321748 PMCID: PMC12047994 DOI: 10.21203/rs.3.rs-6325522/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Background While long-term clinical sequelae following ebolavirus disease (EVD) due to Zaire ebolavirus (EBOV) strain has been characterized, this has not been explored for Sudan ebolavirus (SUDV) strain. Methods We enrolled 87 SUDV survivors from the 2022-2023 outbreak in Uganda, alongside 176 age-, sex-, and location-matched controls. Clinical symptom data were collected at 3-, 9-, 12-, 15-, and 18-and 24-months post-infection. Serum, semen, and breast milk samples were collected and tested for viral RNA. Results Of 86 SUDV survivors, 57.5% reported significantly higher frequencies of clinical symptoms involving musculoskeletal (45.0%, P < 0.001), central nervous system (36.3%, p < 0.001), ophthalmologic (20%, P < 0.001), and respiratory (10%, P < 0.001) systems than those observed among controls. The risk ratio of occurrence was highest for ophthalmologic (20% vs 3.4%, RR = 5.9; p < 0.001) and central nervous systems symptoms (36.3% vs 6.8%, RR = 5.3, p < 0.001), and lowest for reproductive system (13.8% vs 8.5%; RR = 1.6; p > 0.005). Importantly, 50% of SUDV survivors reported persistent multi-systemic symptoms, including low back pain, hand and feet numbness, confusion, and diarrhoea that resulted in inability to perform basic activities of living. Viral RNA was detected in semen for a median duration of 131 days (range: 111-210 days) and in breast milk for a median of 149 days (range: 111-199 days). Conclusions This study demonstrates that SUDV survivors develop long-term clinical sequelae characterized by persistent multi-systemic clinical symptoms. Detection of viral RNA in semen and breastmilk for up to 7 months post-infection suggest prolonged persistence, with the possibility of latency and reactivation of the virus.
Collapse
|
6
|
Ntemafack A, Dzelamonyuy A, Nchinda G, Bopda Waffo A. Mapping Immunological, Host Receptor Binding Determinants, and Cathepsin Cleavage Site of EBOV Glycoprotein Utilizing the Qubevirus Platform. ACS OMEGA 2025; 10:14283-14295. [PMID: 40256529 PMCID: PMC12004191 DOI: 10.1021/acsomega.5c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 04/22/2025]
Abstract
Ebola virus (EBOV) remains a highly infectious human pathogen that causes a severe and lethal disease known as Ebola virus disease (EVD), despite recent progress in vaccine development based on its only surface glycoprotein (GP). In this study, we modeled and inserted four overlapping fragments (F1-4) of the EBOV GP at the C-terminus of the A1 protein of Qubevirus (Qβ) and used the platform to investigate the tropism and immunological functions of the GP by displaying the peptides with 30 overlapping amino acids. The resulting recombinant phages were used to determine their reactivity with GP-specific antibodies and their binding to the recombinant Niemann-Pick C1 (rNPC1) receptor in an immunoassay. In addition, modified, truncated, and C-terminus-tagged fragment F1 named F5 was utilized to map the cathepsin cleavage sites in an enzymatic assay. We demonstrated that a large GP peptide of 200 AA could be fused to A1 and exposed on the Qβ platform in an accessible manner without significantly affecting its viability and infectivity. Fragments F1 (GP1-200), F2 (GP170-370), and F3 (GP350-550) were shown to contain important immune epitopes through binding to anti-GP-specific antibodies. Further, F1 was found to bind rNPC1, thereby suggesting a receptor binding determinant of the GP that was further confirmed in a competitive assay where the recombinant phages bearing the F1 fragment reduced the infectivity of EBOV pseudovirus by 27%. In addition, the viral infectivity was shown to be reduced by 46.39% by a cyclic peptide selected from an RNA Qβ library. Finally, F5 showed the cleavage sites to be AA191-192 and AA194-195 for CatB and L, respectively, which were further validated using a recombinant EBOV glycoprotein. These results provide insights into the antigenicity and tropism characteristic of the glycoprotein, with implications for the development of subunit vaccines or other biologics against Ebola virus disease.
Collapse
Affiliation(s)
- Augustin Ntemafack
- Department
of Biochemistry and Molecular Biology, Indiana
University School of Medicine, 635 Barnhill, Indianapolis, Indiana46202-3082, United States
| | - Aristide Dzelamonyuy
- Department
of Biochemistry and Molecular Biology, Indiana
University School of Medicine, 635 Barnhill, Indianapolis, Indiana46202-3082, United States
| | - Godwin Nchinda
- Laboratory
of Vaccinology and Biobanking, CIRCB, BP 3077 Messa, Yaoundé 03077, Cameroon
| | - Alain Bopda Waffo
- Department
of Biochemistry and Molecular Biology, Indiana
University School of Medicine, 635 Barnhill, Indianapolis, Indiana46202-3082, United States
| |
Collapse
|
7
|
Bowers KH, De Angelis D, Birrell PJ. Modelling with SPEED: a Stochastic Predictor of Early Epidemic Detection. J Theor Biol 2025; 607:112120. [PMID: 40189138 DOI: 10.1016/j.jtbi.2025.112120] [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: 08/30/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 04/20/2025]
Abstract
The frequency of emerging infectious disease outbreaks continues to rise, necessitating predictive frameworks for public health decision-making. This study introduces the Stochastic Predictor of Early Epidemic Detection (SPEED) model, an adaptation of the classic Susceptible-Infected-Recovered model, employing a Gillespie-like algorithm to simulate early-stage stochastic disease transmission. SPEED incorporates individual-level detection probabilities based on the infection time and the lag from GP consultation to lab confirmation. The model dynamically adjusts to public health responses by enhancing testing and reducing detection times once a single case has been identified. SPEED serves two key functionalities. First, as a statistical inference tool refining reproduction number estimates following the detection of a small number of cases. SPEED inference uses specified prior distributions for the reproduction number to provide reliable posterior estimates. Second, to simulate epidemic scenarios under specified values of the reproduction number in order to construct a distribution of the time to subsequent detections. The model is used to evaluate how second case timings can rule out higher values of the reproduction number. Comparisons with simulations under heightened surveillance scenarios demonstrate the model's utility in assessing response efficacy on the initial outbreak spread. Our results demonstrate SPEED applied to a single case of influenza A(H1N2)v, detected through routine flu surveillance on the 23rd November 2023.
Collapse
Affiliation(s)
| | - Daniela De Angelis
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK
| | - Paul J Birrell
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK.
| |
Collapse
|
8
|
Marzi A. One-for-one or one-for-all? Considerations for filovirus vaccine development. PLoS Biol 2025; 23:e3003142. [PMID: 40228023 PMCID: PMC12021285 DOI: 10.1371/journal.pbio.3003142] [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: 03/05/2025] [Revised: 04/24/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
Filoviruses continue to re-emerge in Africa, causing localized public health emergencies. Although vaccination has slowly been implemented for Ebola virus, not for other filoviruses, holistic approaches are needed to broadly protect against filovirus threats.
Collapse
Affiliation(s)
- Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
9
|
Fletcher P, O'Donnell KL, Feldmann F, Rhoderick JF, Clancy CS, Prator CA, Smith BJ, Gunn BM, Feldmann H, Marzi A. Single-dose VSV-Sudan virus vaccine protects from lethal Sudan virus infection within one week: a challenge study in macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645555. [PMID: 40196593 PMCID: PMC11974824 DOI: 10.1101/2025.03.26.645555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background The Sudan virus (SUDV) outbreaks in recent years including the ongoing outbreak in Uganda created a public health emergency beyond the Eastern Africa region. Currently, there are licensed countermeasures for Ebola virus (EBOV); however, there are no licensed vaccines or therapeutics against SUDV. Methods We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the SUDV glycoprotein. Cynomolgus macaques were vaccinated intramuscularly with a single dose of VSV-SUDV either one month or one week prior to SUDV challenge. A third group was vaccinated with a single dose of VSV-EBOV one month prior to SUDV challenge to assess its cross-protective potential, and a control group received an unrelated VSV-based vaccine. Results All vaccinated nonhuman primates (NHPs) developed antigen-specific IgG within 2 weeks of vaccination, including cross-reactive responses. After challenge with a lethal dose of SUDV, all VSV-SUDV-vaccinated NHPs were uniformly protected from disease. In contrast, the VSV-EBOV-vaccinated and control NHPs succumbed to disease between day 5 and 7 after challenge presenting with classical signs of Sudan virus disease associated with high titer viremia, high viral organ load, dysregulated cytokine profiles and typical pathological changes. The humoral immune response in the NHPs vaccinated with VSV-SUDV one month before challenge resulted in a profound and sustained antibody response with a diverse functionality profile which was not observed to the same extend in NHPs vaccinated one week before challenge. Interpretation We demonstrated that a single dose of VSV-SUDV protected NHPs from lethal SUDV infection within one week. The fast-acting nature makes VSV-SUDV an ideal countermeasure for ring vaccination during outbreaks of Sudan virus disease. In contrast, VSV-EBOV provided no relevant protection against SUDV infection in NHPs highlighting the need for species-specific filovirus vaccines.
Collapse
|
10
|
van Tol S, Port JR, Fischer RJ, Gallogly S, Bushmaker T, Griffin A, Schulz JE, Carmody A, Myers L, Crowley DE, Falvo CA, Riopelle JC, Wickenhagen A, Clancy C, Lovaglio J, Shaia C, Saturday G, Prado-Smith J, He Y, Lack J, Martens C, Anzick SL, Kendall LV, Schountz T, Plowright RK, Marzi A, Munster VJ. Jamaican fruit bats' competence for Ebola but not Marburg virus is driven by intrinsic differences. Nat Commun 2025; 16:2884. [PMID: 40133326 PMCID: PMC11937316 DOI: 10.1038/s41467-025-58305-4] [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: 11/19/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Ebola virus (EBOV) and Marburg virus (MARV) are zoonotic filoviruses that cause hemorrhagic fever in humans. Correlative data implicate bats as natural EBOV hosts, but neither a full-length genome nor an EBOV isolate has been found in any bats sampled. Here, we model filovirus infection in the Jamaican fruit bat (JFB), Artibeus jamaicensis, by inoculation with either EBOV or MARV through a combination of oral, intranasal, and subcutaneous routes. Infection with EBOV results in systemic virus replication and oral shedding of infectious virus. MARV replication is transient and does not shed. In vitro, JFB cells replicate EBOV more efficiently than MARV, and MARV infection induces innate antiviral responses that EBOV efficiently suppresses. Experiments using VSV pseudoparticles or replicating VSV expressing the EBOV or MARV glycoprotein demonstrate an advantage for EBOV entry and replication early, respectively, in JFB cells. Overall, this study describes filovirus species-specific phenotypes for both JFB and their cells.
Collapse
Affiliation(s)
- Sarah van Tol
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julia R Port
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Laboratory of Transmission Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert J Fischer
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Shane Gallogly
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Trenton Bushmaker
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Amanda Griffin
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan E Schulz
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Aaron Carmody
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Lara Myers
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Daniel E Crowley
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Caylee A Falvo
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jade C Riopelle
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Arthur Wickenhagen
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Chad Clancy
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jessica Prado-Smith
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Craig Martens
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Sarah L Anzick
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Lon V Kendall
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Tony Schountz
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Raina K Plowright
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Andrea Marzi
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
11
|
Almulhim M, Ghasemian A, Memariani M, Karami F, Yassen ASA, Alexiou A, Papadakis M, Batiha GES. Drug repositioning as a promising approach for the eradication of emerging and re-emerging viral agents. Mol Divers 2025:10.1007/s11030-025-11131-8. [PMID: 40100484 DOI: 10.1007/s11030-025-11131-8] [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: 12/02/2024] [Accepted: 02/08/2025] [Indexed: 03/20/2025]
Abstract
The global impact of emerging and re-emerging viral agents during epidemics and pandemics leads to serious health and economic burdens. Among the major emerging or re-emerging viruses include SARS-CoV-2, Ebola virus (EBOV), Monkeypox virus (Mpox), Hepatitis viruses, Zika virus, Avian flu, Influenza virus, Chikungunya virus (CHIKV), Dengue fever virus (DENV), West Nile virus, Rhabdovirus, Sandfly fever virus, Crimean-Congo hemorrhagic fever (CCHF) virus, and Rift Valley fever virus (RVFV). A comprehensive literature search was performed to identify existing studies, clinical trials, and reviews that discuss drug repositioning strategies for the treatment of emerging and re-emerging viral infections using databases, such as PubMed, Scholar Google, Scopus, and Web of Science. By utilizing drug repositioning, pharmaceutical companies can take advantage of a cost-effective, accelerated, and effective strategy, which in turn leads to the discovery of innovative treatment options for patients. In light of antiviral drug resistance and the high costs of developing novel antivirals, drug repositioning holds great promise for more rapid substitution of approved drugs. Main repositioned drugs have included chloroquine, ivermectin, dexamethasone, Baricitinib, tocilizumab, Mab114 (Ebanga™), ZMapp (pharming), Artesunate, imiquimod, saquinavir, capmatinib, naldemedine, Trametinib, statins, celecoxib, naproxen, metformin, ruxolitinib, nitazoxanide, gemcitabine, Dorzolamide, Midodrine, Diltiazem, zinc acetate, suramin, 5-fluorouracil, quinine, minocycline, trifluoperazine, paracetamol, berbamine, Nifedipine, and chlorpromazine. This succinct review will delve into the topic of repositioned drugs that have been utilized to combat emerging and re-emerging viral pathogens.
Collapse
Affiliation(s)
- Marwa Almulhim
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | - Mojtaba Memariani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Farnaz Karami
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Asmaa S A Yassen
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
12
|
Handa T, Saha A, Narayanan A, Ronzier E, Kumar P, Singla J, Tomar S. Structural Virology: The Key Determinants in Development of Antiviral Therapeutics. Viruses 2025; 17:417. [PMID: 40143346 PMCID: PMC11945554 DOI: 10.3390/v17030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Structural virology has emerged as the foundation for the development of effective antiviral therapeutics. It is pivotal in providing crucial insights into the three-dimensional frame of viruses and viral proteins at atomic-level or near-atomic-level resolution. Structure-based assessment of viral components, including capsids, envelope proteins, replication machinery, and host interaction interfaces, is instrumental in unraveling the multiplex mechanisms of viral infection, replication, and pathogenesis. The structural elucidation of viral enzymes, including proteases, polymerases, and integrases, has been essential in combating viruses like HIV-1 and HIV-2, SARS-CoV-2, and influenza. Techniques including X-ray crystallography, Nuclear Magnetic Resonance spectroscopy, Cryo-electron Microscopy, and Cryo-electron Tomography have revolutionized the field of virology and significantly aided in the discovery of antiviral therapeutics. The ubiquity of chronic viral infections, along with the emergence and reemergence of new viral threats necessitate the development of novel antiviral strategies and agents, while the extensive structural diversity of viruses and their high mutation rates further underscore the critical need for structural analysis of viral proteins to aid antiviral development. This review highlights the significance of structure-based investigations for bridging the gap between structure and function, thus facilitating the development of effective antiviral therapeutics, vaccines, and antibodies for tackling emerging viral threats.
Collapse
Affiliation(s)
- Tanuj Handa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Ankita Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Aarthi Narayanan
- Department of Biology, College of Science, George Mason University, Fairfax, VA 22030, USA;
| | - Elsa Ronzier
- Biomedical Research Laboratory, Institute for Biohealth Innovation, George Mason University, Fairfax, VA 22030, USA;
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Jitin Singla
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| |
Collapse
|
13
|
Musoke D, Atusingwize E, Robins A, Nam S, Bonwitt J, Msukwa C, Rutayisire M, Upenytho G, Kyobe H, Mwebesa H. Barriers to community engagement during the response to an Ebola virus disease outbreak in Uganda. BMJ Glob Health 2025; 10:e017285. [PMID: 40068928 PMCID: PMC11904350 DOI: 10.1136/bmjgh-2024-017285] [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: 08/22/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Uganda reported an outbreak of Ebola virus disease (EVD) in 2022. As part of the outbreak response, government and partners promoted community engagement, which seeks to involve communities in the design, implementation and evaluation of interventions to raise awareness, build trust between communities and partners and create ownership of interventions. This study, therefore, explored barriers to community engagement during the 2022-2023 EVD outbreak response in Uganda. METHODS This qualitative study, conducted in five districts (Kampala, Kassanda, Kyegegwa, Mubende and Wakiso), involved 25 focus group discussions among community members and community health workers (CHWs). In addition, 32 key informant interviews were conducted with staff from the Uganda Ministry of Health, district health officials, local leaders, non-governmental organisation staff and other stakeholders. Data were analysed according to the thematic approach using ATLAS.ti (V.6). RESULTS The main barriers to community engagement identified during the EVD outbreak response are presented under four main themes: (1) delayed consultations between partners and communities; (2) poor communication and misinformation; (3) limited support to human resources; and (4) institutional and coordination challenges. Specifically, these barriers included: limited consultation due to misbelief in community roles; delayed sociocultural discussions; stigma and delayed psychosocial interventions; misinformation, rumours and political influence; poor communication mechanisms; contradictory messages and lack of transparency; language barrier and inappropriate communication media; work overload for CHWs and other community volunteers; failure to prioritise protection of community workers; lack of compensation for CHWs and other community personnel; poor logistical management; inadequate coordination and partner operations; unfavourable institutional structures; and limited funding for emergencies. CONCLUSION The barriers encountered in community engagement during the 2022-2023 EVD outbreak need to be addressed through strengthening guidelines and standard operating procedures, capacity building for partners and communities, as well as adequate financing to ensure Uganda is better prepared for future health emergencies.
Collapse
Affiliation(s)
- David Musoke
- Disease Control and Environmental Health, Makerere University School of Public Health, Kampala, Uganda
| | - Edwinah Atusingwize
- Disease Control and Environmental Health, Makerere University School of Public Health, Kampala, Uganda
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Godard A, Phalkey R, Shepherd S, Rossi S, Tessema MT, Lee JS. Medical oxygen: a necessity or a luxury in humanitarian settings? Lancet Glob Health 2025; 13:e392-e393. [PMID: 39978386 DOI: 10.1016/s2214-109x(24)00509-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/21/2024] [Indexed: 02/22/2025]
Affiliation(s)
| | | | - Susan Shepherd
- Alliance for International Medical Action, New York, NY, USA
| | - Sara Rossi
- Direct Relief International, Santa Barbara, CA, USA
| | | | | |
Collapse
|
15
|
Shurtleff AC, Trefry JC, Dubey S, Sunay MME, Liu K, Chen Z, Eichberg M, Silvera PM, Kwilas SA, Hooper JW, Martin S, Simon JK, Coller BAG, Monath TP. rVSVΔG-ZEBOV-GP Vaccine Is Highly Immunogenic and Efficacious Across a Wide Dose Range in a Nonhuman Primate EBOV Challenge Model. Viruses 2025; 17:341. [PMID: 40143273 PMCID: PMC11945660 DOI: 10.3390/v17030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
The recombinant vesicular stomatitis virus-Zaire Ebolavirus envelope glycoprotein vaccine (rVSVΔG-ZEBOV-GP) was highly effective against Ebola virus disease in a ring vaccination trial conducted during the 2014-2016 outbreak in Guinea and is licensed by regulatory agencies including US FDA, EMA, and prequalified by WHO. Vaccination studies in a nonhuman primate (NHP) model guided initial dose selection for clinical trial evaluation. We summarize two dose-ranging studies with the clinical-grade rVSVΔG-ZEBOV-GP vaccine candidate to assess the impact of dose level on immune responses and efficacy in an NHP Ebola virus (EBOV) challenge model. Forty-six cynomolgus macaques were vaccinated with a wide range of rVSVΔG-ZEBOV-GP doses and challenged 42 days later intramuscularly with 1000 pfu EBOV. Vaccination with rVSVΔG-ZEBOV-GP induced relatively high levels of EBOV-specific IgG and neutralizing antibodies, measured using the same validated assays as used in rVSVΔG-ZEBOV-GP clinical trials. Similar responses were observed across dose groups from 1 × 108 to 1 × 102 pfu. A single vaccination conferred 98% protection from lethal intramuscular EBOV challenge across all dose groups. These results demonstrate that robust antibody titers are induced in NHPs across a wide range of rVSVΔG-ZEBOV-GP vaccine doses, correlating with high levels of protection against death from EBOV challenge.
Collapse
MESH Headings
- Animals
- Ebola Vaccines/immunology
- Ebola Vaccines/administration & dosage
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/immunology
- Ebolavirus/immunology
- Ebolavirus/genetics
- Macaca fascicularis
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Disease Models, Animal
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/administration & dosage
- Vaccination/methods
- Male
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Female
- Vaccine Efficacy
- Vesiculovirus/genetics
- Vesiculovirus/immunology
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Amy C. Shurtleff
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - John C. Trefry
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Sheri Dubey
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Melek M. E. Sunay
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Kenneth Liu
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Ziqiang Chen
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Michael Eichberg
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Peter M. Silvera
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Steve A. Kwilas
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Jay W. Hooper
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Shannon Martin
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Porter Street, Fort Detrick, Frederick, MD 21702, USA; (A.C.S.); (J.C.T.); (M.M.E.S.); (P.M.S.); (S.A.K.); (J.W.H.); (S.M.)
| | - Jakub K. Simon
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Beth-Ann G. Coller
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | | |
Collapse
|
16
|
Bixler SL, Shurtleff AC, Sunay MME, Liu K, Chen Z, Eichberg M, Simon JK, Coller BAG, Dubey S. Durability of Immunogenicity and Protection of rVSV∆G-ZEBOV-GP Vaccine in a Nonhuman Primate EBOV Challenge Model. Viruses 2025; 17:342. [PMID: 40143270 PMCID: PMC11945958 DOI: 10.3390/v17030342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
The rVSVΔG-ZEBOV-GP vaccine demonstrated efficacy in preventing Ebola virus (EBOV) disease in a ring vaccination clinical trial conducted during the 2014-2016 West Africa outbreak and is licensed by regulatory agencies, including the US FDA and the EMA. Here, we present two studies that evaluated the durability of immunogenicity and protection from an EBOV challenge up to ~12 months following vaccination with rVSVΔG-ZEBOV-GP in nonhuman primates (NHPs). Cynomolgus macaques were vaccinated with either one or two doses of rVSVΔG-ZEBOV-GP or a saline control and were challenged intramuscularly with EBOV at a target dose of 1000 pfu at ~4 months (Study 1) or ~8 or ~12 months (Study 2) after the last vaccination. All vaccinated animals developed robust ZEBOV-GP-specific IgG and neutralizing antibody titers, which were sustained until the last time point tested prior to the challenge. The majority of animals (88-93%) challenged with EBOV at ~4 or ~8 months post-vaccination survived, whereas the survival rate was lower (53%) in animals challenged ~12 months post-vaccination. These results demonstrate that both one-dose and two-dose regimens of the rVSVΔG-ZEBOV-GP vaccine induced durable ZEBOV-GP-specific antibody titers in NHPs and provided high levels of protection against a lethal EBOV challenge up to ~8 months post-vaccination. In this stringent challenge model, decreased protection was observed at ~12 months post-vaccination despite sustained antibody levels.
Collapse
Affiliation(s)
- Sandra L. Bixler
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Amy C. Shurtleff
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Melek M. E. Sunay
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Kenneth Liu
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Ziqiang Chen
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Michael Eichberg
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Jakub K. Simon
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Beth-Ann G. Coller
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Sheri Dubey
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| |
Collapse
|
17
|
Prescott JB, Liu KJ, Lander A, Pek NMQ, Jha SK, Bokelmann M, Begur M, Koh PW, Yang H, Lim B, Red-Horse K, Weissman IL, Loh KM, Ang LT. Metabolically purified human stem cell-derived hepatocytes reveal distinct effects of Ebola and Lassa viruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638665. [PMID: 40027809 PMCID: PMC11870522 DOI: 10.1101/2025.02.17.638665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Ebola and Lassa viruses require biosafety-level-4 (BSL4) containment, infect the liver, and cause deadly hemorrhagic fevers. The cellular effects of these viruses, and whether different families of hemorrhagic-fever viruses elicit similar effects, remain fundamental questions in BSL4 virology. Here, we introduce a new metabolic selection approach to create nearly-pure hepatocytes from human pluripotent stem cells, killing non-liver cells by withholding essential nutrients. Unexpectedly, Ebola and Lassa exerted starkly different effects on human hepatocytes. Ebola infection activated the integrated stress response (ISR) and WNT pathways in hepatocytes in vitro and killed them, whereas Lassa did not. Within non-human primates, Ebola likewise infected hepatocytes and activated ISR signaling in vivo . In summary, we present a single-cell transcriptional and chromatin accessibility roadmap of human hepatocyte differentiation, purification, and viral infection.
Collapse
|
18
|
Bu F, Ye G, Turner-Hubbard H, Herbst M, Liu B, Li F. Cryo-EM structure of Sudan ebolavirus glycoprotein complexed with its human endosomal receptor NPC1. Commun Biol 2025; 8:156. [PMID: 39894818 PMCID: PMC11788421 DOI: 10.1038/s42003-025-07613-y] [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: 11/21/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Sudan ebolavirus (SUDV), like Ebola ebolavirus (EBOV), poses a significant threat to global health and security due to its high lethality. However, unlike EBOV, there are no approved vaccines or treatments for SUDV, and its structural interaction with the endosomal receptor NPC1 remains unclear. This study compares the glycoproteins of SUDV and EBOV (in their proteolytically primed forms) and their binding to human NPC1 (hNPC1). The findings reveal that the SUDV glycoprotein binds significantly more strongly to hNPC1 than the EBOV glycoprotein. Using cryo-EM, we determined the structure of the SUDV glycoprotein/hNPC1 complex, identifying four key residues in the SUDV glycoprotein that differ from those in the EBOV glycoprotein and influence hNPC1 binding: Ile79, Ala141, and Pro148 enhance binding, while Gln142 reduces it. Collectively, these residue differences account for SUDV's stronger binding affinity for hNPC1. This study provides critical insights into receptor recognition across all viruses in the ebolavirus genus, including their interactions with receptors in bats, their suspected reservoir hosts. These findings advance our understanding of ebolavirus cell entry, tissue tropism, and host range.
Collapse
Affiliation(s)
- Fan Bu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Center for Emerging Viruses, University of Minnesota, Minneapolis, MN, USA
| | - Gang Ye
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Center for Emerging Viruses, University of Minnesota, Minneapolis, MN, USA
| | - Hailey Turner-Hubbard
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Center for Emerging Viruses, University of Minnesota, Minneapolis, MN, USA
| | - Morgan Herbst
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Center for Emerging Viruses, University of Minnesota, Minneapolis, MN, USA
| | - Bin Liu
- Hormel Institute, University of Minnesota, Austin, MN, USA.
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA.
- Center for Emerging Viruses, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
19
|
Khan A, Sayaf AM, Mohammad A, Alshabrmi FM, Benameur T, Wei DQ, Yeoh KK, Agouni A. Discovery of anti-Ebola virus multi-target inhibitors from traditional Chinese medicine database using molecular screening, biophysical investigation, and binding free energy calculations. J Infect Public Health 2025; 18:102636. [PMID: 39798213 DOI: 10.1016/j.jiph.2024.102636] [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: 05/21/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025] Open
Abstract
INTRODUCTION Ebola virus (EBOV) is a highly lethal RNA virus that causes severe hemorrhagic fever in humans and non-human primates. The lack of effective treatment or vaccine for this pathogen poses a serious threat to a global pandemic. Therefore, it is imperative to explore new drugs and therapies to combat this life-threatening infection. MATERIALS AND METHODS In this study, we employed in silico methods to assess the inhibitory activity of natural products from traditional Chinese medicine (TCM) against four EBOV proteins that are crucial for viral replication and assembly: VP40, VP35, VP30, and VP24. We performed molecular docking of TCM compounds with the EBOV proteins and screened them based on their docking scores, binding free energies, and pharmacokinetic properties. RESULTS Our results pinpointed eight TCM compounds (TCM1797, TCM2872, TCM250, TCM2837, TCM2644, TCM4697, TCM2322, and TCM277) that exhibited superior efficacy in inhibiting all the EBOV proteins compared to the controls. These compounds interacted with key residues of the EBOV proteins through various types of bonds, such as hydrogen bonds, salt bridges, and π-π interactions, forming stable complexes that could disrupt the function of the EBOV proteins. These compounds were found to possess known antiviral activity, acceptable pharmacokinetic properties, and human usage history, which make them promising candidates for anti-EBOV drug development. Moreover, the molecular simulation analysis confirmed the binding stability, structural compactness, and residue flexibility properties of these compounds. Furthermore, the binding free energy results revealed that VP30-TCM2644, VP30-TCM4697, VP35-TCM2837, VP24-TCM250, and VP24-TCM277 complexes exhibit significant binding free energy values compared to the control ligands. Principal Component Analysis (PCA) and Free Energy Landscape (FEL) results revealed the trajectories' motion and conformational energy states. CONCLUSIONS Our findings provide valuable insights into the molecular mechanisms driving the efficacy of TCM drugs against EBOV and suggest novel approaches for the development of anti-EBOV therapies.
Collapse
Affiliation(s)
- Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar; Division of Bioinformatics, Department of Biomedical Sciences, School of Medical and Life Sciences, Sunway University, 5, Jalan Universiti, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | | | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahad M Alshabrmi
- Department of Medical laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Kar Kheng Yeoh
- School of Chemical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
20
|
Mahmoodi S, Amirzakaria JZ, Ghasemian A. A novel multi-epitope peptide vaccine targeting immunogenic antigens of Ebola and monkeypox viruses with potential of immune responses provocation in silico. Biotechnol Appl Biochem 2025; 72:58-74. [PMID: 39128888 DOI: 10.1002/bab.2646] [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: 04/22/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024]
Abstract
The emergence or reemergence of monkeypox (Mpox) and Ebola virus (EBOV) agents causing zoonotic diseases remains a huge threat to human health. Our study aimed at designing a multi-epitope vaccine (MEV) candidate to target both the Mpox and EBOV agents using immunoinformatics tools. Viral protein sequences were retrieved, and potential nonallergenic, nontoxic, and antigenic epitopes were obtained. Next, cytotoxic and helper T-cell (CTL and HTL, respectively) and B-cell (BCL) epitopes were predicted, and those potential epitopes were fused utilizing proper linkers. The in silico cloning and expression processes were implemented using Escherichia coli K12. The immune responses were prognosticated using the C-ImmSim server. The MEV construct (29.53 kDa) included four BCL, two CTL, and four HTL epitopes and adjuvant. The MEV traits were pertinent in terms of antigenicity, non-allergenicity, nontoxicity, physicochemical characters, and stability. The MEV candidate was also highly expressed in E. coli K12. The strong affinity of MEV-TLR3 was confirmed using molecular docking and molecular dynamics simulation analyses. Immune simulation analyses unraveled durable activation and responses of cellular and humoral arms alongside innate immune responses. The designed MEV candidate demonstrated appropriate traits and was promising in the prediction of immune responses against both Mpox and EBOV agents. Further experimental assessments of the MEV are required to verify its efficacy.
Collapse
Affiliation(s)
- Shirin Mahmoodi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Javad Zamani Amirzakaria
- Department of Plant Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
21
|
Peng C, Wu F, Ma Y, Liu G, Huang Y, Tong R, Xu W. Ginkgolic acid inhibits Ebola virus transcription and replication by disrupting the interaction between nucleoprotein and VP30 protein. Antiviral Res 2025; 234:106074. [PMID: 39716669 DOI: 10.1016/j.antiviral.2024.106074] [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: 10/14/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
The Ebola virus, a filovirus, has been responsible for significant human fatalities since its discovery. Despite extensive research, effective small-molecule drugs remain elusive due to its complex pathogenesis. Inhibition of RNA synthesis is a promising therapeutic target, and the VP30 protein plays a critical role in this process. The interaction between VP30 and the nucleoprotein (NP) is essential for viral replication. We identified ginkgolic acid as a small molecule with strong affinity for VP30, which was validated through multiple assays, including thermal shift, surface plasmon resonance, fluorescence polarization, pull-down, and co-immunoprecipitation. The antiviral efficacy of ginkgolic acid was demonstrated in the EBOV transcription- and replication-competent virus-like particle (trVLP) system. Furthermore, we resolved the crystal structure of the VP30-ginkgolic acid complex, revealing two ginkgolic acid molecules located at the VP30/NP interaction interface. This structural information provides insight into the molecular basis of ginkgolic acid's antiviral activity and suggests a novel therapeutic strategy targeting the VP30/NP interaction.
Collapse
Affiliation(s)
- Chiwei Peng
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fang Wu
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Yanhong Ma
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Guolong Liu
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yin Huang
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Rongbiao Tong
- Department of Chemistry, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Wei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Mandal S, Mantilla HM, Loganathan K, Faber H, Sharma A, Gedda M, Yengel E, Goswami DK, Heeney M, Anthopoulos TD. Ultra-Fast Moisture Sensor for Respiratory Cycle Monitoring and Non-Contact Sensing Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414005. [PMID: 39821214 PMCID: PMC11854870 DOI: 10.1002/adma.202414005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/01/2025] [Indexed: 01/19/2025]
Abstract
As human-machine interface hardware advances, better sensors are required to detect signals from different stimuli. Among numerous technologies, humidity sensors are critical for applications across different sectors, including environmental monitoring, food production, agriculture, and healthcare. Current humidity sensors rely on materials that absorb moisture, which can take some time to equilibrate with the surrounding environment, thus slowing their temporal response and limiting their applications. Here, this challenge is tackled by combining a nanogap electrode (NGE) architecture with chicked egg-derived albumen as the moisture-absorbing component. The sensors offer inexpensive manufacturing, high responsivity, ultra-fast response, and selectivity to humidity within a relative humidity range of 10-70% RH. Specifically, the egg albumen-based sensor showed negligible response to relevant interfering species and remained specific to water moisture with a room-temperature responsivity of 1.15 × 104. The nm-short interelectrode distance (circa 20 nm) of the NGE architecture enables fast temporal response, with rise/fall times of 10/28 ms, respectively, making the devices the fastest humidity sensors reported to date based on a biomaterial. By leveraging these features, non-contact moisture sensing and real-time respiratory cycle monitoring suitable for diagnosing chronic diseases such as sleep apnea, asthma, and pulmonary disease are demonstrated.
Collapse
Affiliation(s)
- Suman Mandal
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Harold Mazo Mantilla
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Kalaivanan Loganathan
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Hendrik Faber
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Abhinav Sharma
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Murali Gedda
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Emre Yengel
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Dipak Kumar Goswami
- Organic Electronics LaboratoryDepartment of PhysicsIndian Institute of Technology KharagpurKharagpur721302India
| | - Martin Heeney
- Physical Science and Engineering DivisionKing Abdullah University of Science and Technology (KAUST)Thuwal23955‐6900Saudi Arabia
| | - Thomas D. Anthopoulos
- Henry Royce Institute and Photon Science InstituteDepartment of Electrical and Electronic EngineeringThe University of ManchesterOxford RoadManchesterM13 9PLUK
| |
Collapse
|
23
|
Shen L, Song J, Zhou Y, Yuan X, Seery S, Fu T, Liu X, Liu Y, Shao Z, Li R, Liu K. Assessing the ecological resilience of Ebola virus in Africa and potential influencing factors based on a synthesized model. PLoS Negl Trop Dis 2025; 19:e0012843. [PMID: 39919104 PMCID: PMC11805440 DOI: 10.1371/journal.pntd.0012843] [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: 02/18/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND The Ebola epidemic has persisted in Africa since it was firstly identified in 1976. However, few studies have focused on spatiotemporally assessing the ecological adaptability of this virus and the influence of multiple factors on outbreaks. This study quantitatively explores the ecological adaptability of Ebola virus and its response to different potential natural and anthropogenic factors from a spatiotemporal perspective. METHODOLOGY Based on historical Ebola cases and relevant environmental factors collected from 2014 to 2022 in Africa, the spatiotemporal distribution of Ebola adaptability is characterized by integrating four distinct ecological models into one synthesized spatiotemporal framework. Maxent and Generalized Additive Models were applied to further reveal the potential responses of the Ebola virus niche to its ever-changing environments. FINDINGS Ebola habitats appear to aggregate across the sub-Saharan region and in north Zambia and Angola, covering approximately 16% of the African continent. Countries presently unaffected by Ebola but at increasing risk include Ethiopia, Tanzania, Côte d'Ivoire, Ghana, Cameroon, and Rwanda. In addition, among the thirteen key influencing factors, temperature seasonality and population density were identified as significantly influencing the ecological adaptability of Ebola. Specifically, those regions were prone to minimal seasonal variations in temperature. Both the potential anthropogenic influence and vegetation coverage demonstrate a rise-to-decline impact on the outbreaks of Ebola virus across Africa. CONCLUSIONS Our findings suggest new ways to effectively respond to potential Ebola outbreaks in Sub-Saharan Africa. We believe that this integrated modeling approach and response analysis provide a framework that can be extended to predict risk of other worldwide diseases from a similar epidemic study perspective.
Collapse
Affiliation(s)
- Li Shen
- School of Remote Sensing and Information Engineering, Wuhan University, Wuhan, China
| | - Jiawei Song
- School of Remote Sensing and Information Engineering, Wuhan University, Wuhan, China
| | - Yibo Zhou
- School of Remote Sensing and Information Engineering, Wuhan University, Wuhan, China
| | - Xiaojie Yuan
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Samuel Seery
- Faculty of Health and Medicine, Division of Health Research, Lancaster University, Lancaster, United Kingdom
| | - Ting Fu
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Xihao Liu
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Yihong Liu
- The Third Regiment, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Zhongjun Shao
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Rui Li
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Kun Liu
- Department of Epidemiology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, The Shaanxi Provincial Key Laboratory of Environmental Health Hazard Assessment and Protection, The Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
24
|
Beavis BB, Liu J, Zumbrun EE, Bryan AV, Babka AM, Twenhafel NA, Alves DA, Pitt ML, Nalca A, Zeng X. Sudan Virus Persistence in Immune-Privileged Organs of Nonhuman Primate Survivors. Emerg Infect Dis 2025; 31:275-280. [PMID: 39983711 PMCID: PMC11845127 DOI: 10.3201/eid3102.240983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025] Open
Abstract
After the 2022-2023 Sudan virus (SUDV) disease outbreak in Uganda, we studied SUDV persistence in nonhuman primates that had survived acute infection without therapeutic intervention. We identified SUDV persistence in the vitreous chamber and immediately adjacent tissue in the eyes as well as in the seminiferous tubules in the testes but not in common target organs typically infected during the acute phase of disease. Specifically, SUDV persists primarily in macrophages in the eyes and Sertoli cells in the testes. Ocular and testicular SUDV persistence in nonhuman primates is accompanied by tissue damage, including inflammatory cell invasion. Our study suggests that long-term follow-up efforts are needed to reduce possible recrudescent disease and reignition of outbreaks caused by virus persistence in human survivors of SUDV infection.
Collapse
|
25
|
Liu R, Li J, Salena BJ, Li Y. Aptamer and DNAzyme Based Colorimetric Biosensors for Pathogen Detection. Angew Chem Int Ed Engl 2025; 64:e202418725. [PMID: 39551709 PMCID: PMC11753613 DOI: 10.1002/anie.202418725] [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: 09/28/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
The detection of pathogens is critical for preventing and controlling health hazards across clinical, environmental, and food safety sectors. Functional nucleic acids (FNAs), such as aptamers and DNAzymes, have emerged as versatile molecular tools for pathogen detection due to their high specificity and affinity. This review focuses on the in vitro selection of FNAs for pathogens, with emphasis on the selection of aptamers for specific biomarkers and intact pathogens, including bacteria and viruses. Additionally, the selection of DNAzymes for bacterial detection is discussed. The integration of these FNAs into colorimetric biosensors has enabled the development of simple, cost-effective diagnostic platforms. Both non-catalytic and catalytic colorimetric biosensors are explored, including those based on gold nanoparticles, polydiacetylenes, protein enzymes, G-quadruplexes, and nanozymes. These biosensors offer visible detection through color changes, making them ideal for point-of-care diagnostics. The review concludes by highlighting current challenges and future perspectives for advancing FNA-based colorimetric biosensing technologies for pathogen detection.
Collapse
Affiliation(s)
- Rudi Liu
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Bruno J. Salena
- Department of MedicineMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| |
Collapse
|
26
|
Lee S, Lee J, Min KD. Association Between Deforestation and the Incidence of Snakebites in South Korea. Animals (Basel) 2025; 15:198. [PMID: 39858198 PMCID: PMC11758619 DOI: 10.3390/ani15020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/28/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Snakebites are a significant global health concern; despite various known risk factors, the role of environmental changes, such as deforestation, remains underexplored. Therefore, we investigated the association between deforestation and snakebite incidence in South Korea. Aggregated data were obtained from the National Health Insurance Service (NHIS) and the National Emergency Department Information System (NEDIS) from 2014 to 2021. The main explanatory variable was the level of deforestation by district and year. Additional covariates included altitude, average temperature, population size, proportion of urban land cover, proportion of agricultural land cover, and level of ecological preservation. Twelve statistical models were employed to assess the association between deforestation and snakebite incidence. The odds ratios and relative risks ranged between 1.217 and 1.452 and between 1.078 and 1.175, indicating a significant positive association between these two factors. These findings suggest that deforestation notably increases snakebite risk; collectively, our findings can help develop targeted preventive measures and healthcare strategies to reduce snakebite risk globally.
Collapse
Affiliation(s)
- Seoheui Lee
- Department of Big Data, Chungbuk National University, Cheongju 28644, Chungbuk, Republic of Korea
| | - Junyeong Lee
- Department of Management Information Systems, Chungbuk National University, Cheongju 28644, Chungbuk, Republic of Korea
| | - Kyung-Duk Min
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
27
|
Messingham KN, Richards PT, Fleck A, Patel RA, Djurkovic M, Elliff J, Connell S, Crowe TP, Munoz Gonzalez J, Gourronc F, Dillard JA, Davey RA, Klingelhutz A, Shtanko O, Maury W. Multiple cell types support productive infection and dynamic translocation of infectious Ebola virus to the surface of human skin. SCIENCE ADVANCES 2025; 11:eadr6140. [PMID: 39742475 DOI: 10.1126/sciadv.adr6140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
Ebola virus (EBOV) causes severe human disease. During late infection, EBOV virions are on the skin's surface; however, the permissive skin cell types and the route of virus translocation to the epidermal surface are unknown. We describe a human skin explant model and demonstrate that EBOV infection of human skin via basal media increases in a time-dependent and dose-dependent manner. In the dermis, cells of myeloid, endothelial, and fibroblast origin were EBOV antigen-positive whereas keratinocytes harbored virus in the epidermis. Infectious virus was detected on the apical epidermal surface within 3 days, indicating that virus propagates and traffics through the explants. Purified human fibroblasts and keratinocytes supported EBOV infection ex vivo and both cell types required the phosphatidylserine receptor, AXL, and the endosomal protein, NPC1, for virus entry. This platform identified susceptible cell types and demonstrated dynamic trafficking of EBOV virions. These findings may explain person-to-person transmission via skin contact.
Collapse
Affiliation(s)
- Kelly N Messingham
- Department of Dermatology, University of Iowa, Iowa City, IA 52242, USA
- Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Paige T Richards
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Anthony Fleck
- Department of Dermatology, University of Iowa, Iowa City, IA 52242, USA
| | - Radhika A Patel
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Marija Djurkovic
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jonah Elliff
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Samuel Connell
- Department of Dermatology, University of Iowa, Iowa City, IA 52242, USA
| | - Tyler P Crowe
- Department of Dermatology, University of Iowa, Iowa City, IA 52242, USA
| | - Juan Munoz Gonzalez
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Francoise Gourronc
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Jacob A Dillard
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Aloysius Klingelhutz
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Wendy Maury
- Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
28
|
Marx de Andrade R, Paulos A, Valadas E, Laerte Pinto Junior V. [Disease Caused by Filoviruses: An Update]. ACTA MEDICA PORT 2025; 38:42-48. [PMID: 39566016 DOI: 10.20344/amp.21911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024]
Abstract
The Marburg and Ebola viruses belong to the Filoviridae family and are known to cause emerging zoonotic diseases. These viruses have a high case fatality rate and are easily transmissible from person to person, which makes them capable of triggering outbreaks, including in non-endemic regions, and are also considered agents of bioterrorism. Fruit bats are the natural reservoirs of these filoviruses. Transmission to humans occurs through direct contact with bodily fluids or tissues from infected animals or humans. The most severe form of filovirus disease manifests as mucocutaneous hemorrhage, often accompanied by multiorgan failure, which is the main cause of death. Traditionally, these diseases are classified in the group of viral hemorrhagic fevers, although this term is being abandoned, as there are not always hemorrhagic manifestations or fever in the patient's clinical history. Currently, no specific antiviral treatment for filovirus disease exists, and the therapeutic approach consists of supportive measures. However, for the Zaire Ebola virus (EBOV), monoclonal antibodies have already been licensed for treatment and post-exposure prophylaxis, in addition to three vaccines available. Due to the public health importance and the possibility of cases outside Africa, this review aims to improve clinical knowledge and the approach to suspected cases of FD. Improved surveillance and preparedness for potential global outbreaks are essential measures to effectively respond to these public health threats and to ensure that healthcare professionals are well-informed and prepared to deal with these diseases.
Collapse
Affiliation(s)
| | | | - Emília Valadas
- Clínica Universitária de Doenças Infecciosas. Faculdade de Medicina. Universidade de Lisboa. Lisboa. Portugal
| | - Vitor Laerte Pinto Junior
- Faculdade de Medicina. Universidade de Lisboa. Lisboa; Unidade Local de Saúde da Arrábida. Setúbal. Portugal
| |
Collapse
|
29
|
Fang J, Zhou ZJ, Yuan S, Qiu Y, Ge XY. Lineage classification and selective site identification of Orthoebolavirus zairense. Microbes Infect 2025; 27:105304. [PMID: 38278475 DOI: 10.1016/j.micinf.2024.105304] [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: 11/23/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 01/28/2024]
Abstract
As the high pathogenic species of Filoviridae virus family, Orthoebolavirus zairense (EBOV) shows frequent outbreaks in human in recently years since its first emerging in 1976 in Democratic Republic of the Congo (COD), bringing ongoing risks and burden on public health safety. Here, the phylogenetic relationship among major outbreaks was analyzed. The results showed that EBOV isolates could be divided into four lineages according to spatial and temporal epidemics. Then, the positive selection sites (PSSs) were detected on all proteins of the EBOV, exhibiting lineage characteristic. Particularly, sites in GP and VP24 were identified to be significantly under positive selection, and partial of which were maintained in the latest isolates in 2021. GP and L were found to have high variability between lineages. Substitutions including F443L and F443S in GP, as well as F1610L and I1951V in L could be characteristic of the two large outbreaks in COD (2018) and West Africa (2014), respectively. Further, substitutions of significant PSSs in VP24 and L proteins were visualized for analysis of structural changes, which may affect EBOV pathogenesis. In summary, our results gains insights in genetic characteristic and adaptive evolution of EBOV, which could facilitate gene functional research against EBOV.
Collapse
Affiliation(s)
- Jie Fang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, 27 Tianma Rd., Changsha, Hunan, 410012, China
| | - Zhi-Jian Zhou
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, 27 Tianma Rd., Changsha, Hunan, 410012, China
| | - Shuofeng Yuan
- Department of Microbiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, 27 Tianma Rd., Changsha, Hunan, 410012, China
| | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, 27 Tianma Rd., Changsha, Hunan, 410012, China.
| |
Collapse
|
30
|
Rodríguez E, Muñoz-Fontela C, Escudero-Pérez B. Filovirus Infection in Humanized Mouse Models. Methods Mol Biol 2025; 2877:213-226. [PMID: 39585624 DOI: 10.1007/978-1-0716-4256-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Since the discovery of filoviruses in 1967, there have been more than 40 outbreaks with high case-fatality rates causing more than 30,000 deaths in humans. Filovirus disease (FVD) involves the dysregulation of many host physiological processes. While many advances in the field have taken place since the first outbreaks, the dearth of small animal models that translate the features observed during FVD in humans has limited our understanding of the pathology. Here, we describe the generation and use of human immune system (HIS) mice as a preclinical model to investigate FVD in a human-like immune environment.
Collapse
Affiliation(s)
- Estefanía Rodríguez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany
| | - Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany.
| |
Collapse
|
31
|
Richards PT, Briseño JAA, Brunton BA, Maury W. In Vivo Investigation of Filovirus Glycoprotein-Mediated Infection in a BSL2 Setting. Methods Mol Biol 2025; 2877:183-198. [PMID: 39585622 PMCID: PMC11727417 DOI: 10.1007/978-1-0716-4256-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Highly pathogenic viruses in the Filoviridae family are causative agents of filovirus disease (FVD). Ebola virus (EBOV) is one such member and, of all filoviruses, represents the largest threat to global public health. The study of FVD has been hampered by the lack of tools to study filovirus infection outside maximum containment laboratories. Recombinant vesicular stomatitis virus (VSV) lacking its native glycoprotein and expressing a filovirus glycoprotein (VSV-filo GP) has improved our understanding of GP-mediated host-cell interactions as well as adaptive and humoral immune responses in in vitro and in vivo studies. Furthermore, mouse models suitable for these studies are readily available. Here, we describe multiple injection routes for investigating filovirus GP-mediated infection and pathogenesis using VSV-filo GP and interferon α/β receptor-deficient (Ifnar-/-) mice as models. These tools can be safely used outside maximum containment laboratories, are cost effective, and easy to manipulate.
Collapse
Affiliation(s)
- Paige T Richards
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | | | - Bethany A Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
32
|
Schrell L, Fuchs HL, Dickmanns A, Scheibner D, Olejnik J, Hume AJ, Reineking W, Störk T, Müller M, Graaf-Rau A, Diederich S, Finke S, Baumgärtner W, Mühlberger E, Balkema-Buschmann A, Dobbelstein M. Inhibitors of dihydroorotate dehydrogenase synergize with the broad antiviral activity of 4'-fluorouridine. Antiviral Res 2025; 233:106046. [PMID: 39638153 DOI: 10.1016/j.antiviral.2024.106046] [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: 10/05/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
RNA viruses present a constant threat to human health, often with limited options for vaccination or therapy. Notable examples include influenza viruses and coronaviruses, which have pandemic potential. Filo- and henipaviruses cause more limited outbreaks, but with high case fatality rates. All RNA viruses rely on the activity of a virus-encoded RNA-dependent RNA polymerase (RdRp). An antiviral nucleoside analogue, 4'-Fluorouridine (4'-FlU), targets RdRp and diminishes the replication of several RNA viruses, including influenza A virus and SARS-CoV-2, through incorporation into nascent viral RNA and delayed chain termination. However, the effective concentration of 4'-FlU varied among different viruses, raising the need to fortify its efficacy. Here we show that inhibitors of dihydroorotate dehydrogenase (DHODH), an enzyme essential for pyrimidine biosynthesis, can synergistically enhance the antiviral effect of 4'-FlU against influenza A viruses, SARS-CoV-2, henipaviruses, and Ebola virus. Even 4'-FlU-resistant mutant influenza A virus was re-sensitized towards 4'-FlU by DHODH inhibition. The addition of uridine rescued influenza A virus replication, strongly suggesting uridine depletion as a mechanism of this synergy. 4'-FlU was also highly effective against SARS-CoV-2 in a hamster model of COVID. We propose that the impairment of endogenous uridine synthesis by DHODH inhibition enhances the incorporation of 4'-FlU into viral RNAs. This strategy may be broadly applicable to enhance the efficacy of pyrimidine nucleoside analogues for antiviral therapy.
Collapse
Affiliation(s)
- Leon Schrell
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Hannah L Fuchs
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Antje Dickmanns
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - David Scheibner
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Adam J Hume
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Martin Müller
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Annika Graaf-Rau
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Sandra Diederich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
33
|
Goldstein N, McLean C, Gaddah A, Doua J, Keshinro B, Bus-Jacobs L, Hendriks J, Luhn K, Robinson C, Douoguih M. Lot-to-lot consistency, immunogenicity, and safety of the Ad26.ZEBOV, MVA-BN-Filo Ebola virus vaccine regimen: A phase 3, randomized, double-blind, placebo-controlled trial. Hum Vaccin Immunother 2024; 20:2327747. [PMID: 38523332 PMCID: PMC10965106 DOI: 10.1080/21645515.2024.2327747] [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: 12/19/2023] [Accepted: 03/05/2024] [Indexed: 03/26/2024] Open
Abstract
This phase-3, double-blind, placebo-controlled study (NCT04228783) evaluated lot-to-lot consistency of the Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine regimen. Participants were randomized (6:6:6:1) to receive the two-dose regimen from three consecutively manufactured lots of Ad26.ZEBOV on Day 1 paired with three consecutively manufactured lots of MVA-BN-Filo on Day 57 (Groups 1-3) or two doses of placebo (Group 4). An additional cohort also received an Ad26.ZEBOV booster or placebo 4 months post-dose 2. Equivalence of the immunogenicity at 21 days post-dose 2 between any two groups was demonstrated if the 95% confidence interval (CI) of the Ebola virus glycoprotein (EBOV GP)-binding antibody geometric mean concentration (GMC) ratio was entirely within the prespecified margin of 0.5-2.0. Lot-to-lot consistency (i.e., consecutive lots can be consistently manufactured) was accomplished if equivalence was shown for all three pairwise comparisons. Results showed that the primary objective in the per-protocol immunogenicity subset (n = 549) was established for each pairwise comparison (Group 1 vs 2: GMC ratio = 0.9 [95% CI: 0.8, 1.1], Group 1 vs 3: 0.9 [0.8, 1.1], Group 2 vs 3: 1.0 [0.9, 1.2]). Equivalence of the three groups for the Ad26.ZEBOV component only was also demonstrated at 56 days post-dose 1. EBOV GP-binding antibody responses (post-vaccination concentrations >2.5-fold from baseline) were observed in 419/421 (99.5%) vaccine recipients at 21 days post-dose 2 and 445/460 (96.7%) at 56 days post-dose 1. In the booster cohort (n = 39), GMCs increased 9.0- and 11.8-fold at 7 and 21 days post-booster, respectively, versus pre-booster. Ad26.ZEBOV, MVA-BN-Filo was well tolerated, and no safety issues were identified.
Collapse
Affiliation(s)
- Neil Goldstein
- Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Chelsea McLean
- Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | | | | | | | | | - Jenny Hendriks
- Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Kerstin Luhn
- Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | | | | |
Collapse
|
34
|
Zumbrun EE, Garvey CB, Wells JB, Lynn GC, Van Tongeren SA, Steffens JT, Wetzel KS, Wetzel DL, Esham HL, Garza NL, Lee ED, Scruggs JL, Rossi FD, Brown ES, Weidner JM, Gomba LM, O’Brien KA, Jay AN, Zeng X, Akers KS, Kallgren PA, Englund E, Meinig JM, Kugelman JR, Moore JL, Bloomfield HA, Norris SL, Bryan T, Scheuerell CH, Walters J, Mollova N, Blair C, Babusis D, Cihlar T, Porter DP, Singh B, Hedskog C, Bavari S, Warren TK, Bannister R. A Randomized, Blinded, Vehicle-Controlled Dose-Ranging Study to Evaluate and Characterize Remdesivir Efficacy Against Ebola Virus in Rhesus Macaques. Viruses 2024; 16:1934. [PMID: 39772240 PMCID: PMC11680158 DOI: 10.3390/v16121934] [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: 11/26/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Ebola virus (EBOV) causes severe disease in humans, with mortality as high as 90%. The small-molecule antiviral drug remdesivir (RDV) has demonstrated a survival benefit in EBOV-exposed rhesus macaques. Here, we characterize the efficacy of multiple intravenous RDV dosing regimens on survival of rhesus macaques 42 days after intramuscular EBOV exposure. Thirty rhesus macaques underwent surgical implantation of telemetry devices for the fine-scale monitoring of body temperature and activity, as well as central venous catheters, to enable treatment administration and blood collection. Treatment, consisting of a loading dose of RDV followed by once-daily maintenance doses for 11 days, was initiated 4 days after virus exposure when all animals were exhibiting disease signs consistent with incipient EBOV disease as well as quantifiable levels of EBOV RNA in plasma. In the RDV treatment groups receiving loading/maintenance doses of 5/2.5 mg/kg, 10/5 mg/kg, and 20/10 mg/kg, a total of 6 of 8 (75%), 7 of 8 (87.5%), and 5 of 7 (71.4%) animals survived, respectively. In the vehicle control group, one of seven animals (14.3%) survived. The improved survival rate compared to the control group was statistically significant only for the 10/5 mg/kg RDV treatment group. This treatment regimen also resulted in a significantly lower systemic viral load compared to the vehicle control after a single RDV treatment. All three RDV regimens produced a significantly lower systemic viral load after two treatments. For most animals, RDV treatment, regardless of dose, resulted in the amelioration of many of the clinical-pathological changes associated with EBOV disease in this model.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Carly B. Garvey
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jay B. Wells
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Ginger C. Lynn
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Sean A. Van Tongeren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jesse T. Steffens
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kelly S. Wetzel
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Darrell L. Wetzel
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Heather L. Esham
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Nicole L. Garza
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Eric D. Lee
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Jennifer L. Scruggs
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Franco D. Rossi
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Elizabeth S. Brown
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jessica M. Weidner
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Laura M. Gomba
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kristan A. O’Brien
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Alexandra N. Jay
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Kristen S. Akers
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Paul A. Kallgren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Ethan Englund
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - J. Matthew Meinig
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Jeffrey R. Kugelman
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Joshua L. Moore
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Holly A. Bloomfield
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Sarah L. Norris
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Tameka Bryan
- PharPoint Research, Inc., Wilmington, NC 28401, USA;
| | | | - Jesse Walters
- Labcorp Early Development Laboratories, Madison, WI 53704, USA; (C.H.S.); (J.W.)
| | - Nevena Mollova
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Christiana Blair
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Darius Babusis
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Tomas Cihlar
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Danielle P. Porter
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Bali Singh
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Charlotte Hedskog
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Travis K. Warren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Roy Bannister
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| |
Collapse
|
35
|
Zalwango MG, Paige S, Migisha R, Nakafeero Simbwa B, Nsubuga EJ, Asio A, Kabami Z, Zalwango JF, Kawungezi PC, Wanyana MW, King P, Naiga HN, Agaba B, Zavuga R, Earle-Richardson G, Kwesiga B, Bulage L, Kadobera D, Ario AR, Harris JR. Stigma among ebola disease survivors in Mubende and Kassanda districts, Central Uganda, 2022. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003272. [PMID: 39680556 DOI: 10.1371/journal.pgph.0003272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/17/2024] [Indexed: 12/18/2024]
Abstract
Ebola disease survivors often experience stigma in multiple forms, including felt (perceived) stigma, enacted (action-based) stigma, and institutional stigma. On September 20, 2022, Uganda declared a Sudan Virus Disease (species orthoebolavirus sudanense) outbreak after a patient with confirmed Sudan virus (SUDV) infection was identified in Mubende District. The outbreak led to 142 confirmed and 22 probable cases over the next two months. We examined the types of stigma experienced by survivors and their household members and its effect on their well-being. We conducted a qualitative study during January 2023 in Mubende and Kassanda Districts. We conducted in-depth and key informant interviews with ten SUDV disease survivors, ten household members of SUDV disease survivors, and ten key informants (district officials and health workers in the affected communities). Interviews were recorded, translated, transcribed, and analyzed thematically. Survivors reported experiencing isolation and rejection by community members and loss of work. They reported being denied purchases at shops or having their money collected in a basket and disinfected (enacted stigma), which led to self-isolation (felt stigma). Educational institutions denied admission to some students from affected homes, while parents of children in some affected families stopped sending children to school due to verbal abuse from students and teachers (structural stigma). Prolonged SUDV disease symptoms and additional attention to survivors from responders (including home visits by health workers, public distribution of support items, and conspicuous transport from home to the survivor's clinic) were perceived as aggravating both felt and enacted stigma. Even after the outbreak had been declared over, survivors felt that they were still considered a threat to the community. Survivors experienced mainly enacted stigma which was aggravated by the outbreak response and control activities such as additional attention to survivors from responders. Strengthening community engagement to counteract stigma, rethinking response activities that aggravate stigma, integrated response interventions by partners, private distribution of support items, and increasing awareness and sensitization could reduce stigma among the Ebola disease survivors in future responses.
Collapse
Affiliation(s)
- Marie Gorreti Zalwango
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Sarah Paige
- United States Agency for International Development, Kampala, Uganda
| | - Richard Migisha
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Brenda Nakafeero Simbwa
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Edirisa Junior Nsubuga
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Alice Asio
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Zainah Kabami
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Jane Frances Zalwango
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Peter Chris Kawungezi
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Mercy Wendy Wanyana
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Patrick King
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Hellen Nelly Naiga
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Brian Agaba
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Robert Zavuga
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Giulia Earle-Richardson
- National Center for Emerging Zoonotic and Infectious Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Benon Kwesiga
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Lilian Bulage
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Daniel Kadobera
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Alex Riolexus Ario
- Uganda Public Health Fellowship Program-Uganda National Institute of Public Health, Kampala, Uganda
| | - Julie R Harris
- Division of Global Health Protection, US Centers for Disease Control and Prevention, Kampala, Uganda
| |
Collapse
|
36
|
Xu JQ, Zhang WY, Fu JJ, Fang XZ, Gao CG, Li C, Yao L, Li QL, Yang XB, Ren LH, Shu HQ, Peng K, Wu Y, Zhang DY, Qiu Y, Zhou X, Yao YM, Shang Y. Viral sepsis: diagnosis, clinical features, pathogenesis, and clinical considerations. Mil Med Res 2024; 11:78. [PMID: 39676169 PMCID: PMC11648306 DOI: 10.1186/s40779-024-00581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Sepsis, characterized as life-threatening organ dysfunction resulting from dysregulated host responses to infection, remains a significant challenge in clinical practice. Despite advancements in understanding host-bacterial interactions, molecular responses, and therapeutic approaches, the mortality rate associated with sepsis has consistently ranged between 10 and 16%. This elevated mortality highlights critical gaps in our comprehension of sepsis etiology. Traditionally linked to bacterial and fungal pathogens, recent outbreaks of acute viral infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), among other regional epidemics, have underscored the role of viral pathogenesis in sepsis, particularly when critically ill patients exhibit classic symptoms indicative of sepsis. However, many cases of viral-induced sepsis are frequently underdiagnosed because standard evaluations typically exclude viral panels. Moreover, these viruses not only activate conventional pattern recognition receptors (PRRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) but also initiate primary antiviral pathways such as cyclic guanosine monophosphate adenosine monophosphate (GMP-AMP) synthase (cGAS)-stimulator of interferon genes (STING) signaling and interferon response mechanisms. Such activations lead to cellular stress, metabolic disturbances, and extensive cell damage that exacerbate tissue injury while leading to a spectrum of clinical manifestations. This complexity poses substantial challenges for the clinical management of affected cases. In this review, we elucidate the definition and diagnosis criteria for viral sepsis while synthesizing current knowledge regarding its etiology, epidemiology, and pathophysiology, molecular mechanisms involved therein as well as their impact on immune-mediated organ damage. Additionally, we discuss clinical considerations related to both existing therapies and advanced treatment interventions, aiming to enhance the comprehensive understanding surrounding viral sepsis.
Collapse
Affiliation(s)
- Ji-Qian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wan-Ying Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Ji Fu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Gang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chang Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lu Yao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi-Lan Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Bo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Le-Hao Ren
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua-Qing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Ying Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, 430072, China
| | - Ding-Yu Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yang Qiu
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Xi Zhou
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and the Fourth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
37
|
Fan P, Sun B, Liu Z, Fang T, Ren Y, Zhao X, Song Z, Yang Y, Li J, Yu C, Chen W. A pan-orthoebolavirus neutralizing antibody encoded by mRNA effectively prevents virus infection. Emerg Microbes Infect 2024; 13:2432366. [PMID: 39560055 PMCID: PMC11590195 DOI: 10.1080/22221751.2024.2432366] [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: 09/09/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
Orthoebolavirus is a genus of hazardous pathogens that has caused over 30 outbreaks. However, currently approved therapies are limited in scope, as they are only effective against the Ebola virus and lack cross-protection against other orthoebolaviruses. Here, we demonstrate that a previously isolated human-derived antibody, 2G1, can recognize the glycoprotein (GP) of every orthoebolavirus species. The cryo-electron microscopy structure of 2G1 Fab in complex with the GPΔMucin trimer reveals that 2G1 binds a quaternary pocket formed by three subunits from two GP protomers. 2G1 recognizes highly conserved epitopes among filoviruses and achieves neutralization by blocking GP proteolysis. We designed an efficient mRNA module capable of producing test antibodies at expression levels exceeding 1500 ng/mL in vitro. The lipid nanoparticle (LNP)-encapsulated mRNA-2G1 exhibited potent neutralizing activities against the HIV-pseudotyped Ebola and Sudan viruses that were 19.8 and 12.5 times that of IgG format, respectively. In mice, the antibodies encoded by the mRNA-2G1-LNP peaked within 24 h, effectively blocking the invasion of pseudoviruses with no apparent liver toxicity. This study suggests that the 2G1 antibody and its mRNA formulation represent promising candidate interventions for orthoebolavirus disease, and it provides an efficient mRNA framework applicable to antibody-based therapies.
Collapse
Affiliation(s)
- Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yi Ren
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zhenwei Song
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Wei Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| |
Collapse
|
38
|
Li W, Yang W, Liu X, Zhou W, Wang S, Wang Z, Zhao Y, Feng N, Wang T, Wu M, Ge L, Xia X, Yan F. Fully human monoclonal antibodies against Ebola virus possess complete protection in a hamster model. Emerg Microbes Infect 2024; 13:2392651. [PMID: 39155772 PMCID: PMC11348817 DOI: 10.1080/22221751.2024.2392651] [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: 05/05/2024] [Revised: 07/14/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Ebola disease is a lethal viral hemorrhagic fever caused by ebolaviruses within the Filoviridae family with mortality rates of up to 90%. Monoclonal antibody (mAb) based therapies have shown great potential for the treatment of EVD. However, the potential emerging ebolavirus isolates and the negative effect of decoy protein on the therapeutic efficacy of antibodies highlight the necessity of developing novel antibodies to counter the threat of Ebola. Here, 11 fully human mAbs were isolated from transgenic mice immunized with GP protein and recombinant vesicular stomatitis virus-bearing GP (rVSV-EBOV GP). These mAbs were divided into five groups according to their germline genes and exhibited differential binding activities and neutralization capabilities. In particular, mAbs 8G6, 2A4, and 5H4 were cross-reactive and bound at least three ebolavirus glycoproteins. mAb 4C1 not only exhibited neutralizing activity but no cross-reaction with sGP. mAb 7D8 exhibited the strongest neutralizing capacity. Further analysis on the critical residues for the bindings of 4C1 and 8G6 to GPs was conducted using antibodies complementarity-determining regions (CDRs) alanine scanning. It has been shown that light chain CDR3 played a crucial role in binding and neutralization and that any mutation in CDRs could not improve the binding of 4C1 to sGP. Importantly, mAbs 7D8, 8G6, and 4C1 provided complete protections against EBOV infection in a hamster lethal challenge model when administered 12 h post-infection. These results support mAbs 7D8, 8G6, and 4C1 as potent antibody candidates for further investigations and pave the way for further developments of therapies and vaccines.
Collapse
Affiliation(s)
- Wujian Li
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Wanying Yang
- Department of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xueqin Liu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Wujie Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Zhenshan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Meng Wu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| |
Collapse
|
39
|
Liu T, Cao Y, Weng J, Gao S, Jin Z, Zhang Y, Yang Y, Zhang H, Xia C, Yin X, Luo Y, He Q, Jiang H, Wang L, Zhang Z. Hepatitis E virus infects human testicular tissue and Sertoli cells. Emerg Microbes Infect 2024; 13:2332657. [PMID: 38517709 PMCID: PMC11057402 DOI: 10.1080/22221751.2024.2332657] [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: 02/15/2024] [Accepted: 03/14/2024] [Indexed: 03/24/2024]
Abstract
Globally, hepatitis E virus (HEV) infections are prevalent. The finding of high viral loads and persistent viral shedding in ejaculate suggests that HEV replicates within the human male genital tract, but its target organ is unknown and appropriate models are lacking. We aimed to determine the HEV tropism in the human testis and its potential influence on male reproductive health. We conducted an ex vivo culture of human testis explants and in vitro culture of primary human Sertoli cells. Clinically derived HEV genotype 1 (HEV1) and HEV3 virions, as well as rat-derived HEV-C1, were used for inoculation. Transcriptomic analysis was performed on testis tissues collected from tacrolimus-treated rabbits with chronic HEV3 infection. Our findings reveal that HEV3, but not HEV1 or HEV-C1, can replicate in human testis explants and primary human Sertoli cells. Tacrolimus treatment significantly enhanced the replication efficiency of HEV3 in testis explants and enabled successful HEV1 infection in Sertoli cells. HEV3 infection disrupted the secretion of several soluble factors and altered the cytokine microenvironment within primary human Sertoli cells. Finally, intratesticular transcriptomic analysis of immunocompromised rabbits with chronic HEV infection indicated downregulation of genes associated with spermatogenesis. HEV can infect the human testicular tissues and Sertoli cells, with increased replication efficiency when exposed to tacrolimus treatment. These findings shed light on how HEV may persist in the ejaculate of patients with chronic hepatitis E and provide valuable ex vivo tools for studying countermeasures.
Collapse
Affiliation(s)
- Tianxu Liu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Yalei Cao
- Department of Urology, Peking University Third Hospital, Beijing, People’s Republic of China
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Jiaming Weng
- Department of Urology, Peking University Third Hospital, Beijing, People’s Republic of China
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Songzhan Gao
- Department of Andrology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Zirun Jin
- Department of Urology, Peking University Third Hospital, Beijing, People’s Republic of China
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Yun Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Yuzhuo Yang
- Department of Urology, Peking University First Hospital, Beijing, People’s Republic of China
| | - He Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Changyou Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Yong Luo
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Qiyu He
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Hui Jiang
- Department of Urology, Peking University First Hospital, Beijing, People’s Republic of China
| | - Lin Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing, People’s Republic of China
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, People’s Republic of China
| |
Collapse
|
40
|
Nabi W, Merle H, Abroug N, Jean-Charles A, Ksiaa I, Jelliti B, Khairallah M. [Viral uveitis in the tropics]. J Fr Ophtalmol 2024; 47:104342. [PMID: 39509945 DOI: 10.1016/j.jfo.2024.104342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 11/15/2024]
Abstract
Tropical regions are characterized by a hot and humid climate that promotes the emergence or resurgence of endemic or epidemic levels of microorganisms such as viruses. Emerging viral infections are mostly inapparent, mild, or non-specific (flu-like syndrome), but they may include serious, potentially disabling or life-threatening complications. A wide array of ocular manifestations has been recently described in association with specific emerging viral infections, including arboviruses, Human T-cell Lymphotropic Virus Type 1 (HTLV-1), and Ebola. Emerging viral disease should be considered in the differential diagnosis of ocular inflammatory conditions in patients living in or returning from endemic regions. Diagnosis of emerging viral infection is confirmed with either real time polymerase chain reaction or serology. Management consists of supportive care, as there are currently no specific antiviral drug options. Corticosteroids are often used for the treatment of associated ocular inflammation. Most patients have a good visual prognosis, but there may be permanent visual impairment in some due to ocular structural complications. Prophylactic measures are essential to prevent human infection and disease.
Collapse
Affiliation(s)
- W Nabi
- Service d'ophtalmologie, faculté de médecine, CHU Fattouma Bourguiba, université de Monastir, Monastir, Tunisie
| | - H Merle
- EA4537, Inserm CIC 1424, service d'ophtalmologie, centre hospitalier universitaire de Martinique, université des Antilles, Martinique
| | - N Abroug
- Service d'ophtalmologie, faculté de médecine, CHU Fattouma Bourguiba, université de Monastir, Monastir, Tunisie
| | - A Jean-Charles
- EA4537, Inserm CIC 1424, service d'ophtalmologie, centre hospitalier universitaire de Martinique, université des Antilles, Martinique
| | - I Ksiaa
- Service d'ophtalmologie, faculté de médecine, CHU Fattouma Bourguiba, université de Monastir, Monastir, Tunisie
| | - B Jelliti
- Service d'ophtalmologie, faculté de médecine, CHU Fattouma Bourguiba, université de Monastir, Monastir, Tunisie
| | - M Khairallah
- Service d'ophtalmologie, faculté de médecine, CHU Fattouma Bourguiba, université de Monastir, Monastir, Tunisie.
| |
Collapse
|
41
|
Bosa HK, Kamara N, Aragaw M, Wayengera M, Katoto PDMC, Ihekweazu C, Fallah MP, Douno M, Agyarko RK, Mbala P, Traoré MS, Talisuna A, Bangura J, Mwebesa HG, Bousso A, Joshua O, Squire JS, Nyenswah T, Nelson TV, Maeda J, Olu OO, Woldemariam YT, Djoudalbaye B, Ngongo AN, Raji T, Kasolo FC, Fall IS, Ouma Ogwell A, Aceng JR, Kaseya J. How to prepare for the next inevitable Ebola outbreak: lessons from West Africa. Nat Med 2024; 30:3413-3416. [PMID: 39420216 DOI: 10.1038/s41591-024-03295-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Affiliation(s)
| | - Neema Kamara
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | - Merawi Aragaw
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia.
| | | | | | | | - Mosoka P Fallah
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | - Moussa Douno
- Université Gamal Abdel de Conakry, Conakry, Guinea
| | | | - Placide Mbala
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | | | - Ambrose Talisuna
- World Health Organization Regional Office for Africa, Brazzaville, Republic of Congo
| | - James Bangura
- Health and Development in Action, Freetown, Sierra Leone
| | | | | | | | | | - Tolbert Nyenswah
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Justin Maeda
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | - Olushayo Oluseun Olu
- World Health Organization Regional Office for Africa, Brazzaville, Republic of Congo
| | | | | | | | - Tajudeen Raji
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | | | | | - Ahmed Ouma Ogwell
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | | | - Jean Kaseya
- Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| |
Collapse
|
42
|
Fan L, Wang Y, Huang H, Wang Z, Liang C, Yang X, Ye P, Lin J, Shi W, Zhou Y, Yan H, Long Z, Wang Z, Liu L, Qian J. RNA binding motif 4 inhibits the replication of ebolavirus by directly targeting 3'-leader region of genomic RNA. Emerg Microbes Infect 2024; 13:2300762. [PMID: 38164794 PMCID: PMC10773643 DOI: 10.1080/22221751.2023.2300762] [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: 09/29/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Ebola virus (EBOV) belongs to Filoviridae family possessing single-stranded negative-sense RNA genome, which is a serious threat to human health. Nowadays, no therapeutics have been proven to be successful in efficiently decreasing the mortality rate. RNA binding proteins (RBPs) are reported to participate in maintaining cell integrity and regulation of viral replication. However, little is known about whether and how RBPs participate in regulating the life cycle of EBOV. In our study, we found that RNA binding motif protein 4 (RBM4) inhibited the replication of EBOV in HEK293T and Huh-7 cells by suppressing viral mRNA production. Such inhibition resulted from the direct interaction between the RRM1 domain of RBM4 and the "CU" enrichment elements located in the PE1 and TSS of the 3'-leader region within the viral genome. Simultaneously, RBM4 could upregulate the expression of some cytokines involved in the host innate immune responses to synergistically exert its antiviral function. The findings therefore suggest that RBM4 might serve as a novel target of anti-EBOV strategy.
Collapse
Affiliation(s)
- Linjin Fan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Yulong Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Hongxin Huang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Zequn Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Chudan Liang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Xiaofeng Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Pengfei Ye
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Jingyan Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Wendi Shi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Yuandong Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Huijun Yan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Zhenyu Long
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Zhongyi Wang
- Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Linna Liu
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jun Qian
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
| |
Collapse
|
43
|
Bu F, Ye G, Morsheimer K, Mendoza A, Turner-Hubbard H, Herbst M, Spiller B, Wadzinski BE, Eaton B, Anantpadma M, Yang G, Liu B, Davey R, Li F. Discovery of Nanosota-EB1 and -EB2 as Novel Nanobody Inhibitors Against Ebola Virus Infection. PLoS Pathog 2024; 20:e1012817. [PMID: 39715280 PMCID: PMC11723632 DOI: 10.1371/journal.ppat.1012817] [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: 09/27/2024] [Revised: 01/10/2025] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
The Ebola filovirus (EBOV) poses a serious threat to global health and national security. Nanobodies, a type of single-domain antibody, have demonstrated promising therapeutic potential. We identified two anti-EBOV nanobodies, Nanosota-EB1 and Nanosota-EB2, which specifically target the EBOV glycoprotein (GP). Cryo-EM and biochemical data revealed that Nanosota-EB1 binds to the glycan cap of GP1, preventing its protease cleavage, while Nanosota-EB2 binds to critical membrane-fusion elements in GP2, stabilizing it in the pre-fusion state. Nanosota-EB2 is a potent neutralizer of EBOV infection in vitro and offers excellent protection in a mouse model of EBOV challenge, while Nanosota-EB1 provides moderate neutralization and protection. Nanosota-EB1 and Nanosota-EB2 are the first nanobodies shown to inhibit authentic EBOV. Combined with our newly developed structure-guided in vitro evolution approach, they lay the foundation for nanobody-based therapies against EBOV and other viruses within the ebolavirus genus.
Collapse
Affiliation(s)
- Fan Bu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gang Ye
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kimberly Morsheimer
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Alise Mendoza
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Hailey Turner-Hubbard
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Morgan Herbst
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Benjamin Spiller
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Brett Eaton
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Manu Anantpadma
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Ge Yang
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Bin Liu
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Robert Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
44
|
Markandan K, Tiong YW, Sankaran R, Subramanian S, Markandan UD, Chaudhary V, Numan A, Khalid M, Walvekar R. Emergence of infectious diseases and role of advanced nanomaterials in point-of-care diagnostics: a review. Biotechnol Genet Eng Rev 2024; 40:3438-3526. [PMID: 36243900 DOI: 10.1080/02648725.2022.2127070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022]
Abstract
Infectious outbreaks are the foremost global public health concern, challenging the current healthcare system, which claims millions of lives annually. The most crucial way to control an infectious outbreak is by early detection through point-of-care (POC) diagnostics. POC diagnostics are highly advantageous owing to the prompt diagnosis, which is economical, simple and highly efficient with remote access capabilities. In particular, utilization of nanomaterials to architect POC devices has enabled highly integrated and portable (compact) devices with enhanced efficiency. As such, this review will detail the factors influencing the emergence of infectious diseases and methods for fast and accurate detection, thus elucidating the underlying factors of these infections. Furthermore, it comprehensively highlights the importance of different nanomaterials in POCs to detect nucleic acid, whole pathogens, proteins and antibody detection systems. Finally, we summarize findings reported on nanomaterials based on advanced POCs such as lab-on-chip, lab-on-disc-devices, point-of-action and hospital-on-chip. To this end, we discuss the challenges, potential solutions, prospects of integrating internet-of-things, artificial intelligence, 5G communications and data clouding to achieve intelligent POCs.
Collapse
Affiliation(s)
- Kalaimani Markandan
- Temasek Laboratories, Nanyang Technological University, Nanyang Drive, Singapore
- Faculty of Engineering, Technology and Built Environment, UCSI University, Kuala Lumpur, Malaysia
| | - Yong Wei Tiong
- NUS Environmental Research Institute, National University of Singapore, Engineering Drive, Singapore
| | - Revathy Sankaran
- Graduate School, University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Sakthinathan Subramanian
- Department of Materials & Mineral Resources Engineering, National Taipei University of Technology (NTUT), Taipei, Taiwan
| | | | - Vishal Chaudhary
- Research Cell & Department of Physics, Bhagini Nivedita College, University of Delhi, New Delhi, India
| | - Arshid Numan
- Graphene & Advanced 2D Materials Research Group (GAMRG), School of Engineering and Technology, Sunway University, Petaling Jaya, Selangor, Malaysia
- Sunway Materials Smart Science & Engineering (SMS2E) Research Cluster School of Engineering and Technology, Sunway University, Selangor, Malaysia
| | - Mohammad Khalid
- Graphene & Advanced 2D Materials Research Group (GAMRG), School of Engineering and Technology, Sunway University, Petaling Jaya, Selangor, Malaysia
- Sunway Materials Smart Science & Engineering (SMS2E) Research Cluster School of Engineering and Technology, Sunway University, Selangor, Malaysia
| | - Rashmi Walvekar
- Department of Chemical Engineering, School of Energy and Chemical Engineering, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| |
Collapse
|
45
|
Wanninger TG, Saldarriaga OA, Arroyave E, Millian DE, Comer JE, Paessler S, Stevenson HL. Hepatic and pulmonary macrophage activity in a mucosal challenge model of Ebola virus disease. Front Immunol 2024; 15:1439971. [PMID: 39635525 PMCID: PMC11615675 DOI: 10.3389/fimmu.2024.1439971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background The inflammatory macrophage response contributes to severe Ebola virus disease, with liver and lung injury in humans. Objective We sought to further define the activation status of hepatic and pulmonary macrophage populations in Ebola virus disease. Methods We compared liver and lung tissue from terminal Ebola virus (EBOV)-infected and uninfected control cynomolgus macaques challenged via the conjunctival route. Gene and protein expression was quantified using the nCounter and GeoMx Digital Spatial Profiling platforms. Macrophage phenotypes were further quantified by digital pathology analysis. Results Hepatic macrophages in the EBOV-infected group demonstrated a mixed inflammatory/non-inflammatory profile, with upregulation of CD163 protein expression, associated with macrophage activation syndrome. Hepatic macrophages also showed differential expression of gene sets related to monocyte/macrophage differentiation, antigen presentation, and T cell activation, which were associated with decreased MHC-II allele expression. Moreover, hepatic macrophages had enriched expression of genes and proteins targetable with known immunomodulatory therapeutics, including S100A9, IDO1, and CTLA-4. No statistically significant differences in M1/M2 gene expression were observed in hepatic macrophages compared to controls. The significant changes that occurred in both the liver and lung were more pronounced in the liver. Conclusion These data demonstrate that hepatic macrophages in terminal conjunctivally challenged cynomolgus macaques may express a unique inflammatory profile compared to other macaque models and that macrophage-related pharmacologically druggable targets are expressed in both the liver and the lung in Ebola virus disease.
Collapse
Affiliation(s)
- Timothy G. Wanninger
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Omar A. Saldarriaga
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Esteban Arroyave
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Daniel E. Millian
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jason E. Comer
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Heather L. Stevenson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
46
|
Wang F, Hu F, Zhang Y, Li X, Ma Q, Wang X, Peng N. A Novel High-Throughput Sample-in-Result-Out Device for the Rapid Detection of Viral Nucleic Acids. BIOSENSORS 2024; 14:549. [PMID: 39590008 PMCID: PMC11591587 DOI: 10.3390/bios14110549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/26/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) molecular diagnostic technology is one of the most reliable diagnostic tools for infectious diseases due to its short reaction time, high sensitivity, and excellent specificity. However, compared with fluorescent polymerase chain reaction (PCR) technology, CRISPR molecular diagnostic technology lacks high-throughput automated instrumentation and standardized detection reagents for high sensitivity, limiting its large-scale clinical application. In this study, a high-throughput automated device was developed by combining reagent lyophilization, extraction-free technology, and a one-pot consumable system. This innovative approach enabled the rapid sample-in-result-out detection of 48 samples in 25 min and demonstrated high sensitivity and specificity for the qualitative analysis of clinical samples. The obtained results show that the detection limit of the designed system for African swine fever virus (ASFV) is 0.5 copies/μL. As a proof concept, a single-tube dual-target nucleic acid detection method was developed, achieving a detection limit of 5 copies/μL for the ORF1ab and N genes of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) within 45 min. The method is highly specific, reliable, and stable, providing a feasible solution for the clinical application of CRISPR nucleic acid detection technology.
Collapse
Affiliation(s)
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi’an Jiaotong University, Xi’an 710054, China; (F.W.); (Y.Z.); (X.L.); (Q.M.); (X.W.); (N.P.)
| | | | | | | | | | | |
Collapse
|
47
|
Filion A, Sundaram M, Schmidt JP, Drake JM, Stephens PR. Evidence of repeated zoonotic pathogen spillover events at ecological boundaries. Front Public Health 2024; 12:1435233. [PMID: 39568607 PMCID: PMC11577354 DOI: 10.3389/fpubh.2024.1435233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/01/2024] [Indexed: 11/22/2024] Open
Abstract
Anthropogenic modifications to the landscape have altered several ecological processes worldwide, creating new ecological boundaries at the human/wildlife interface. Outbreaks of zoonotic pathogens often occur at these ecological boundaries, but the mechanisms behind new emergences remain drastically understudied. Here, we test for the influence of two types of ecosystem boundaries on spillover risk: (1) biotic transition zones such as species range edges and transitions between ecoregions and (2) land use transition zones where wild landscapes occur in close proximity to heavily impacted areas of high human population density. Using ebolavirus as a model system and an ensemble machine learning modeling framework, we investigated the role of likely reservoir (bats) and accidental host (primates) range edges and patterns of land use (defined using SEDAC categories) on past spillover events. Our results show that overlapping species range edges and heightened habitat diversity increase ebolavirus outbreaks risk. Moreover, we show that gradual transition zones, represent by high proportion of rangelands, acts as a buffer to reduces outbreak risks. With increasing landscape changes worldwide, we provide novel ecological and evolutionary insights into our understanding of zoonotic pathogen emergence and highlight the risk of aggressively developing ecological boundaries.
Collapse
Affiliation(s)
- Antoine Filion
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| | - Mekala Sundaram
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
- Savannah River Ecology Laboratory, University of Georgia, Aiken, SC, United States
| | - John Paul Schmidt
- Odum School of Ecology and Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - John M Drake
- Odum School of Ecology and Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Patrick R Stephens
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
48
|
Bates JN, Kamara A, Bereteh MS, Barrera D, Moses L, Sheriff A, Sesay F, Yillah MS, Grant DS, Lamin J, Anglewicz P. Long-term physical and mental health outcomes of Ebola Virus Disease survivors in Kenema District, Sierra Leone: A cross-sectional survey. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003421. [PMID: 39495749 PMCID: PMC11534246 DOI: 10.1371/journal.pgph.0003421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/02/2024] [Indexed: 11/06/2024]
Abstract
The 2013-2016 Ebola Virus Disease (EVD) epidemic in West Africa was the deadliest in history, with over 28,000 cases. Numerous physical and mental health symptoms have been reported in EVD survivors, although there is limited prior research on how the health of survivors compares to the general population. We conducted a survey of EVD survivors in Kenema District, Sierra Leone and a population-based sample of community members who lived in EVD-affected areas but were not diagnosed with EVD, and compared resulting data about self-reported symptoms, duration, and severity between EVD survivors and community members through multivariate regression models. This study found that more than six years after the epidemic, survivors were significantly more likely to experience both physical and mental health symptoms than community members, with respective adjusted incidence rate ratios (IRRs) of 2.65 (95% CI, 2.28-3.09), p < 0.001, and 11.95 (95% CI, 6.58-21.71), p < 0.001. The most highly reported physical health symptoms experienced by EVD survivors were joint pain (75.5%), headaches (67.3%), and vision problems (44.5%), and the most prevalent psychological symptoms were spells of terror and panic (25.5%) and difficulty falling asleep or staying asleep (20.0%). EVD survivors were significantly more likely than community members to report the symptoms as lasting for a longer period, a median of 6.0 (3.0-7.0) years, and with higher severity. The results indicated that six years after the epidemic, EVD survivors in Kenema District, Sierra Leone are experiencing worse physical and mental health than their peers. These findings of the long-term, debilitating health issues following EVD infection should be considered when designing and implementing future epidemic responses.
Collapse
Affiliation(s)
- Jenna N. Bates
- School of Global Health, University of Copenhagen, Copenhagen, Denmark
| | - Abdulai Kamara
- School of Community Health Sciences, Njala University, Bo, Sierra Leone
| | | | - Denise Barrera
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Lina Moses
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Allieu Sheriff
- Sierra Leone Association of Ebola Survivors, Kenema, Sierra Leone
| | - Fudia Sesay
- Sierra Leone Association of Ebola Survivors, Kenema, Sierra Leone
| | | | - Donald S. Grant
- Kenema Government Hospital, Kenema, Sierra Leone
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Joseph Lamin
- School of Community Health Sciences, Njala University, Bo, Sierra Leone
| | - Philip Anglewicz
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
49
|
Li J, Eagles DA, Tucker IJ, Pereira Schmidt AC, Deplazes E. Secondary structure propensities of the Ebola delta peptide E40 in solution and model membrane environments. Biophys Chem 2024; 314:107318. [PMID: 39226875 DOI: 10.1016/j.bpc.2024.107318] [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: 02/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The Ebola delta peptide is an amphipathic, 40-residue peptide encoded by the Ebola virus, referred to as E40. The membrane-permeabilising activity of the E40 delta peptide has been demonstrated in cells and lipid vesicles suggesting the E40 delta peptide likely acts as a viroporin. The lytic activity of the peptide increases in the presence of anionic lipids and a disulphide bond in the C-terminal part of the peptide. Previous in silico work predicts the peptide to show a partially helical structure, but there is no experimental information on the structure of E40. Here, we use circular dichroism spectroscopy to report the secondary structure propensities of the reduced and oxidised forms of the E40 peptide in water, detergent micelles, and lipid vesicles composed of neutral and anionic lipids (POPC and POPG, respectively). Results indicate that the peptide is predominately a random coil in solution, and the disulphide bond has a small but measurable effect on peptide conformation. Secondary structure analysis shows large uncertainties and dependence on the reference data set and, in our system, cannot be used to accurately determine the secondary structure motifs of the peptide in membrane environments. Nevertheless, the spectra can be used to assess the relative changes in secondary structure propensities of the peptide depending on the solvent environment and disulphide bond. In POPC-POPG vesicles, the peptide transitions from a random coil towards a more structured conformation, which is even more pronounced in negatively charged SDS micelles. In vesicles, the effect depends on the peptide-lipid ratio, likely resulting from vesicle surface saturation. Further experiments with zwitterionic POPC vesicles and DPC micelles show that both curvature and negatively charged lipids can induce a change in conformation, with the two effects being cumulative. Electrostatic screening from Na+ ions reduced this effect. The oxidised form of the peptide shows a slightly lower propensity for secondary structure and retains a more random coil conformation even in the presence of PG-PC vesicles.
Collapse
Affiliation(s)
- Jiayu Li
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - David A Eagles
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Isaac J Tucker
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | | | - Evelyne Deplazes
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia.
| |
Collapse
|
50
|
Branda F, Pavia G, Ciccozzi A, Quirino A, Marascio N, Matera G, Romano C, Locci C, Azzena I, Pascale N, Sanna D, Casu M, Ceccarelli G, Ciccozzi M, Scarpa F. Zoonotic Paramyxoviruses: Evolution, Ecology, and Public Health Strategies in a Changing World. Viruses 2024; 16:1688. [PMID: 39599803 PMCID: PMC11599060 DOI: 10.3390/v16111688] [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: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The family Paramyxoviridae includes a number of negative RNA viruses known for their wide host range and significant zoonotic potential. In recent years, there has been a surge in the identification of emerging zoonotic paramyxoviruses, particularly those hosted by bat species, which serve as key reservoirs. Among these, the genera Henipavirus and Pararubulavirus are of particular concern. Henipaviruses, including the highly pathogenic Hendra and Nipah viruses, have caused severe outbreaks with high mortality rates in both humans and animals. In contrast, zoonotic pararubulaviruses such as the Menangle virus typically induce mild symptoms or remain asymptomatic in human hosts. This review summarizes current knowledge on the evolution, ecology, and epidemiology of emerging zoonotic paramyxoviruses, focusing on recently discovered viruses and their potential to cause future epidemics. We explore the molecular mechanisms underlying host-switching events, viral replication strategies, and immune evasion tactics that facilitate interspecies transmission. In addition, we discuss ecological factors influencing virus emergence, including changes in bat populations and habitats and the role of wildlife-human interfaces. We also examine the public health impact of these emerging viruses, underlining the importance of enhanced surveillance, developing improved diagnostic tools, and implementing proactive strategies to prevent potential outbreaks. By providing a comprehensive overview of recent advances and gaps in knowledge, this review aims to inform future research directions and public health policies related to zoonotic paramyxoviruses.
Collapse
Affiliation(s)
- Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Alessandra Ciccozzi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Chiara Locci
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Ilenia Azzena
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Noemi Pascale
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
- Department of Chemical Physical Mathematical and Natural Sciences, University of Sassari, 07100 Sassari, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University Hospital Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy;
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| |
Collapse
|