1
|
Eslamkhah S, Aslan ES, Yavas C, Akcalı N, Batur LK, Abuaisha A, Yildirim EE, Solak M, White KN. Mpox virus (MPXV): comprehensive analysis of pandemic risks, pathophysiology, treatments, and mRNA vaccine development. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6143-6163. [PMID: 39777535 DOI: 10.1007/s00210-024-03649-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Mpox, formerly known as monkeypox, is a zoonotic disease caused by the Mpox virus (MPXV), which has recently attracted global attention due to its potential for widespread outbreaks. Initially identified in 1958, MPXV primarily spreads to humans through contact with infected wild animals, particularly rodents. Historically confined to Africa, the virus has expanded beyond endemic regions, with notable outbreaks in Europe and North America in 2022, especially among men who have sex with men (MSM). The World Health Organization (WHO) has declared the current Mpox outbreak a Public Health Emergency of International Concern. This review explores the epidemiology, pathophysiology, and clinical manifestations of MPXV, along with current treatment strategies and the role of mRNA vaccines. It emphasizes the importance of understanding the changing dynamics of Mpox transmission, which are influenced by factors such as waning immunity from smallpox vaccinations and increased global interconnectedness. The potential for developing multi-epitope vaccines that can stimulate robust immune responses is highlighted, showcasing how bioinformatics can facilitate the identification of immunogenic antigens. Continued research and investment in vaccine development are crucial to address the urgent need for effective candidates that can protect at-risk populations. In summary, this review underscores the necessity for proactive public health measures and collaborative efforts among healthcare authorities, researchers, and communities to mitigate the impact of Mpox and enhance global preparedness for future outbreaks.
Collapse
Affiliation(s)
- Sajjad Eslamkhah
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Elif Sibel Aslan
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Cuneyd Yavas
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Nermin Akcalı
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Lutfiye Karcıoglu Batur
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Asmaa Abuaisha
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Erva Esma Yildirim
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34015, Turkey
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey
| | - Mustafa Solak
- Biruni University Research Center (B@MER), Biruni University, Istanbul, 34015, Turkey.
- Department of Medical Genetics, Faculty of Medicine, Biruni University, Merkezefendi Mahallesi G/75 Sk. No: 1-13, Zeytinburnu, Istanbul, 34010, Turkey.
| | - Kenneth N White
- School of Human Sciences, London Metropolitan University, London, UK
| |
Collapse
|
2
|
Wu J, Zhang X. Exploring monkeypox virus antibody levels: insights from human immunological research. Virol J 2025; 22:175. [PMID: 40450351 DOI: 10.1186/s12985-025-02748-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/19/2025] [Indexed: 06/03/2025] Open
Abstract
Monkeypox(mpox) is a zoonotic disease caused by the monkeypox virus (MPXV), which was previously endemic to West and Central Africa. However, it has recently appeared in several non-endemic countries beyond Africa. On July 23, 202 WHO declared mpox outbreak a public health emergency of international concern, a declaration reaffirmed on August 14, 2024. In this context, understanding the antibody levels of MPXV in the population has become crucial, especially given the historical cross-protection provided by smallpox vaccination. To provide a comprehensive overview of the current understanding of MPXV antibody levels and the protective efficacy of smallpox vaccination, we conducted a review of the existing literature. We reviewed relevant studies published in peer-reviewed journals from 1958 to 2025, focusing on those that reported research on MPXV antibodies and the effects of smallpox vaccination. Here, we review the research progress of MPXV and smallpox virus(VARV) in epidemiology, etiology, mutation and mechanism of virus infection, clinical characteristics and vaccine application. In addition, the differences in MPXV levels in different populations and the cross-protective effect of smallpox vaccine against mpox were also discussed. Our review indicates that MPXV antibody levels are closely related to the level of immunity in the population, particularly among individuals who have received smallpox vaccination. This narrative review aims to synthesize existing evidence on the role of smallpox vaccination in protecting against mpox and to offer evidence-based guidance for public health policy. We aim to establish a theoretical foundation and practical recommendations for future research and mpox prevention strategies.
Collapse
Affiliation(s)
- Jing Wu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Xiaomin Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
3
|
Kurosawa N, Ozawa T, Ozawa K, Shimojima M, Kawahara M, Kasuya F, Okada W, Nagashima M, Sadamasu K, Itamochi M, Tani H, Morinaga Y, Yuhara K, Okamoto J, Ichikawa H, Kawahata T, Yamazaki T, Isobe M. Development and clinical evaluation of a MPXV antigen-detecting rapid diagnostic test. J Virol Methods 2025; 337:115164. [PMID: 40345596 DOI: 10.1016/j.jviromet.2025.115164] [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: 08/02/2024] [Revised: 03/15/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
To address the global emergence of mpox after the 2022 epidemic, a rapid and accurate diagnostic tool is needed at the point of care to identify individuals infected with mpox virus (MPXV) to prevent and control the spread of the virus. We designed an antigen-detecting rapid diagnostic test that exclusively detects MPXV without cross-reacting with the vaccinia virus by developing monoclonal antibodies against the MPXV nuclear capsid protein A5L (MPXV-A5L). The test results indicated that the detection limits were established at 0.5 ng/mL for MPXV-A5L and 4.4 × 102 ∼ 2.1 × 103 pfu/mL for MPXV culture fluid. Clinical samples collected from MPXV patients showed a high sensitivity of 87 % at a qPCR cycle threshold of 25 or lower, with a specificity of 100 % for samples that tested negative in the qPCR. The test is an ideal rapid diagnostic tool for supporting clinical decision-making for people suspected of having MPXV infection in resource-poor settings.
Collapse
Affiliation(s)
- Nobuyuki Kurosawa
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan; Center for Advanced Antibody Drug Development, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan.
| | - Tatsuhiko Ozawa
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan; Center for Advanced Antibody Drug Development, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan
| | - Kousei Ozawa
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Graduate School of Pharma-Medical Sciences, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Madoka Kawahara
- Department of Virology I, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Fumi Kasuya
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shinjuku-ku, Tokyo, Japan
| | - Wakaba Okada
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shinjuku-ku, Tokyo, Japan
| | - Mami Nagashima
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shinjuku-ku, Tokyo, Japan
| | - Kenji Sadamasu
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shinjuku-ku, Tokyo, Japan
| | - Masae Itamochi
- Department of Virology, Toyama Institute of Health, 7-1 Nakataikoyama, Imizu-shi, Toyama 939-0363, Japan
| | - Hideki Tani
- Department of Virology, Toyama Institute of Health, 7-1 Nakataikoyama, Imizu-shi, Toyama 939-0363, Japan
| | - Yoshitomo Morinaga
- Center for Advanced Antibody Drug Development, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan; Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kosuke Yuhara
- Biotechnology Research Laboratory, TOYOBO Co., Ltd., 10-24, Toyo-Cho, Tsuruga-Shi, Fukui 914-8550, Japan
| | - Jun Okamoto
- Biotechnology Research Laboratory, TOYOBO Co., Ltd., 10-24, Toyo-Cho, Tsuruga-Shi, Fukui 914-8550, Japan
| | - Haruna Ichikawa
- Biotechnology Research Laboratory, TOYOBO Co., Ltd., 10-24, Toyo-Cho, Tsuruga-Shi, Fukui 914-8550, Japan
| | - Takashi Kawahata
- Biotechnology Research Laboratory, TOYOBO Co., Ltd., 10-24, Toyo-Cho, Tsuruga-Shi, Fukui 914-8550, Japan
| | - Tomomi Yamazaki
- Biotechnology Research Laboratory, TOYOBO Co., Ltd., 10-24, Toyo-Cho, Tsuruga-Shi, Fukui 914-8550, Japan
| | - Masaharu Isobe
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan; Center for Advanced Antibody Drug Development, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama 930-8555, Japan
| |
Collapse
|
4
|
Zhang Y, Wan Y, Guo C, Zhu Z, Qiu C, Lu J, Zhou Y, Zheng J, Dai F, Cheng X, Deng K, Wang W, Wang Y, Zhang W. Novel derivatives of brincidofovir and (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine inhibit orthopoxviruses and human adenoviruses more potently than brincidofovir. Signal Transduct Target Ther 2025; 10:114. [PMID: 40210872 PMCID: PMC11985979 DOI: 10.1038/s41392-025-02207-w] [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: 11/01/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 04/12/2025] Open
Abstract
Brincidofovir (BCV) and tecovirimat are the only two chemical drugs that have been approved to treat smallpox and can be requested for monkeypox (Mpox) treatment through a single-patient Emergency Investigational New Drug (EIND) application. Disappointedly, the efficacy of tecovirimat manifested in recent clinical trials is far from being satisfactory, while the clinical efficacy of BCV is still inconclusive. Given that monkeypox virus (MPXV), variola and other emerging orthopoxviruses are posing serious threats to global health, it is urgent to develop better therapeutics. In this study, we tested the antiviral effects of three novel prodrugs, which were designed based on previously reported parent drugs, either (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine ((S)-HPMPC, cidofovir) or (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine ((S)-HPMPA). We found that one of the (S)-HPMPA-based prodrugs, ODE-(S)-HPMPA formate, exhibited significantly better anti-orthopoxvirus activity than BCV both in vitro and in vivo, which also inhibited human adenovirus type 2 and type 21 more efficiently than BCV. Most strikingly, the EC50 and EC90 of ODE-(S)-HPMPA formate against MPXV were more than 40-fold lower than those of BCV. In contrast, we observed that the anti-herpes simplex virus type 1 (HSV-1) activities of the (S)-HPMPA-based prodrugs were less effective than those of the cidofovir-based prodrugs (BCV and BCV formate), especially in vivo. Moreover, we showed for the first time that cytidine and adenine analog combined therapies could provide mice with complete protection against lethal challenges of both vaccinia and HSV-1. Collectively, we propose that both the ODE-(S)-HPMPA formate and the BCV/ODE-(S)-HPMPA formate combination are worth further investigations for their potential clinical applications.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
- Department of laboratory medicine, Shanghai Public Health Clinical Center, Shanghai, China
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yanmin Wan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China.
- Department of radiology, Shanghai Public Health Clinical Center, Shanghai, China.
| | - Cuiyuan Guo
- Department of laboratory medicine, Shanghai Public Health Clinical Center, Shanghai, China
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, China
| | - Zhaoqin Zhu
- Department of laboratory medicine, Shanghai Public Health Clinical Center, Shanghai, China
- Biosafety Level 3 Laboratory, Shanghai Public Health Clinical Center, Shanghai, China
| | - Chao Qiu
- Institutes of biomedical sciences & Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Jiasheng Lu
- School of Life Sciences, Fudan University, Shanghai, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Risen (Shanghai) Pharma Tech Co. Ltd., Shanghai, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jiaojiao Zheng
- Biosafety Level 3 Laboratory, Shanghai Public Health Clinical Center, Shanghai, China
| | - Fahui Dai
- Biosafety Level 3 Laboratory, Shanghai Public Health Clinical Center, Shanghai, China
| | - Xiaoyang Cheng
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
- Department of laboratory medicine, Shanghai Public Health Clinical Center, Shanghai, China
| | - Kunlu Deng
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
- Department of laboratory medicine, Shanghai Public Health Clinical Center, Shanghai, China
| | - Wanhai Wang
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, China
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China.
| |
Collapse
|
5
|
Li Y, Wang L, Chen S. An overview of the progress made in research into the Mpox virus. Med Res Rev 2025; 45:788-812. [PMID: 39318037 DOI: 10.1002/med.22085] [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: 03/26/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Mpox is a zoonotic illness caused by the Mpox virus (MPXV), a member of the Orthopoxvirus family. Although a few cases have been reported outside Africa, it was originally regarded as an endemic disease limited to African countries. However, the Mpox outbreak of 2022 was remarkable in that the infection spread to more than 123 countries worldwide, causing thousands of infections and deaths. The ongoing Mpox outbreak has been declared as a public health emergency of international concern by the World Health Organization. For a better management and control of the epidemic, this review summarizes the research advances and important scientific findings on MPXV by reviewing the current literature on epidemiology, clinical characteristics, diagnostic methods, prevention and treatment measures, and animal models of MPXV. This review provides useful information to raise awareness about the transmission, symptoms, and protective measures of MPXV, serving as a theoretical guide for relevant institutions to control MPXV.
Collapse
Affiliation(s)
- Yansheng Li
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lianrong Wang
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Shi Chen
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
6
|
Tseng KK, Koehler H, Becker DJ, Gibb R, Carlson CJ, Pilar Fernandez MD, Seifert SN. Viral genomic features predict Orthopoxvirus reservoir hosts. Commun Biol 2025; 8:309. [PMID: 40000824 PMCID: PMC11862092 DOI: 10.1038/s42003-025-07746-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Orthopoxviruses (OPVs), including the causative agents of smallpox and mpox have led to devastating outbreaks in human populations worldwide. However, the discontinuation of smallpox vaccination, which also provides cross-protection against related OPVs, has diminished global immunity to OPVs more broadly. We apply machine learning models incorporating both host ecological and viral genomic features to predict likely reservoirs of OPVs. We demonstrate that incorporating viral genomic features in addition to host ecological traits enhanced the accuracy of potential OPV host predictions, highlighting the importance of host-virus molecular interactions in predicting potential host species. We identify hotspots for geographic regions rich with potential OPV hosts in parts of southeast Asia, equatorial Africa, and the Amazon, revealing high overlap between regions predicted to have a high number of potential OPV host species and those with the lowest smallpox vaccination coverage, indicating a heightened risk for the emergence or establishment of zoonotic OPVs. Our findings can be used to target wildlife surveillance, particularly related to concerns about mpox establishment beyond its historical range.
Collapse
Affiliation(s)
- Katie K Tseng
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Heather Koehler
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| | - Daniel J Becker
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| | - Rory Gibb
- Centre for Biodiversity and Environment Research, Department of Genetics, Evolution and Environment, University College London, London, UK
- People & Nature Lab, UCL East, University College London, London, UK
| | - Colin J Carlson
- Department of Epidemiology of Microbial Diseases, Yale University School of Public Health, New Haven, CT, USA
| | | | - Stephanie N Seifert
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.
| |
Collapse
|
7
|
Zhu J, Jia X, Ren S, Zhang Z, Li H, Wang J, Song B, Wu W, Peng C. Inhibition of Polo-Like Kinase 1 Dampens the Replication of Vaccinia Virus in Mammalian Cells. J Med Virol 2025; 97:e70240. [PMID: 39953955 DOI: 10.1002/jmv.70240] [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: 10/20/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Since the eradication of smallpox, zoonotic poxviruses, such as the mpox virus (MPXV), continue to pose a threat to public health. Identifying drugs that reduce poxvirus infection and replication, as well as understanding their molecular mechanisms, is essential for epidemic control. Polo-like kinase 1 (PLK1) has been shown to facilitate vaccinia virus (VACV) infection and replication. This study confirms the effects of the PLK1 inhibitors HMN-214 and ON-01910 on VACV replication in A549 cells. Both viral titers and DNA loads were significantly reduced in treated cells after infection. Additionally, ON-01910 demonstrated broad-spectrum antiviral activity against the lumpy skin disease virus (LSDV) and the infectious bovine rhinotracheitis virus (IBRV) in vitro. PLK1 knockdown in A549 cells also led to a reduction in VACV protein expression, viral titers, and DNA levels. Further analysis showed that VACV infection leads to the accumulation of PLK1 near viral factories. However, despite its strong in vitro effects, ON-01910 did not significantly reduce VACV replication in mice. These findings highlight the critical role of PLK1 in VACV replication and its potential as a target for antiviral therapy against orthopoxviruses.
Collapse
Affiliation(s)
- Junda Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xuejiao Jia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuning Ren
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zihui Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hua Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baifen Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenxue Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chen Peng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
8
|
Andersen-Ranberg E, Nymo IH, Jokelainen P, Emelyanova A, Jore S, Laird B, Davidson RK, Ostertag S, Bouchard E, Fagerholm F, Skinner K, Acquarone M, Tryland M, Dietz R, Abass K, Rautio A, Hammer S, Evengård B, Thierfelder T, Stimmelmayr R, Jenkins E, Sonne C. Environmental stressors and zoonoses in the Arctic: Learning from the past to prepare for the future. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:176869. [PMID: 39423885 DOI: 10.1016/j.scitotenv.2024.176869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The risk of zoonotic disease transmission from animals to humans is elevated for people in close contact with domestic and wild animals. About three-quarters of all known human infectious diseases are zoonotic, and potential health impacts of these diseases are higher where infectious disease surveillance and access to health care and public health services are limited. This is especially the case for remote circumarctic regions, where drivers for endemic, emerging, and re-emerging zoonotic diseases include anthropogenic influences, such as pollution by long-range transport of industrial chemicals, climate change, loss of biodiversity and ecosystem alterations. In addition to these, indirect effects including natural changes in food web dynamics, appearance of invasive species and thawing permafrost also affect the risk of zoonotic disease spill-over. In other words, the Arctic represents a changing world where pollution, loss of biodiversity and habitat, and maritime activity are likely driving forward occurrence of infectious diseases. As a broad international consortium with a wide range of expertise, we here describe a selection of case studies highlighting the importance of a One Health approach to zoonoses in the circumarctic, encompassing human health, animal health, and environmental health aspects. The cases highlight critical gaps in monitoring and current knowledge, focusing on environmental stressors and lifestyle factors, and they are examples of current occurrences in the Arctic that inform on critically needed actions to prepare us for the future. Through these presentations, we recommend measures to enhance awareness and management of existing and emerging zoonoses with epidemic and pandemic potential while also focusing on the impacts of various environmental stressors and lifestyle factors on zoonoses in the Arctic.
Collapse
Affiliation(s)
- Emilie Andersen-Ranberg
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, Dyrlægevej 16, 1870 Frederiksberg, Denmark.
| | - Ingebjørg H Nymo
- Norwegian Veterinary Institute, Holtveien 66, 9016 Tromsø, Norway; Department of Arctic and Marine Biology, UiT - The Arctic University of Norway, Framstredet 39, Breivika, 9019 Tromsø, Norway
| | - Pikka Jokelainen
- Infectious Disease Preparedness, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Anastasia Emelyanova
- Thule Institute, University of Oulu, Paavo Havaksen tie 3, 90570 Oulu, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland
| | - Solveig Jore
- Department of Zoonotic, Food & Waterborne Infections, Norwegian Institute of Public Health, Postbox 222 Skøyen, 0213 Oslo, Norway
| | - Brian Laird
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | | | - Sonja Ostertag
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | - Emilie Bouchard
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, St Hyacinthe J2T 1B3, Canada; Department of Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon S7N 5B4, Canada
| | - Freja Fagerholm
- Department of Clinical Microbiology and the Arctic Center, Umeå University, Johan Bures Väg 5, 90187 Umeå, Sweden
| | - Kelly Skinner
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | - Mario Acquarone
- Arctic Monitoring and Assessment Programme, Hjalmar Johansens gate 14, 9007 Tromsø, Norway
| | - Morten Tryland
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, Anne Evenstads Veg 80, 2480 Koppang, Norway
| | - Rune Dietz
- Aarhus University, Faculty of Technological Sciences, Department of Ecoscience, Frederiksborgvej 399, 4000 Roskilde, Denmark
| | - Khaled Abass
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland; Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, postbox 27272, United Arab Emirates
| | - Arja Rautio
- Thule Institute, University of Oulu, Paavo Havaksen tie 3, 90570 Oulu, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland
| | - Sjúrður Hammer
- Faroese Environment Agency, Traðagøta 38, 165 Argir, Faroe Islands; University of the Faroe Islands, Vestara Bryggja 15, 100 Tórshavn, Faroe Islands
| | - Birgitta Evengård
- Department of Clinical Microbiology and the Arctic Center, Umeå University, Johan Bures Väg 5, 90187 Umeå, Sweden
| | - Tomas Thierfelder
- Department of Energy and Technology, Swedish University of Agricultural Sciences, postbox 75651, Uppsala, Sweden
| | - Raphaela Stimmelmayr
- Department of Wildlife management, North Slope Borough, postbox 69, 99723 Utqiagvik, AK, USA
| | - Emily Jenkins
- Department of Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon S7N 5B4, Canada
| | - Christian Sonne
- Aarhus University, Faculty of Technological Sciences, Department of Ecoscience, Frederiksborgvej 399, 4000 Roskilde, Denmark.
| |
Collapse
|
9
|
Tseng KK, Koehler H, Becker DJ, Gibb R, Carlson CJ, del Pilar Fernandez M, Seifert SN. Viral genomic features predict Orthopoxvirus reservoir hosts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.26.564211. [PMID: 37961540 PMCID: PMC10634857 DOI: 10.1101/2023.10.26.564211] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Orthopoxviruses (OPVs), including the causative agents of smallpox and mpox have led to devastating outbreaks in human populations worldwide. However, the discontinuation of smallpox vaccination, which also provides cross-protection against related OPVs, has diminished global immunity to OPVs more broadly. We apply machine learning models incorporating both host ecological and viral genomic features to predict likely reservoirs of OPVs. We demonstrate that incorporating viral genomic features in addition to host ecological traits enhanced the accuracy of potential OPV host predictions, highlighting the importance of host-virus molecular interactions in predicting potential host species. We identify hotspots for geographic regions rich with potential OPV hosts in parts of southeast Asia, equatorial Africa, and the Amazon, revealing high overlap between regions predicted to have a high number of potential OPV host species and those with the lowest smallpox vaccination coverage, indicating a heightened risk for the emergence or establishment of zoonotic OPVs. Our findings can be used to target wildlife surveillance, particularly related to concerns about mpox establishment beyond its historical range.
Collapse
Affiliation(s)
- Katie K. Tseng
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, United States of America
| | - Heather Koehler
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Daniel J. Becker
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Rory Gibb
- Centre for Biodiversity and Environment Research, Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- People & Nature Lab, UCL East, University College London, Stratford, London, United Kindom
| | - Colin J. Carlson
- Center for Global Health Science and Security, Georgetown University, Washington, DC, United States of America
| | - Maria del Pilar Fernandez
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, United States of America
| | - Stephanie N. Seifert
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, United States of America
| |
Collapse
|
10
|
Ehmann R, Donoso Mantke O, McCulloch E, Yousef A, Ricketts A, Staines H, Bugert JJ, Wölfel R, Niesters HGM. International external quality assessment study for detection of monkeypox virus by PCR supporting laboratory preparedness during the 2022-2023 mpox outbreak and beyond. J Clin Virol 2024; 175:105741. [PMID: 39488925 DOI: 10.1016/j.jcv.2024.105741] [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: 04/27/2024] [Revised: 09/20/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Diagnostic capabilities and correspondent External Quality Assessments (EQA) are key for outbreak preparedness. To support diagnostic facilities with a quality assessment of newly established monkeypox virus (MPXV) molecular diagnostic workflows, Quality Control for Molecular Diagnostics (QCMD) and the Bundeswehr Institute of Microbiology (IMB) piloted an international EQA study conducting four challenges from autumn 2022 to summer 2023 during the global mpox outbreak. OBJECTIVES To assess the performance (sensitivity/specificity) of molecular assays used by diagnostic laboratories. STUDY DESIGN Inactivated EQA panels were prepared and distributed containing seven samples of clade Ia and clade IIb MPXV strains at different viral concentrations, two specificity controls with other zoonotic orthopoxviruses (vaccinia and cowpox virus) and a negative control. Assessment was based on reported qualitative testing results. RESULTS In this outbreak-related EQA study, a total of 192 laboratories from 37 countries reported 346 qualitative datasets. Overall, core samples were correctly detected by approximately 92 % of participants in all four challenges. While sensitivity performance was acceptable in at least 90 % of datasets correctly reported even for educational MPXV-positive samples with low viral concentration [102 genome equivalents (GE)/mL], several laboratories reported the educational specificity controls as false positives or were unable to differentiate MPXV from related orthopoxviruses. CONCLUSIONS Mpox is now a globally occurring infection with a demand for quality-assured diagnostic capabilities. The newly established EQA scheme presented here, offers a multi-purpose panel for orthopoxviruses with a focus on MPXV which will continue to ensure diagnostic quality in clinical settings with up-to-date sample panels.
Collapse
Affiliation(s)
- Rosina Ehmann
- Bundeswehr Institute of Microbiology (IMB), Munich, Germany
| | - Oliver Donoso Mantke
- Quality Control for Molecular Diagnostics (QCMD), Unit 5, Technology Terrace, Todd Campus, West of Scotland Science Park, Glasgow G20 0XA, United Kingdom.
| | - Elaine McCulloch
- Quality Control for Molecular Diagnostics (QCMD), Unit 5, Technology Terrace, Todd Campus, West of Scotland Science Park, Glasgow G20 0XA, United Kingdom
| | - Amani Yousef
- Quality Control for Molecular Diagnostics (QCMD), Unit 5, Technology Terrace, Todd Campus, West of Scotland Science Park, Glasgow G20 0XA, United Kingdom
| | - Alastair Ricketts
- Quality Control for Molecular Diagnostics (QCMD), Unit 5, Technology Terrace, Todd Campus, West of Scotland Science Park, Glasgow G20 0XA, United Kingdom
| | - Harry Staines
- Sigma Statistical Services, Balmullo, United Kingdom
| | | | - Roman Wölfel
- Bundeswehr Institute of Microbiology (IMB), Munich, Germany
| | - Hubert G M Niesters
- The University of Groningen, University Medical Center Groningen, Division of Clinical Virology, Department of Medical Microbiology and Infection Prevention, Groningen, the Netherlands
| |
Collapse
|
11
|
Premraj A, Aleyas AG, Nautiyal B, Rasool TJ. First report of a chemokine from camelids: Dromedary CXCL8 is induced by poxvirus and heavy metal toxicity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105261. [PMID: 39241936 DOI: 10.1016/j.dci.2024.105261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Low molecular weight proteins, known as chemokines, facilitate the migration and localization of immune cells to the site of infection and injury. One of the first chemokines identified, CXCL8 functions as a key neutrophil activator, recruiting neutrophils to sites of inflammation. Several viral infections, including zoonotic coronaviruses and poxviruses, have been reported to induce the expression of CXCL8. Dromedary camels are known to harbor several potentially zoonotic pathogens, but critical immune molecules such as chemokines remain unidentified. We report here the identification of CXCL8 from the dromedary camel - the first chemokine identified from camelids. The complete dromedary CXCL8 cDNA sequence as well as the corresponding gene sequence from dromedary and two New World camelids - alpaca and llama were cloned. CXCL8 mRNA expression was relatively higher in PBMC, spleen, lung, intestine, and liver. Poly(I:C) and lipopolysaccharide stimulated CXCL8 expression in vitro, while interferon treatment inhibited it. In vitro infection with potentially zoonotic camelpox virus induced the expression of CXCL8 in camel kidney cells. Toxicological studies on camelids have been limited, and no biomarkers have been identified. Hence, we also evaluated CXCL8 mRNA expression as a potential biomarker to assess heavy metal toxicity in camel kidney cells in vitro. CXCL8 expression was increased after in vitro exposure to heavy metal compounds of cobalt and cadmium, suggesting potential utility as a biomarker for renal toxicity in camels. The results of our study demonstrate that camel CXCL8 plays a significant role in immunomodulatory and induced toxicity responses in dromedary camels.
Collapse
Affiliation(s)
- Avinash Premraj
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Abi George Aleyas
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Binita Nautiyal
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Thaha Jamal Rasool
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates.
| |
Collapse
|
12
|
Mohamed Abdoul-Latif F, Ainane A, Mohamed H, Merito Ali A, Houmed Aboubaker I, Jutur PP, Ainane T. Mpox Resurgence: A Multifaceted Analysis for Global Preparedness. Viruses 2024; 16:1737. [PMID: 39599851 PMCID: PMC11598846 DOI: 10.3390/v16111737] [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: 10/12/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
This study provides an in-depth analysis of mpox, encompassing its history, characteristics, epidemiology, diagnostics, treatment options, and the ongoing evolution of the virus and its transmission dynamics. Mpox, though once successfully eradicated, has re-emerged with new modes of transmission and a broader host range. Genomic analyses have revealed the virus's adaptability, posing challenges for diagnostics and vaccine efficacy. The epidemiology has shifted from sporadic zoonotic transmission in rural Africa to a significant presence in urban areas, particularly impacting high-risk populations. Advancements in diagnostics and therapeutics offer hope, but challenges persist. This work underscores the critical need for enhanced surveillance, vaccination strategies, and continued research to bolster global health systems and preparedness for future outbreaks.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Medicinal Research Institute, Center for Research and Study of Djibouti, Djibouti P.O. Box 486, Djibouti; (H.M.); (A.M.A.); (I.H.A.)
| | - Ayoub Ainane
- Superior School of Technology, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco;
| | - Houda Mohamed
- Medicinal Research Institute, Center for Research and Study of Djibouti, Djibouti P.O. Box 486, Djibouti; (H.M.); (A.M.A.); (I.H.A.)
- Peltier Hospital of Djibouti, Djibouti P.O. Box 2123, Djibouti
| | - Ali Merito Ali
- Medicinal Research Institute, Center for Research and Study of Djibouti, Djibouti P.O. Box 486, Djibouti; (H.M.); (A.M.A.); (I.H.A.)
| | - Ibrahim Houmed Aboubaker
- Medicinal Research Institute, Center for Research and Study of Djibouti, Djibouti P.O. Box 486, Djibouti; (H.M.); (A.M.A.); (I.H.A.)
- Peltier Hospital of Djibouti, Djibouti P.O. Box 2123, Djibouti
| | - Pannaga Pavan Jutur
- Omics of Algae Group, Industrial Biotechnology, International Centre for Genetic Engineering and Bio-Technology, Aruna Asaf Ali Marg, New Delhi 110067, India;
| | - Tarik Ainane
- Superior School of Technology, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco;
| |
Collapse
|
13
|
Pattanaik A, Lodha L, Marate S, K D, Sushma Bhandarkar B, V S, Ashtaputre N, Mani RS. Buffalopox: An emerging zoonotic challenge. Infect Dis Now 2024; 54:104954. [PMID: 39033879 DOI: 10.1016/j.idnow.2024.104954] [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: 03/28/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
As a variant of Vaccinia virus, Buffalopox virus is known to cause Buffalopox disease. In recent times, sporadic outbreaks of the infection in humans have been reported, especially in the endemic countries of Southeast Asia. Though mortality has not been high, associated morbidity is significant. Due to waning cross-protective immunity against smallpox, Buffalopox virus is one of the several orthopox viruses likely to emerge or reemerge. To combat this virus, early recognition, isolation, and management of the infection in animals and humans is of prime importance. In addition, vaccination in animals and humans at risk of acquiring infection is essential as a means of limiting animal-to-animal and animal-to-human spread of the virus. With this in mind, a collaborative approach between the animal and human health sectors is indispensable.
Collapse
Affiliation(s)
- Amrita Pattanaik
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Lonika Lodha
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India
| | - Srilatha Marate
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Dhanya K
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India
| | - B Sushma Bhandarkar
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Sreelakshmi V
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Nidhi Ashtaputre
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Reeta S Mani
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India.
| |
Collapse
|
14
|
Parnian R, Heydarifard F, Mousavi FS, Heydarifard Z, Zandi M. Innate Immune Response to Monkeypox Virus Infection: Mechanisms and Immune Escape. J Innate Immun 2024; 16:413-424. [PMID: 39137733 PMCID: PMC11521483 DOI: 10.1159/000540815] [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: 06/06/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The reemergence of monkeypox virus (Mpox, formerly monkeypox) in 2022 in non-endemic countries has raised significant concerns for global health due to its high transmissibility and mortality rate. A major challenge in combating Mpox is its ability to evade the host's innate immune system, the first line of defense against viral infections. SUMMARY Mpox encodes various proteins that interfere with key antiviral pathways and mechanisms, such as the nuclear factor kappa B signaling, cytokine production, complement and inflammasome activation, and chemokine binding. These proteins modulate the expression and function of innate immune mediators, such as interferons, interleukins, and Toll-like receptors, and impair the recruitment and activation of innate immune cells, such as natural killer cells. By suppressing or altering these innate immune responses, Mpox enhances its replication and infection in the host tissues and organs, leading to systemic inflammation, tissue damage, and organ failure. KEY MESSAGES This study reveals new insights into the molecular and cellular interactions between Mpox and the host's innate immune system. It identifies potential targets and strategies for antiviral interventions, highlighting the importance of understanding these interactions to develop effective treatments and improve global health responses to Mpox outbreaks.
Collapse
Affiliation(s)
- Reza Parnian
- Department of Virology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fatemeh Heydarifard
- Department of Veterinary, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Fatemeh Sadat Mousavi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Heydarifard
- Department of Virology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Milad Zandi
- Department of Microbiology, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
15
|
Shchelkunov SN, Yakubitskiy SN, Titova KA, Pyankov SA, Shulgina IS, Starostina EV, Borgoyakova MB, Kisakov DN, Karpenko LI, Shchelkunova GA, Sergeev AA. An Attenuated and Highly Immunogenic Variant of the Vaccinia Virus. Acta Naturae 2024; 16:82-89. [PMID: 39188266 PMCID: PMC11345087 DOI: 10.32607/actanaturae.27384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/05/2024] [Indexed: 08/28/2024] Open
Abstract
The vaccinia virus (VACV) has been used for prophylactic immunization against smallpox for many decades. However, the VACV-based vaccine had been highly reactogenic. Therefore, after the eradication of smallpox, the World Health Organization in 1980 recommended that vaccination against this infection be discontinued. As a result, there has been a rise in the occurrence of orthopoxvirus infections in humans in recent years, with the most severe being the 2022 monkeypox epidemic that reached all continents. Thus, it is crucial to address the pressing matter of developing safe and highly immunogenic vaccines for new generations to combat orthopoxvirus infections. In a previous study, we created a LAD strain by modifying the LIVP (L) VACV strain, which is used as a first-generation smallpox vaccine in Russia. This modification involved introducing mutations in the A34R gene to enhance extracellular virion production and deleting the A35R gene to counteract the antibody response to the viral infection. In this study, a strain LADA was created with an additional deletion in the DNA of the LAD strain ati gene. This ati gene directs the production of a major non-virion immunogen. The findings indicate that the LADA VACV variant exhibits lower levels of reactogenicity in BALB/c mice during intranasal infection, as compared to the original L strain. Following intradermal immunization with a 105 PFU dose, both the LAD and LADA strains were found to induce a significantly enhanced cellular immune response in mice when compared to the L strain. At the same time, the highest level of virus-specific IFN-γ producing cells for the LAD variant was detected on the 7th day post-immunization (dpi), whereas for LADA, it was observed on 14 dpi. The LAD and LADA strains induced significantly elevated levels of VACV-specific IgG compared to the original L strain, particularly between 28 and 56 dpi. The vaccinated mice were intranasally infected with the cowpox virus at a dose of 460 LD50 to assess the protective immunity at 62 dpi. The LADA virus conferred complete protection to mice, with the LAD strain providing 70% protection and the parent strain L offering protection to only 60% of the animals.
Collapse
Affiliation(s)
- S. N. Shchelkunov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - S. N. Yakubitskiy
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - K. A. Titova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - S. A. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - I. S. Shulgina
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - E. V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - M. B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - D. N. Kisakov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - L. I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - G. A. Shchelkunova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| | - A. A. Sergeev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russian Federation
| |
Collapse
|
16
|
Sun Y, Nie W, Tian D, Ye Q. Human monkeypox virus: Epidemiologic review and research progress in diagnosis and treatment. J Clin Virol 2024; 171:105662. [PMID: 38432097 DOI: 10.1016/j.jcv.2024.105662] [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: 12/22/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Monkeypox virus (MPXV) is responsible for causing a zoonotic disease called monkeypox (mpox), which sporadically infects humans in West and Central Africa. It first infected humans in 1970 and, along with the variola virus, belongs to the genus Orthopoxvirus in the poxvirus family. Since the World Health Organization declared the MPXV outbreak a "Public Health Emergency of International Concern" on July 23, 2022, the number of infected patients has increased dramatically. To control this epidemic and address this previously neglected disease, MPXV needs to be better understood and reevaluated. In this review, we cover recent research on MPXV, including its genomic and pathogenic characteristics, transmission, mutations and mechanisms, clinical characteristics, epidemiology, laboratory diagnosis, and treatment measures, as well as prevention of MPXV infection in light of the 2022 and 2023 global outbreaks. The 2022 MPXV outbreak has been primarily associated with close intimate contact, including sexual activity, with most cases diagnosed among men who have sex with men. The incubation period of MPXV infection usually lasts from 6 to 13 days, and symptoms include fever, muscle pains, headache, swollen lymph nodes, and a characteristic painful rash, including several stages, such as macules, papules, blisters, pustules, scabs, and scab shedding involving the genitals and anus. Polymerase chain reaction (PCR) is usually used to detect MPXV in skin lesion material. Treatment includes supportive care, antivirals, and intravenous vaccinia immune globulin. Smallpox vaccines have been designed with four givens emergency approval for use against MPXV infection.
Collapse
Affiliation(s)
- Yanhong Sun
- Department of Clinical Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Wenjian Nie
- Department of Clinical Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Dandan Tian
- Department of Clinical Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Qing Ye
- Department of Clinical Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China.
| |
Collapse
|
17
|
Yang CH, Song AL, Qiu Y, Ge XY. Cross-species transmission and host range genes in poxviruses. Virol Sin 2024; 39:177-193. [PMID: 38272237 PMCID: PMC11074647 DOI: 10.1016/j.virs.2024.01.007] [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: 06/20/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The persistent epidemic of human mpox, caused by mpox virus (MPXV), raises concerns about the future spread of MPXV and other poxviruses. MPXV is a typical zoonotic virus which can infect human and cause smallpox-like symptoms. MPXV belongs to the Poxviridae family, which has a relatively broad host range from arthropods to vertebrates. Cross-species transmission of poxviruses among different hosts has been frequently reported and resulted in numerous epidemics. Poxviruses have a complex linear double-strand DNA genome that encodes hundreds of proteins. Genes related to the host range of poxvirus are called host range genes (HRGs). This review briefly introduces the taxonomy, phylogeny and hosts of poxviruses, and then comprehensively summarizes the current knowledge about the cross-species transmission of poxviruses. In particular, the HRGs of poxvirus are described and their impacts on viral host range are discussed in depth. We hope that this review will provide a comprehensive perspective about the current progress of researches on cross-species transmission and HRG variation of poxviruses, serving as a valuable reference for academic studies and disease control in the future.
Collapse
Affiliation(s)
- Chen-Hui Yang
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China
| | - A-Ling Song
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China
| | - Ye Qiu
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China.
| | - Xing-Yi Ge
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China.
| |
Collapse
|
18
|
Jacob-Dolan C, Ty D, Hope D, McMahan K, Liu J, Powers OC, Cotter CA, Sciacca M, Wu C, Borducchi E, Bouffard E, Richter H, Velasco J, Teow E, Boursiquot M, Cook A, Feliciano K, Yalley-Ogunro J, Seaman MS, Pessiant L, Lewis MG, Andersen H, Moss B, Barouch DH. Comparison of the immunogenicity and protective efficacy of ACAM2000, MVA, and vectored subunit vaccines for Mpox in rhesus macaques. Sci Transl Med 2024; 16:eadl4317. [PMID: 38536937 DOI: 10.1126/scitranslmed.adl4317] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/29/2024] [Indexed: 05/23/2025]
Abstract
The 2022-2023 mpox outbreak triggered vaccination efforts using smallpox vaccines that were approved for mpox, including modified vaccinia Ankara (MVA; JYNNEOS), which is a safer alternative to live replicating vaccinia virus (ACAM2000). Here, we compare the immunogenicity and protective efficacy of JYNNEOS by the subcutaneous or intradermal routes, ACAM2000 by the percutaneous route, and subunit Ad35 vector-based L1R/B5R or L1R/B5R/A27L/A33R vaccines by the intramuscular route in rhesus macaques. All vaccines provided robust protection against high-dose intravenous mpox virus challenge with the current outbreak strain, with ACAM2000 providing near complete protection and JYNNEOS and Ad35 vaccines providing robust but incomplete protection. Protection correlated with neutralizing antibody responses as well as L1R/M1R- and B5R/B6R-specific binding antibody responses, although additional immune responses likely also contributed to protection. This study demonstrates the protective efficacy of multiple vaccine platforms against mpox virus challenge, including both current clinical vaccines and vectored subunit vaccines.
Collapse
Affiliation(s)
- Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Darren Ty
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Olivia C Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MA 20852, USA
| | - Michela Sciacca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Cindy Wu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Erica Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Emily Bouffard
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Hannah Richter
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | | | | | | | | | | | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | | | | | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MA 20852, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
19
|
Chakraborty P, Kumar R, Karn S, Raviya DD, Mondal P. Poxviruses as Agents of Biological Warfare: The Importance of Ensuring Ethical Standards for Research with Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:399-412. [PMID: 38801593 DOI: 10.1007/978-3-031-57165-7_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Historically, biological agents have been used to target various populations. One of the earliest examples could be the catastrophic effect of smallpox in Australia in the eighteenth century (as alleged by some historians). Modern biological techniques can be used to both create or provide protection against various agents of biological warfare. Any microorganism (viruses, bacteria, and fungi) or its toxins can be used as biological agents. Minnesota Department of Health has listed Smallpox (variola major) as a category A bioterrorism agent, even though it has been eradicated in 1980 through an extensive vaccination campaign. Category A agents are considered the highest risk to public health. Laboratory-associated outbreaks of poxviruses could cause unprecedented occupational hazards. Only two WHO-approved BSL-4 facilities in the United States and Russia are allowed to perform research on the variola virus. So, poxviruses present themselves as a classical case of a dual-use dilemma, since research with them can be used for both beneficial and harmful purposes. Although the importance of ethics in scientific research requires no further elaboration, ethical norms assume greater significance during experimentation with poxviruses. In this chapter, we will update the readers on the sensitive nature of conducting research with poxviruses, and how these viruses can be a source of potential biological weapons. Finally, specified ethical guidelines are explored to ensure safe research practices in virology.
Collapse
Affiliation(s)
- Prasenjit Chakraborty
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India.
| | - Randhir Kumar
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Sanjay Karn
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Dharmiben D Raviya
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Priya Mondal
- Laboratory of Cell Biology, National Cancer Institute, National Institute of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
20
|
Biernacka Z, Gregorczyk-Zboroch K, Lasocka I, Ostrowska A, Struzik J, Gieryńska M, Toka FN, Szulc-Dąbrowska L. Ectromelia Virus Affects the Formation and Spatial Organization of Adhesive Structures in Murine Dendritic Cells In Vitro. Int J Mol Sci 2023; 25:558. [PMID: 38203729 PMCID: PMC10779027 DOI: 10.3390/ijms25010558] [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: 10/29/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Ectromelia virus (ECTV) is a causative agent of mousepox. It provides a suitable model for studying the immunobiology of orthopoxviruses, including their interaction with the host cell cytoskeleton. As professional antigen-presenting cells, dendritic cells (DCs) control the pericellular environment, capture antigens, and present them to T lymphocytes after migration to secondary lymphoid organs. Migration of immature DCs is possible due to the presence of specialized adhesion structures, such as podosomes or focal adhesions (FAs). Since assembly and disassembly of adhesive structures are highly associated with DCs' immunoregulatory and migratory functions, we evaluated how ECTV infection targets podosomes and FAs' organization and formation in natural-host bone marrow-derived DCs (BMDC). We found that ECTV induces a rapid dissolution of podosomes at the early stages of infection, accompanied by the development of larger and wider FAs than in uninfected control cells. At later stages of infection, FAs were predominantly observed in long cellular extensions, formed extensively by infected cells. Dissolution of podosomes in ECTV-infected BMDCs was not associated with maturation and increased 2D cell migration in a wound healing assay; however, accelerated transwell migration of ECTV-infected cells towards supernatants derived from LPS-conditioned BMDCs was observed. We suggest that ECTV-induced changes in the spatial organization of adhesive structures in DCs may alter the adhesiveness/migration of DCs during some conditions, e.g., inflammation.
Collapse
Affiliation(s)
- Zuzanna Biernacka
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Karolina Gregorczyk-Zboroch
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Iwona Lasocka
- Department of Biology of Animal Environment, Institute of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Justyna Struzik
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Małgorzata Gieryńska
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Felix N. Toka
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| |
Collapse
|
21
|
Andrade FM, Faria GN, Ramos ML, Franco S, Sardinha T, Gouveia S. Beyond Borders: Monkeypox Case on Madeira Island. Cureus 2023; 15:e50715. [PMID: 38234948 PMCID: PMC10792706 DOI: 10.7759/cureus.50715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
Human infection with monkeypox virus (MPXV) is characterized by a pox-like rash in various areas, including the anogenital region, and is accompanied by fever, headaches, fatigue, myalgias, and lymphadenopathy. These symptoms may occur a few days before the rash or simultaneously. Nonspecific and vague symptoms, particularly in the recent outbreak of the MPXV, have led to unrecognized or very mild prodromes, which can delay diagnosis. Diagnosis involves laboratory confirmation through polymerase chain reaction (PCR). The symptomatology of MPX is self-limiting, resolving in about two to four weeks. Therefore, the therapeutic approach includes supportive care, monitoring, intervention for possible complications (e.g., bacterial superinfection, cellulitis, and bronchopneumonia), and the implementation of preventive contact measures. This clinical case emphasizes the importance of conducting a thorough medical history and maintaining a high level of clinical suspicion, even in the absence of a history of contact with suspected or confirmed cases and in regions without active or suspected infectious cases.
Collapse
Affiliation(s)
- Filipa M Andrade
- Family Medicine, Centro de Saúde do Bom Jesus, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Funchal, PRT
| | - Guilherme N Faria
- Family Medicine, Centro de Saúde do Porto da Cruz, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Funchal, PRT
| | - Maria L Ramos
- Family Medicine, Centro de Saúde Dr. Rui Adriano de Freitas, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Funchal, PRT
| | - Susana Franco
- Family Medicine, Centro de Saúde Faial, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Funchal, PRT
| | - Tiago Sardinha
- Family Medicine, Centro de Saúde do Caniço, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Caniço, PRT
| | - Sara Gouveia
- Family Medicine, Centro de Saúde do Bom Jesus, Serviço de Saúde da Região Autónoma da Madeira (SESARAM), Funchal, PRT
| |
Collapse
|
22
|
Li Z, Sinha A, Zhang Y, Tanner N, Cheng HT, Premsrirut P, Carlow CKS. Extraction-free LAMP assays for generic detection of Old World Orthopoxviruses and specific detection of Mpox virus. Sci Rep 2023; 13:21093. [PMID: 38036581 PMCID: PMC10689478 DOI: 10.1038/s41598-023-48391-z] [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: 07/21/2023] [Accepted: 11/26/2023] [Indexed: 12/02/2023] Open
Abstract
Mpox is a neglected zoonotic disease endemic in West and Central Africa. The Mpox outbreak with more than 90,000 cases worldwide since 2022 generated great concern about future outbreaks and highlighted the need for a simple and rapid diagnostic test. The Mpox virus, MPV, is a member of the Orthopoxvirus (OPV) genus that also contains other pathogenic viruses including variola virus, vaccinia virus, camelpox virus, and cowpox virus. Phylogenomic analysis of 200 OPV genomes identified 10 distinct phylogroups with the New World OPVs placed on a very long branch distant from the Old World OPVs. Isolates derived from infected humans were found to be distributed across multiple phylogroups interspersed with isolates from animal sources, indicating the zoonotic potential of these viruses. In this study, we developed a simple and sensitive colorimetric LAMP assay for generic detection of Old World OPVs. We also developed an MPV-specific probe that differentiates MPV from other OPVs in the N1R LAMP assay. In addition, we described an extraction-free protocol for use directly with swab eluates in LAMP assays, thereby eliminating the time and resources needed to extract DNA from the sample. Our direct LAMP assays are well-suited for low-resource settings and provide a valuable tool for rapid and scalable diagnosis and surveillance of OPVs and MPV.
Collapse
Affiliation(s)
- Zhiru Li
- Molecular Genetics and Genomics Division, New England Biolabs, Ipswich, MA, 01938, USA.
| | - Amit Sinha
- Molecular Genetics and Genomics Division, New England Biolabs, Ipswich, MA, 01938, USA
| | - Yinhua Zhang
- Molecular Genetics and Genomics Division, New England Biolabs, Ipswich, MA, 01938, USA
| | - Nathan Tanner
- Molecular Genetics and Genomics Division, New England Biolabs, Ipswich, MA, 01938, USA
| | | | | | - Clotilde K S Carlow
- Molecular Genetics and Genomics Division, New England Biolabs, Ipswich, MA, 01938, USA
| |
Collapse
|
23
|
Gao Z, He X, Chen G, Fang Y, Meng Z, Tian H, Zhang H, Jing Z. The Viral Protein Poly(A) Polymerase Catalytic Subunit Interacts with Guanylate-Binding Proteins 2 to Antagonize the Antiviral Ability of Targeting Ectromelia Virus. Int J Mol Sci 2023; 24:15750. [PMID: 37958732 PMCID: PMC10648259 DOI: 10.3390/ijms242115750] [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: 09/18/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The recent spread of the monkeypox virus among humans has heightened concerns regarding orthopoxvirus infections. Consequently, conducting a comprehensive study on the immunobiology of the monkeypox virus is imperative for the development of effective therapeutics. Ectromelia virus (ECTV) closely resembles the genetic and disease characteristics of monkeypox virus, making it a valuable research tool for studying orthopoxvirus-host interactions. Guanylate-binding proteins (GBPs), highly expressed interferon-stimulated genes (ISGs), have antagonistic effects against various intracellular pathogenic microorganisms. Our previous research has shown that GBP2 has a mild but statistically significant inhibitory effect on ECTV infection. The presence of a significant number of molecules in the poxvirus genome that encode the host immune response raises questions about whether it also includes proteins that counteract the antiviral activity of GBP2. Using IP/MS and co-IP technology, we discovered that the poly(A) polymerase catalytic subunit (PAPL) protein of ECTV is a viral regulatory molecule that interacts with GBP2. Further studies have shown that PAPL antagonizes the antiviral activity of GBP2 by reducing its protein levels. Knocking out the PAPL gene of ECTV with the CRISPR/Cas9 system significantly diminishes the replication ability of the virus, indicating the indispensable role of PAPL in the replication process of ECTV. In conclusion, our study presents preliminary evidence supporting the significance of PAPL as a virulence factor that can interact with GBP2.
Collapse
Affiliation(s)
- Zhenzhen Gao
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Xiaobing He
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Guohua Chen
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Yongxiang Fang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zejing Meng
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Huihui Tian
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Hui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zhizhong Jing
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
24
|
Borges V, Duque MP, Martins JV, Vasconcelos P, Ferreira R, Sobral D, Pelerito A, de Carvalho IL, Núncio MS, Borrego MJ, Roemer C, Neher RA, O'Driscoll M, Rocha R, Lopo S, Neves R, Palminha P, Coelho L, Nunes A, Isidro J, Pinto M, Santos JD, Mixão V, Santos D, Duarte S, Vieira L, Martins F, Machado J, Veríssimo VC, Grau B, Peralta-Santos A, Neves J, Caldeira M, Pestana M, Fernandes C, Caria J, Pinto R, Póvoas D, Maltez F, Sá AI, Salvador MB, Teófilo E, Rocha M, Moneti V, Duque LM, E Silva FF, Baptista T, Vasconcelos J, Casanova S, Mansinho K, Alves JV, Alves J, Silva A, Alpalhão M, Brazão C, Sousa D, Filipe P, Pacheco P, Peruzzu F, de Jesus RP, Ferreira L, Mendez J, Jordão S, Duarte F, Gonçalves MJ, Pena E, Silva CN, Guimarães AR, Tavares M, Freitas G, Cordeiro R, Gomes JP. Viral genetic clustering and transmission dynamics of the 2022 mpox outbreak in Portugal. Nat Med 2023; 29:2509-2517. [PMID: 37696933 PMCID: PMC10579057 DOI: 10.1038/s41591-023-02542-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023]
Abstract
Pathogen genome sequencing during epidemics enhances our ability to identify and understand suspected clusters and investigate their relationships. Here, we combine genomic and epidemiological data of the 2022 mpox outbreak to better understand early viral spread, diversification and transmission dynamics. By sequencing 52% of the confirmed cases in Portugal, we identified the mpox virus sublineages with the highest impact on case numbers and fitted them into a global context, finding evidence that several international sublineages probably emerged or spread early in Portugal. We estimated a 62% infection reporting rate and that 1.3% of the population of men who have sex with men in Portugal were infected. We infer the critical role played by sexual networks and superspreader gatherings, such as sauna attendance, in the dissemination of mpox virus. Overall, our findings highlight genomic epidemiology as a tool for the real-time monitoring and control of mpox epidemics, and can guide future vaccine policy in a highly susceptible population.
Collapse
Affiliation(s)
- Vítor Borges
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Mariana Perez Duque
- Epidemiology and Statistics Division, Directorate-General of Health, Lisbon, Portugal
- Pathogen Dynamics Group, Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - João Vieira Martins
- Epidemiology and Statistics Division, Directorate-General of Health, Lisbon, Portugal
| | - Paula Vasconcelos
- Public Health Emergency Centre, Directorate-General of Health, Lisbon, Portugal
| | - Rita Ferreira
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Daniel Sobral
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Ana Pelerito
- Emergency Response and Biopreparedness Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Isabel Lopes de Carvalho
- Emergency Response and Biopreparedness Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Maria Sofia Núncio
- Emergency Response and Biopreparedness Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Maria José Borrego
- National Reference Laboratory for Sexually Transmitted Infections, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Cornelius Roemer
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Richard A Neher
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Megan O'Driscoll
- Pathogen Dynamics Group, Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Raquel Rocha
- National Reference Laboratory for Sexually Transmitted Infections, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Sílvia Lopo
- National Reference Laboratory for Sexually Transmitted Infections, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Raquel Neves
- National Reference Laboratory for Sexually Transmitted Infections, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Paula Palminha
- National Reference Laboratory for Sexually Transmitted Infections, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Luís Coelho
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Alexandra Nunes
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal
| | - Joana Isidro
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Miguel Pinto
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - João Dourado Santos
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Verónica Mixão
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Daniela Santos
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Silvia Duarte
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Luís Vieira
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Fátima Martins
- Technical Board, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Jorge Machado
- Department Coordination, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Vítor Cabral Veríssimo
- Epidemiology and Statistics Division, Directorate-General of Health, Lisbon, Portugal
- Public Health Unit, ACES Cascais, ARSLVT, Cascais, Portugal
| | - Berta Grau
- Epidemiology and Statistics Division, Directorate-General of Health, Lisbon, Portugal
- Public Health Emergency Centre, Directorate-General of Health, Lisbon, Portugal
- ECDC Fellowship Programme, Field Epidemiology path (EPIET), European Centre for Disease Prevention and Control (ECDC), Solna, Sweden
- Directorate of Information and Analysis, Directorate-General of Health, Lisbon, Portugal
| | - André Peralta-Santos
- Directorate of Information and Analysis, Directorate-General of Health, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), Escola Nacional de Saúde Pública, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - José Neves
- Serviço de Dermatovenereologia, Consulta de DST, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Margarida Caldeira
- Serviço de Dermatovenereologia, Consulta de DST, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Mafalda Pestana
- Serviço de Dermatovenereologia, Consulta de DST, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Cândida Fernandes
- Serviço de Dermatovenereologia, Consulta de DST, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - João Caria
- Serviço de Doenças Infeciosas, Hospital de Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Raquel Pinto
- Serviço de Doenças Infeciosas, Hospital de Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Diana Póvoas
- Serviço de Doenças Infeciosas, Hospital de Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Fernando Maltez
- Serviço de Doenças Infeciosas, Hospital de Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Ana Isabel Sá
- Unidade de Doenças Sexualmente Transmissíveis da Lapa, Lisbon, Portugal
| | | | - Eugénio Teófilo
- GAT - Grupo de Ativistas em Tratamentos, Av. Paris, Lisbon, Portugal
| | - Miguel Rocha
- GAT - Grupo de Ativistas em Tratamentos, Av. Paris, Lisbon, Portugal
| | - Virginia Moneti
- GAT - Grupo de Ativistas em Tratamentos, Av. Paris, Lisbon, Portugal
| | - Luis Miguel Duque
- GAT - Grupo de Ativistas em Tratamentos, Av. Paris, Lisbon, Portugal
| | | | - Teresa Baptista
- GAT - Grupo de Ativistas em Tratamentos, Intendente, Lisbon, Portugal
| | - Joana Vasconcelos
- Serviço de Doenças Infeciosas e Medicina Tropical, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Sara Casanova
- Serviço de Doenças Infeciosas e Medicina Tropical, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Kamal Mansinho
- Serviço de Doenças Infeciosas e Medicina Tropical, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - João Vaz Alves
- Serviço de Doenças Infeciosas e Medicina Tropical, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - João Alves
- Serviço de Dermatovenereologia, Hospital Garcia de Orta, Almada, Portugal
| | - António Silva
- Serviço de Dermatovenereologia, Hospital Garcia de Orta, Almada, Portugal
| | - Miguel Alpalhão
- Dermatology Department, Centro Hospitalar Universitário Lisboa Norte EPE, Lisbon, Portugal
- Dermatology Research Unit (PFilipe Lab), Instituto de Medicina Molecular João Lobo Antunes, University of Lisbon, Lisbon, Portugal
- Dermatology University Clinic, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Cláudia Brazão
- Dermatology Department, Centro Hospitalar Universitário Lisboa Norte EPE, Lisbon, Portugal
| | - Diogo Sousa
- Dermatology Department, Centro Hospitalar Universitário Lisboa Norte EPE, Lisbon, Portugal
| | - Paulo Filipe
- Dermatology Department, Centro Hospitalar Universitário Lisboa Norte EPE, Lisbon, Portugal
- Dermatology Research Unit (PFilipe Lab), Instituto de Medicina Molecular João Lobo Antunes, University of Lisbon, Lisbon, Portugal
- Dermatology University Clinic, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Patrícia Pacheco
- Serviço de Infeciologia, Hospital Professor Doutor Fernando Fonseca, Amadora, Portugal
| | - Francesca Peruzzu
- Serviço de Infeciologia, Hospital Professor Doutor Fernando Fonseca, Amadora, Portugal
| | | | - Luís Ferreira
- Serviço Infeciologia do CHUP, Largo Professor Abel Salazar, Porto, Portugal
| | - Josefina Mendez
- Serviço Infeciologia do CHUP, Largo Professor Abel Salazar, Porto, Portugal
| | - Sofia Jordão
- Serviço de Doenças Infeciosas, Hospital Pedro Hispano - ULS Matosinhos, Matosinhos, Portugal
| | - Frederico Duarte
- Serviço de Doenças Infeciosas, Hospital Pedro Hispano - ULS Matosinhos, Matosinhos, Portugal
| | - Maria João Gonçalves
- Serviço de Doenças Infeciosas, Hospital Pedro Hispano - ULS Matosinhos, Matosinhos, Portugal
| | - Eduarda Pena
- Serviço de Doenças Infeciosas, Hospital Pedro Hispano - ULS Matosinhos, Matosinhos, Portugal
| | - Claúdio Nunes Silva
- Serviço de Doenças Infeciosas, Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | - Margarida Tavares
- Serviço de Doenças Infeciosas, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Graça Freitas
- National Health Authority, Directorate-General of Health, Lisbon, Portugal
| | - Rita Cordeiro
- Emergency Response and Biopreparedness Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - João Paulo Gomes
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal.
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal.
| |
Collapse
|
25
|
Li E, Guo X, Hong D, Gong Q, Xie W, Li T, Wang J, Chuai X, Chiu S. Duration of humoral immunity from smallpox vaccination and its cross-reaction with Mpox virus. Signal Transduct Target Ther 2023; 8:350. [PMID: 37709783 PMCID: PMC10502045 DOI: 10.1038/s41392-023-01574-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/11/2023] [Accepted: 07/26/2023] [Indexed: 09/16/2023] Open
Abstract
The ongoing pandemic caused by mpox virus (MPXV) has become an international public health emergency that poses a significant threat to global health. The vaccinia virus Tiantan strain (VTT) was used to vaccinate against smallpox in China 42 years ago. It is urgent to assess the level of immunity to smallpox in individuals vaccinated 43 or more years ago and evaluate their immunological susceptibility to MPXV. Here, we recruited 294 volunteers and detected the level of residual humoral immunity, including the vaccinia-specific IgG level and neutralizing antibody titer, and the cross-antibodies of MPXV A29L, B6R, A35R, and M1R. Our results showed that the humoral immunity from the smallpox vaccine in the population still remains, and VTT-specific NAb levels wane with age. The majority of the population pre-1981 who should be immunized with VTT still maintains certain levels of MPXV-specific antibodies, in particular, targeting A35R and B6R antigens. Furthermore, we separately analyzed the correlations between the OD450 values of VTT-specific IgG and A35R-specific IgG, B6R-specific IgG, and A29L-specific IgG with plasma samples diluted 1:40, showing a linear correlation (p < 0.0001). Our findings suggest that most Chinese populations still maintain VTT-specific IgG antibodies for 42 or more years after smallpox vaccination and could provide some level of protection against MPXV.
Collapse
Affiliation(s)
- Entao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoping Guo
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dongxiang Hong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qizan Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenyu Xie
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tingting Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jian Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Xia Chuai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
| | - Sandra Chiu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China.
| |
Collapse
|
26
|
Wei ZK, Zhao YC, Wang ZD, Sui LY, Zhao YH, Liu Q. Animal models of mpox virus infection and disease. INFECTIOUS MEDICINE 2023; 2:153-166. [PMID: 38073883 PMCID: PMC10699680 DOI: 10.1016/j.imj.2023.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 01/07/2025]
Abstract
Mpox (monkeypox) virus (MPXV), which causes a mild smallpox-like disease, has been endemic in Africa for several decades, with sporadic cases occurring in other parts of the world. However, the most recent outbreak of mpox mainly among men that have sex with men has affected several continents, posing serious global public health concerns. The infections exhibit a wide spectrum of clinical presentation, ranging from asymptomatic infection to mild, severe disease, especially in immunocompromised individuals, young children, and pregnant women. Some therapeutics and vaccines developed for smallpox have partial protective and therapeutic effects against MPXV historic isolates in animal models. However, the continued evolution of MPXV has produced multiple lineages, leading to significant gaps in the knowledge of their pathogenesis that constrain the development of targeted antiviral therapies and vaccines. MPXV infections in various animal models have provided a central platform for identification and comparison of diseased pathogenesis between the contemporary and historic isolates. In this review, we discuss the susceptibility of various animals to MPXV, and describe the key pathologic features of rodent, rabbit and nonhuman primate models. We also provide application examples of animal models in elucidating viral pathogenesis and evaluating effectiveness of vaccine and antiviral drugs. These animal models are essential to understand the biology of MPXV contemporary isolates and to rapidly test potential countermeasures. Finally, we list some remaining scientific questions of MPXV that can be resolved by animal models.
Collapse
Affiliation(s)
- Zheng-Kai Wei
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, China
| | - Yi-Cheng Zhao
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| | - Ze-Dong Wang
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| | - Li-Yan Sui
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| | - Ying-Hua Zhao
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| | - Quan Liu
- Department of Infectious Diseases, Center of Infectious diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, China
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| |
Collapse
|
27
|
Chiem K, Nogales A, Lorenzo M, Morales Vasquez D, Xiang Y, Gupta YK, Blasco R, de la Torre JC, Martínez-Sobrido L. Identification of In Vitro Inhibitors of Monkeypox Replication. Microbiol Spectr 2023; 11:e0474522. [PMID: 37278625 PMCID: PMC10434227 DOI: 10.1128/spectrum.04745-22] [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: 11/19/2022] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
Monkeypox virus (MPXV) infections in humans have historically been restricted to regions of endemicity in Africa. However, in 2022, an alarming number of MPXV cases were reported globally, with evidence of person-to-person transmission. Because of this, the World Health Organization (WHO) declared the MPXV outbreak a public health emergency of international concern. The supply of MPXV vaccines is limited, and only two antivirals, tecovirimat and brincidofovir, approved by the U.S. Food and Drug Administration (FDA) for the treatment of smallpox, are currently available for the treatment of MPXV infection. Here, we evaluated 19 compounds previously shown to inhibit different RNA viruses for their ability to inhibit orthopoxvirus infections. We first used recombinant vaccinia virus (rVACV) expressing fluorescence (mScarlet or green fluorescent protein [GFP]) and luciferase (Nluc) reporter genes to identify compounds with antiorthopoxvirus activity. Seven compounds from the ReFRAME library (antimycin A, mycophenolic acid, AVN-944, pyrazofurin, mycophenolate mofetil, azaribine, and brequinar) and six compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) showed inhibitory activity against rVACV. Notably, the anti-VACV activity of some of the compounds in the ReFRAME library (antimycin A, mycophenolic acid, AVN-944, mycophenolate mofetil, and brequinar) and all the compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) were confirmed with MPXV, demonstrating their inhibitory activity in vitro against two orthopoxviruses. IMPORTANCE Despite the eradication of smallpox, some orthopoxviruses remain important human pathogens, as exemplified by the recent 2022 monkeypox virus (MPXV) outbreak. Although smallpox vaccines are effective against MPXV, access to those vaccines is limited. In addition, current antiviral treatment against MPXV infections is limited to the use of the FDA-approved drugs tecovirimat and brincidofovir. Thus, there is an urgent need to identify novel antivirals for the treatment of MPXV infection and other potentially zoonotic orthopoxvirus infections. Here, we show that 13 compounds, derived from two different libraries, previously found to inhibit several RNA viruses, also inhibit VACV. Notably, 11 compounds also displayed inhibitory activity against MPXV.
Collapse
Affiliation(s)
- Kevin Chiem
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Aitor Nogales
- Animal Health Research Centre, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Maria Lorenzo
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | | | - Yan Xiang
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yogesh K. Gupta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
28
|
Zhou Y, Chen Z. Mpox: a review of laboratory detection techniques. Arch Virol 2023; 168:221. [PMID: 37543543 PMCID: PMC10404179 DOI: 10.1007/s00705-023-05848-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/04/2023] [Indexed: 08/07/2023]
Abstract
Mpox (formerly monkeypox) is a zoonotic disease caused by monkeypox virus (MPXV), which, like smallpox, is characterised by skin rashes. While the world is currently grappling with the coronavirus disease 2019 pandemic, the appearance of MPXV has presented a global threat and raised concerns worldwide. Since May 2022, MPXV has spread rapidly in non-endemic mpox areas. As of 27 June 2023, the virus has spread to more than 112 countries and regions, with over 88,060 laboratory-confirmed cases and 147 deaths. Thus, measures to control the mpox epidemic are urgently needed. As the principal methods for identifying and monitoring mpox, laboratory detection techniques play an important role in mpox diagnosis. This review summarises the currently-used laboratory techniques for MPXV detection, discusses progress in improving these methods, and compares the benefits and limitations of various diagnostic detection methods. Currently, nucleic acid amplification tests, such as the polymerase chain reaction, are the most commonly used. Immunological methods have also been applied to diagnose the disease, which can help us discover new features of MPXV, improve diagnostic accuracy, track epidemic trends, and guide future prevention and control strategies, which are also vital for controlling mpox epidemics. This review provides a resource for the scientific community and should stimulate more research and development in alternative diagnostics to be applied to this and future public health crises.
Collapse
Affiliation(s)
- Yunfan Zhou
- School of Medicine, Guangzhou Higher Education Mega Centre, South China University of Technology, Panyu District, Guangzhou, 510006, China.
| | - Zixin Chen
- School of Medicine, Guangzhou Higher Education Mega Centre, South China University of Technology, Panyu District, Guangzhou, 510006, China
| |
Collapse
|
29
|
Martínez-Fernández DE, Fernández-Quezada D, Casillas-Muñoz FAG, Carrillo-Ballesteros FJ, Ortega-Prieto AM, Jimenez-Guardeño JM, Regla-Nava JA. Human Monkeypox: A Comprehensive Overview of Epidemiology, Pathogenesis, Diagnosis, Treatment, and Prevention Strategies. Pathogens 2023; 12:947. [PMID: 37513794 PMCID: PMC10384102 DOI: 10.3390/pathogens12070947] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Monkeypox virus (MPXV) is an emerging zoonotic virus that belongs to the Orthopoxvirus genus and presents clinical symptoms similar to those of smallpox, such as fever and vesicular-pustular skin lesions. However, the differential diagnosis between smallpox and monkeypox is that smallpox does not cause lymphadenopathy but monkeypox generates swelling in the lymph nodes. Since the eradication of smallpox, MPXV has been identified as the most common Orthopoxvirus to cause human disease. Despite MPXV being endemic to certain regions of Africa, the current MPXV outbreak, which began in early 2022, has spread to numerous countries worldwide, raising global concern. As of the end of May 2023, over 87,545 cases and 141 deaths have been reported, with most cases identified in non-endemic countries, primarily due to human-to-human transmission. To better understand this emerging threat, this review presents an overview of key aspects of MPXV infection, including its animal reservoirs, modes of transmission, animal models, epidemiology, clinical and immunological features, diagnosis, treatments, vaccines, and prevention strategies. The material presented here provides a comprehensive understanding of MPXV as a disease, while emphasizing the significance and unique characteristics of the 2022 outbreak. This offers valuable information that can inform future research and aid in the development of effective interventions.
Collapse
Affiliation(s)
| | - David Fernández-Quezada
- Department of Neurosciences, University Center for Health Science (CUCS), University of Guadalajara, Guadalajara 44340, Mexico
| | | | | | - Ana Maria Ortega-Prieto
- Department of Microbiology, University of Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Málaga, Spain
| | - Jose M Jimenez-Guardeño
- Department of Microbiology, University of Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Málaga, Spain
| | - Jose Angel Regla-Nava
- Department of Microbiology and Pathology, University Center for Health Science (CUCS), University of Guadalajara, Guadalajara 44340, Mexico
| |
Collapse
|
30
|
Ushkalenko ND, Ersh AV, Filatov PV, Poltavchenko AG. [The rapid ELISA method for detection of orthopoxviruses]. Vopr Virusol 2023; 68:242-251. [PMID: 37436415 DOI: 10.36233/0507-4088-178] [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: 06/14/2023] [Indexed: 07/13/2023]
Abstract
INTRODUCTION Following the successful eradication of smallpox, mass vaccination against this disease was discontinued in 1980. The unvaccinated population continues to be at risk of infection due to military use of variola virus or exposure to monkeypox virus in Africa and non-endemic areas. In cases of these diseases, rapid diagnosis is of great importance, since the promptness and effectiveness of therapeutic and quarantine measures depend on it. The aim of work is to develop a kit of reagents for enzyme-linked immunosorbent assay (ELISA) for fast and highly sensitive detection of orthopoxviruses (OPV) in clinical samples. MATERIALS AND METHODS The efficiency of virus detection was evaluated by single-stage ELISA in the cryolisate of CV-1 cell culture samples infected with vaccinia, cowpox, rabbitpox, and ectromelia viruses, as well as in clinical samples of infected rabbits and mice. RESULTS The method of rapid ELISA was shown to allow the detection of OPV in crude viral samples in the range of 5.0 1025.0 103 PFU/ml, and in clinical samples with a viral load exceeding 5 103 PFU/ml. CONCLUSIONS The assay involves a minimum number of operations and can be performed within 45 minutes, which makes it possible to use it in conditions of a high level of biosecurity. Rapid ELISA method was developed using polyclonal antibodies, which significantly simplifies and reduces the cost of manufacturing a diagnostic system.
Collapse
Affiliation(s)
- N D Ushkalenko
- State Research Center of Virology and Biotechnology "Vector" of Rospotrebnadzor
| | - A V Ersh
- State Research Center of Virology and Biotechnology "Vector" of Rospotrebnadzor
| | - P V Filatov
- State Research Center of Virology and Biotechnology "Vector" of Rospotrebnadzor
| | - A G Poltavchenko
- State Research Center of Virology and Biotechnology "Vector" of Rospotrebnadzor
| |
Collapse
|
31
|
Srivastava S, Kumar S, Jain S, Mohanty A, Thapa N, Poudel P, Bhusal K, Al-Qaim ZH, Barboza JJ, Padhi BK, Sah R. The Global Monkeypox (Mpox) Outbreak: A Comprehensive Review. Vaccines (Basel) 2023; 11:1093. [PMID: 37376482 DOI: 10.3390/vaccines11061093] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 06/29/2023] Open
Abstract
Monkeypox (Mpox) is a contagious illness that is caused by the monkeypox virus, which is part of the same family of viruses as variola, vaccinia, and cowpox. It was first detected in the Democratic Republic of the Congo in 1970 and has since caused sporadic cases and outbreaks in a few countries in West and Central Africa. In July 2022, the World Health Organization (WHO) declared a public-health emergency of international concern due to the unprecedented global spread of the disease. Despite breakthroughs in medical treatments, vaccines, and diagnostics, diseases like monkeypox still cause death and suffering around the world and have a heavy economic impact. The 85,189 reported cases of Mpox as of 29 January 2023 have raised alarm bells. Vaccines for the vaccinia virus can protect against monkeypox, but these immunizations were stopped after smallpox was eradicated. There are, however, treatments available once the illness has taken hold. During the 2022 outbreak, most cases occurred among men who had sex with men, and there was a range of 7-10 days between exposure and the onset of symptoms. Three vaccines are currently used against the Monkeypox virus. Two of these vaccines were initially developed for smallpox, and the third is specifically designed for biological-terrorism protection. The first vaccine is an attenuated, nonreplicating smallpox vaccine that can also be used for immunocompromised individuals, marketed under different names in different regions. The second vaccine, ACAM2000, is a recombinant second-generation vaccine initially developed for smallpox. It is recommended for use in preventing monkeypox infection but is not recommended for individuals with certain health conditions or during pregnancy. The third vaccine, LC16m8, is a licensed attenuated smallpox vaccine designed to lack the B5R envelope-protein gene to reduce neurotoxicity. It generates neutralizing antibodies to multiple poxviruses and broad T-cell responses. The immune response takes 14 days after the second dose of the first two vaccines and 4 weeks after the ACAM2000 dose for maximal immunity development. The efficacy of these vaccines in the current outbreak of monkeypox is uncertain. Adverse events have been reported, and a next generation of safer and specific vaccines is needed. Although some experts claim that developing vaccines with a large spectrum of specificity can be advantageous, epitope-focused immunogens are often more effective in enhancing neutralization.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India
| | - Shagun Jain
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India
| | - Aroop Mohanty
- Department of Clinical Microbiology, All India Institute of Medical Sciences, Gorakhpur 273008, India
| | - Neeraj Thapa
- Nepal Medical College, Jorpati, Kathmandu 44600, Nepal
| | | | - Krishna Bhusal
- Lumbini Medical College, Tansen-11, Pravas, Palpa 32500, Nepal
| | - Zahraa Haleem Al-Qaim
- Department of Anesthesia Techniques, Al-Mustaqbal University College, Hilla 51001, Iraq
| | - Joshuan J Barboza
- Escuela de Medicina, Universidad César Vallejo, Trujillo 13007, Peru
| | - Bijaya Kumar Padhi
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, India
| |
Collapse
|
32
|
Luques MN, Oliveira RL, Hir S, Nunes DDS, Higa LM, Mendonça AF, Pereira LA, Sousa F, Castiñeiras TMPP, Tanuri A, Damaso CR. Co-circulation of vaccinia and monkeypox viruses in rural areas of Brazil: Importance of differential molecular diagnosis. Travel Med Infect Dis 2023; 53:102578. [PMID: 37088362 PMCID: PMC10122557 DOI: 10.1016/j.tmaid.2023.102578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Affiliation(s)
- Matheus Nobrega Luques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Régis Linhares Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Samuel Hir
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Desiree Dos Santos Nunes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luiza M Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Flávia Mendonça
- Laboratório Estadual de Saúde Pública Dr. Giovanni Cysneiros (LACEN/GO), Goiânia, GO, Brazil
| | - Luiz Augusto Pereira
- Laboratório Estadual de Saúde Pública Dr. Giovanni Cysneiros (LACEN/GO), Goiânia, GO, Brazil
| | - Fabrício Sousa
- Coordenação de Zoonoses, Secretaria Estadual de Saúde, Goiânia, GO, Brazil
| | - Terezinha Marta Pereira Pinto Castiñeiras
- Núcleo de Enfrentamento e Estudos de Doenças Infecciosas Emergentes e Reemergentes (NEEDIER), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Clarissa R Damaso
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
33
|
Chiem K, Nogales A, Lorenzo M, Vasquez DM, Xiang Y, Gupta YK, Blasco R, de la Torre JC, Mart Nez-Sobrido L. Antivirals against monkeypox infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537483. [PMID: 37131608 PMCID: PMC10153157 DOI: 10.1101/2023.04.19.537483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Monkeypox virus (MPXV) infection in humans are historically restricted to endemic regions in Africa. However, in 2022, an alarming number of MPXV cases have been reported globally with evidence of person-to-person transmission. Because of this, the World Health Organization (WHO) declared the MPXV outbreak a public health emergency of international concern. MPXV vaccines are limited and only two antivirals, tecovirimat and brincidofovir, approved by the United States (US) Food and Drug Administration (FDA) for the treatment of smallpox, are currently available for the treatment of MPXV infection. Here, we evaluated 19 compounds previously shown to inhibit different RNA viruses for their ability to inhibit Orthopoxvirus infections. We first used recombinant vaccinia virus (rVACV) expressing fluorescence (Scarlet or GFP) and luciferase (Nluc) reporter genes to identify compounds with anti-Orthopoxvirus activity. Seven compounds from the ReFRAME library (antimycin A, mycophenolic acid, AVN- 944, pyrazofurin, mycophenolate mofetil, azaribine, and brequinar) and six compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) showed antiviral activity against rVACV. Notably, the anti-VACV activity of some of the compounds in the ReFRAME library (antimycin A, mycophenolic acid, AVN- 944, mycophenolate mofetil, and brequinar) and all the compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) were confirmed with MPXV, demonstrating the broad-spectrum antiviral activity against Orthopoxviruses and their potential to be used for the antiviral treatment of MPXV, or other Orthopoxvirus, infections. IMPORTANCE Despite the eradication of smallpox, some Orthopoxviruses remain important human pathogens, as exemplified by the recent 2022 monkeypox virus (MPXV) outbreak. Although smallpox vaccines are effective against MPXV, there is presently limited access to those vaccines. In addition, current antiviral treatment against MPXV infections is limited to the use of the FDA-approved drugs tecovirimat and brincidofovir. Thus, there is an urgent need to identify novel antivirals for the treatment of MPXV, and other potentially zoonotic Orthopoxvirus infections. Here, we show that thirteen compounds, derived from two different libraries, previously found to inhibit several RNA viruses, exhibit also antiviral activity against VACV. Notably, eleven compounds also displayed antiviral activity against MPXV, demonstrating their potential to be incorporated into the therapeutic armamentarium to combat Orthopoxvirus infections.
Collapse
|
34
|
Kumar P, Chaudhary B, Yadav N, Devi S, Pareek A, Alla S, Kajal F, Nowrouzi-Kia B, Chattu VK, Gupta MM. Recent Advances in Research and Management of Human Monkeypox Virus: An Emerging Global Health Threat. Viruses 2023; 15:v15040937. [PMID: 37112916 PMCID: PMC10146223 DOI: 10.3390/v15040937] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
In 2003, the United States saw an epidemic of monkeypox that was later traced back to rodents of West Africa infected with the monkeypox virus (MPXV). Disease in the United States seemed less severe than the smallpox-like disease in the Democratic Republic of the Congo (DRC). In this study, researchers analyzed data from Central Africa: two distinct MPXV clades were confirmed by sequencing the genomes of MPXV isolates from Western Africa, the United States, and Central Africa. By comparing open reading frames across MPXV clades, scientists can infer which virus proteins might account for the observed variation in pathogenicity in humans. Monkeypox can be prevented and controlled with a better understanding of MPXV's molecular etiology and epidemiological and clinical features. In light of the current outbreaks worldwide, we provide updated information on monkeypox for medical professionals in this review.
Collapse
Affiliation(s)
- Parveen Kumar
- Shri Ram College of Pharmacy, Karnal 132116, Haryana, India
| | - Benu Chaudhary
- Guru Gobind Singh College of Pharmacy, Yamunanagar 135001, Haryana, India
| | - Nishant Yadav
- B.S. Anangpuria Institute of Pharmacy, Faridabad 121004, Haryana, India
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sujatha Alla
- Department of Engineering Management & Systems Engineering, Frank Batten College of Engineering, Old Dominion University, Norfolk, VA 23529, USA
- Center for Technology and Innovations, Global Health Research and Innovations Canada, Toronto, ON M1J 2W8, Canada
| | - Fnu Kajal
- Department of Health Promotion Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Behdin Nowrouzi-Kia
- Department of Occupational Science and Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
| | - Vijay Kumar Chattu
- Department of Occupational Science and Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Department of Community Medicine, Faculty of Medicine, Datta Meghe Institute of Medical Sciences, Wardha 442107, Maharashtra, India
- Center for Transdisciplinary Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Madan Mohan Gupta
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine 3303, Trinidad and Tobago
| |
Collapse
|
35
|
Van Reempts A, De Meester L, Blot K, Candaele AS, Beele H, Van Dorpe J, Huis In 't Veld D. A Belgian student with black eschars. Acta Clin Belg 2023; 78:180-184. [PMID: 35731612 DOI: 10.1080/17843286.2022.2090181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Human cowpox virus infection is a rare zoonotic disease. Cowpox virus is a member of the Orthopoxvirus genus, like smallpox. Over the last years records of cowpox virus transmission from pet cats and pet rats to humans in Europe have increased. This observation may result from the loss of cross-immunity against orthopoxviruses after discontinuation of routine smallpox vaccination in the 1980s. CASE PRESENTATION We report the first case of a human cowpox infection in an unvaccinated Belgian citizen. This 19-year-old student presented with multiple necrotic skin lesions on the chin, the scalp and the pubic region, and with cervical lymphadenopathy and flu-like symptoms. The diagnosis of human cowpox was based on electron microscopic findings and PCR examination performed on a skin biopsy of the pubic lesion. Close contact with cats (her domestic cats or cats from a local shelter) was probably the source of transmission. Spreading of the lesions was likely the result of autoinoculation. After six months all lesions spontaneously healed with atrophic scars. DISCUSSION To enhance awareness of this rare viral zoonosis and to verify the suspected increase in incidence and symptom severity after cessation of smallpox vaccination, one could argue whether human cowpox should become a notifiable disease.
Collapse
Affiliation(s)
- Astrid Van Reempts
- Department of Dermatology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Liesbet De Meester
- Department of General Internal Medicine and Infectious Diseases, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Koen Blot
- Department Epidemiology of Infectious Diseases, Sciensano, Ixelles, Belgium
| | - Ann-Sophie Candaele
- Department of Pathology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Hilde Beele
- Department of Dermatology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Diana Huis In 't Veld
- Department of General Internal Medicine and Infectious Diseases, Ghent University, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
36
|
Shabani S, Rashidi M, Radgoudarzi S, Jebali A. The validation of artificial anti-monkeypox antibodies by in silico and experimental approaches. Immun Inflamm Dis 2023; 11:e834. [PMID: 37102640 PMCID: PMC10091375 DOI: 10.1002/iid3.834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/06/2023] [Accepted: 03/25/2023] [Indexed: 04/28/2023] Open
Abstract
As a result of smallpox immunization programs that ended more than 40 years ago, a significant portion of the world's population is not immune. Moreover, due to the lack of anti-monkeypox drugs and vaccines against monkeypox, the spread of this virus may be the beginning of another challenge. In this study, novel antibodies against monkeypox virus were modeled based on a heavy chain of human antibody and a small peptide fragment. Docking of modeled antibodies with C19L protein showed the range of docking energy, and root-mean-square deviation (RMSD) was from -124 to -154 kcal/mL and 4-6 angstrom, respectively. Also, docking of modeled antibodies-C19L complex with gamma Fc receptor type I illustrated the range of docking energy, and RMSD was from -132 to -155 kcal/ml and 5-7 angstrom, respectively. Moreover, molecular dynamics simulation showed that antibody 62 had the highest stability with the lowest energy level and RMSD. Interestingly, no modeled antibodies had immunogenicity, allergenicity, and toxicity. Although all of them had good stability, only antibodies 25, 28, 54, and 62 had a half-life of >10 h. Moreover, the interaction between C19L protein and anti-C19L antibodies (wild-type and synthetic) was evaluated by the SPR method. We found that KD in synthetic antibodies was lower than wild antibody. In terms of δH°, TδS°, and δG°, the results were consistent with binding parameters. Here, the lowest value of thermodynamic parameters was obtained for antibody 62. These data show that the synthetic antibodies, especially antibody 62, had a higher affinity than the wild-type antibody.
Collapse
Affiliation(s)
- Sadeq Shabani
- Department of Biological SciencesFlorida International UniversityMiamiFloridaUSA
- Biomolecular Science InstituteFlorida International UniversityMiamiFloridaUSA
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of MedicineMazandaran University of Medical SciencesSariIran
- The Health of Plant and Livestock Products Research CenterMazandaran University of Medical SciencesSariIran
| | - Shakila Radgoudarzi
- I.M. Sechenov First Moscow State Medical University (Первый МГМУ им)MoscowRussia
| | - Ali Jebali
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical ScienceIslamic Azad UniversityTehranIran
| |
Collapse
|
37
|
Falendysz EA, Lopera JG, Rocke TE, Osorio JE. Monkeypox Virus in Animals: Current Knowledge of Viral Transmission and Pathogenesis in Wild Animal Reservoirs and Captive Animal Models. Viruses 2023; 15:905. [PMID: 37112885 PMCID: PMC10142277 DOI: 10.3390/v15040905] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Mpox, formerly called monkeypox, is now the most serious orthopoxvirus (OPXV) infection in humans. This zoonotic disease has been gradually re-emerging in humans with an increasing frequency of cases found in endemic areas, as well as an escalating frequency and size of epidemics outside of endemic areas in Africa. Currently, the largest known mpox epidemic is spreading throughout the world, with over 85,650 cases to date, mostly in Europe and North America. These increased endemic cases and epidemics are likely driven primarily by decreasing global immunity to OPXVs, along with other possible causes. The current unprecedented global outbreak of mpox has demonstrated higher numbers of human cases and greater human-to-human transmission than previously documented, necessitating an urgent need to better understand this disease in humans and animals. Monkeypox virus (MPXV) infections in animals, both naturally occurring and experimental, have provided critical information about the routes of transmission; the viral pathogenicity factors; the methods of control, such as vaccination and antivirals; the disease ecology in reservoir host species; and the conservation impacts on wildlife species. This review briefly described the epidemiology and transmission of MPXV between animals and humans and summarizes past studies on the ecology of MPXV in wild animals and experimental studies in captive animal models, with a focus on how animal infections have informed knowledge concerning various aspects of this pathogen. Knowledge gaps were highlighted in areas where future research, both in captive and free-ranging animals, could inform efforts to understand and control this disease in both humans and animals.
Collapse
Affiliation(s)
| | | | - Tonie E. Rocke
- U.S. Geological Survey, National Wildlife Health Center, Madison, WI 53711, USA
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
- Global Health Institute, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
38
|
Kandeel M. Current Clinical Trials for the Monkeypox Virus. DR. SULAIMAN AL HABIB MEDICAL JOURNAL 2023. [PMCID: PMC9990049 DOI: 10.1007/s44229-023-00029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Background Monkeypox (MPX) is a zoonotic Orthopoxvirus causing smallpox-like symptoms. Before April 2022, MPX cases outside Africa were rare. The virus can spread through skin-to-skin contact, sexual contact, respiratory droplets, and household items such as towels and blankets. Aim This study was aimed at highlighting the dire need for vaccination and treatment against this infection. Several in-process clinical trials that may help overcome MPX infection are discussed. Methods A search for recent clinical studies was conducted in the clinicaltrials.gov database. Results A total of 15 trials were identified. After February 2022, 14 new trials were launched. Of the 15 trials, 9 were observational studies, 3 were treatment studies and 3 were preventive studies. MPX clinical trial topics were classified into four broad categories: MPX virus shedding and clearance; response to MPX vaccine; antiviral treatment for MPX; and awareness regarding MPX. One medication, tecovirimat, and two vaccines are currently in clinical trials. Conclusions Few treatments and vaccines are under evaluation. Although multiple trials have been conducted, evidence to determine the present state of MPX infection is currently insufficient. Global collaboration is required to achieve complete understanding of the epidemiology, prevention and control of MPX.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, 31982 Al-Ahsa Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh, 33516 Egypt
| |
Collapse
|
39
|
Neuberger J, Brailsford SR, Mallinson G, Murphy MF, Simmonds P. Challenges for the maintaining the microbiological safety of the UK blood supply. Clin Med (Lond) 2023; 23:151-156. [PMID: 36806203 PMCID: PMC11046496 DOI: 10.7861/clinmed.2022-0401] [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: 02/22/2023]
Abstract
The supply of blood, blood products and components in the UK, as elsewhere, is safe, although there is no cause for complacency. Use of blood, blood products and components is not without risk of morbidity and mortality. Transfusion-transmitted infections (TTIs) continue to occur and may severely affect the health and welfare of recipients. As indicated by recent and current inquiries, public interest in these TTIs is huge. The risk of TTI can be mitigated but not abolished. Measures to reduce risk include screening of donors, testing of donations and, where appropriate, treatment of donations. The introduction of newer screening tests might identify some infectious donations but come at a cost, which could exceed a justifiable limit. Thus, the recognition, detection, reporting and investigation of cases of possible TTIs need to be improved. Recipients of blood should understand that, although transfusion in the UK is safe, it is not free of risk and so should be provided with full information so that properly informed consent can be given.
Collapse
Affiliation(s)
| | | | - Gary Mallinson
- Joint UK Blood Transfusion and Tissue Transplantation Services Professional Advisory Committee (JPAC), NHS Blood and Transplant, Bristol
| | - Michael F Murphy
- NHS Blood & Transplant, Oxford University Hospitals NHS Foundation Trust and University of Oxford, Oxford
| | | |
Collapse
|
40
|
Perveen N, Kundu B, Sudalaimuthuasari N, Al-Maskari RS, Muzaffar SB, Al-Deeb MA. Virome diversity of Hyalomma dromedarii ticks collected from camels in the United Arab Emirates. Vet World 2023; 16:439-448. [PMID: 37041826 PMCID: PMC10082741 DOI: 10.14202/vetworld.2023.439-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/30/2023] [Indexed: 03/18/2023] Open
Abstract
Background and Aim: Viruses are important components of the microbiome of ticks. Ticks are capable of transmitting several serious viral diseases to humans and animals. Hitherto, the composition of viral communities in Hyalomma dromedarii ticks associated with camels in the United Arab Emirates (UAE) remains unexplored. This study aimed to characterize the RNA virome diversity in male and female H. dromedarii ticks collected from camels in Al Ain, UAE.
Materials and Methods: We collected ticks, extracted, and sequenced RNA, using Illumina (NovaSeq 6000) and Oxford Nanopore (MinION).
Results: From the total generated sequencing reads, 180,559 (~0.35%) and 197,801 (~0.34%) reads were identified as virus-related reads in male and female tick samples, respectively. Taxonomic assignment of the viral sequencing reads was accomplished based on bioinformatic analyses. Further, viral reads were classified into 39 viral families. Poxiviridae, Phycodnaviridae, Phenuiviridae, Mimiviridae, and Polydnaviridae were the most abundant families in the tick viromes. Notably, we assembled the genomes of three RNA viruses, which were placed by phylogenetic analyses in clades that included the Bole tick virus.
Conclusion: Overall, this study attempts to elucidate the RNA virome of ticks associated with camels in the UAE and the results obtained from this study improve the knowledge of the diversity of viruses in H. dromedarii ticks.
Keywords: camels, Hyalomma dromedarii, nanopore technology, UAE, viral diversity, virome analysis, whole genome sequencing.
Collapse
Affiliation(s)
- Nighat Perveen
- Department of Biology, United Arab Emirates University, Al-Ain, P.O. Box 15551, UAE
| | - Biduth Kundu
- Department of Biology, United Arab Emirates University, Al-Ain, P.O. Box 15551, UAE
| | | | | | - Sabir Bin Muzaffar
- Department of Biology, United Arab Emirates University, Al-Ain, P.O. Box 15551, UAE
| | - Mohammad Ali Al-Deeb
- Department of Biology, United Arab Emirates University, Al-Ain, P.O. Box 15551, UAE
| |
Collapse
|
41
|
Orthopoxvirus Zoonoses—Do We Still Remember and Are Ready to Fight? Pathogens 2023; 12:pathogens12030363. [PMID: 36986285 PMCID: PMC10052541 DOI: 10.3390/pathogens12030363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
The eradication of smallpox was an enormous achievement due to the global vaccination program launched by World Health Organization. The cessation of the vaccination program led to steadily declining herd immunity against smallpox, causing a health emergency of global concern. The smallpox vaccines induced strong, humoral, and cell-mediated immune responses, protecting for decades after immunization, not only against smallpox but also against other zoonotic orthopoxviruses that now represent a significant threat to public health. Here we review the major aspects regarding orthopoxviruses’ zoonotic infections, factors responsible for viral transmissions, as well as the emerging problem of the increased number of monkeypox cases recently reported. The development of prophylactic measures against poxvirus infections, especially the current threat caused by the monkeypox virus, requires a profound understanding of poxvirus immunobiology. The utilization of animal and cell line models has provided good insight into host antiviral defenses as well as orthopoxvirus evasion mechanisms. To survive within a host, orthopoxviruses encode a large number of proteins that subvert inflammatory and immune pathways. The circumvention of viral evasion strategies and the enhancement of major host defenses are key in designing novel, safer vaccines, and should become the targets of antiviral therapies in treating poxvirus infections.
Collapse
|
42
|
Steinman L, Patarca R, Haseltine W. Experimental encephalomyelitis at age 90, still relevant and elucidating how viruses trigger disease. J Exp Med 2023; 220:213807. [PMID: 36652203 PMCID: PMC9880878 DOI: 10.1084/jem.20221322] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/28/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
20 yr ago, a tribute appeared in this journal on the 70th anniversary of an animal model of disseminated encephalomyelitis, abbreviated EAE for experimental autoimmune encephalomyelitis. "Observations on Attempts to Produce Disseminated Encephalomyelitis in Monkeys" appeared in the Journal of Experimental Medicine on February 21, 1933. Rivers and colleagues were trying to understand what caused neurological reactions to viral infections like smallpox, vaccinia, and measles, and what triggered rare instances of encephalomyelitis to smallpox vaccines. The animal model known as EAE continues to display its remarkable utility. Recent research, since the 70th-anniversary tribute, helps explain how Epstein-Barr virus triggers multiple sclerosis via molecular mimicry to a protein known as GlialCAM. Proteins with multiple domains similar to GlialCAM, tenascin, neuregulin, contactin, and protease kinase C inhibitors are present in the poxvirus family. These observations take us a full circle back to Rivers' first paper on EAE, 90 yr ago.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences and Pediatrics, Stanford University, Stanford, CA, USA,Correspondence to Lawrence Steinman:
| | | | | |
Collapse
|
43
|
Lozano C, Grenga L, Gallais F, Miotello G, Bellanger L, Armengaud J. Mass spectrometry detection of monkeypox virus: Comprehensive coverage for ranking the most responsive peptide markers. Proteomics 2023; 23:e2200253. [PMID: 35969374 DOI: 10.1002/pmic.202200253] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/21/2022] [Accepted: 08/11/2022] [Indexed: 01/20/2023]
Abstract
The recent and sudden outbreak of monkeypox in numerous non-endemic countries requires expanding its surveillance immediately and understanding its origin and spread. As learned from the COVID-19 pandemic, appropriate detection techniques are crucial to achieving such a goal. Mass spectrometry has the advantages of a rapid response, low analytical interferences, better precision, and easier multiplexing to detect various pathogens and their variants. In this proteomic dataset, we report experimental data on the proteome of the monkeypox virus (MPXV) recorded by state-of-the-art shotgun proteomics, including data-dependent and data-independent acquisition for comprehensive coverage. We highlighted 152 viral proteins, corresponding to an overall proteome coverage of 79.5 %. Among the 1371 viral peptides detected, 35 peptides with the most intense signals in mass spectrometry were selected, representing a subset of 13 viral proteins. Their relevance as potential candidate markers for virus detection by targeted mass spectrometry is discussed. This report should assist the rapid development of mass spectrometry-based tests to detect a pathogen of increasing concern.
Collapse
Affiliation(s)
- Clément Lozano
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | - Lucia Grenga
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | - Fabrice Gallais
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | - Guylaine Miotello
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | - Laurent Bellanger
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| |
Collapse
|
44
|
Shchelkunova GA, Shchelkunov SN. Smallpox, Monkeypox and Other Human Orthopoxvirus Infections. Viruses 2022; 15:103. [PMID: 36680142 PMCID: PMC9865299 DOI: 10.3390/v15010103] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Considering that vaccination against smallpox with live vaccinia virus led to serious adverse effects in some cases, the WHO, after declaration of the global eradication of smallpox in 1980, strongly recommended to discontinue the vaccination in all countries. This led to the loss of immunity against not only smallpox but also other zoonotic orthopoxvirus infections in humans over the past years. An increasing number of human infections with zoonotic orthopoxviruses and, first of all, monkeypox, force us to reconsider a possible re-emergence of smallpox or a similar disease as a result of natural evolution of these viruses. The review contains a brief analysis of the results of studies on genomic organization and evolution of human pathogenic orthopoxviruses, development of modern methods for diagnosis, vaccination, and chemotherapy of smallpox, monkeypox, and other zoonotic human orthopoxvirus infections.
Collapse
Affiliation(s)
| | - Sergei N. Shchelkunov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, 630559 Novosibirsk, Russia
| |
Collapse
|
45
|
Muacevic A, Adler JR, AlAnazi MM, Ayyashi MJ, Khubrani AA, Khormi YB, Shbeir LA, Alatif SI, Alfagih AE. The Global Human Monkeypox Outbreak and Management: A Comprehensive Literature Review. Cureus 2022; 14:e32557. [PMID: 36654643 PMCID: PMC9840451 DOI: 10.7759/cureus.32557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Monkeypox (MPX) belongs to the genus Orthopoxvirus (OPV), family Poxviridae, and sub-family Chordopoxvirinae. Human monkeypox (HMPX) is a viral zoonotic illness caused by the monkeypox virus (MPXV). Several non-endemic countries have confirmed MPX cases across the globe. Therefore, consider an outbreak to be a global health emergency. MPXV transmits from animals to humans via infected animals, and there is currently human-to-human transmission, notably among guys who have sexual relations with males. Healthcare interventions are required to stop outbreaks. These include strict isolation and care for MPX patients while they are still contagious or until the skin lesions dry out and crust over. JYNNEOS was approved as a vaccine for the prevention of MPXV. Tecovirimat is licensed to treat severe MPX or risk developing a serious disease. We should encourage international cooperation to conduct clinical trials investigating the effectiveness and safety of MPXV vaccines and antiviral medications. Precautions must be taken at the global level to prevent an MPXV outbreak.
Collapse
|
46
|
Evaluation of Rapid Dot-Immunoassay for Detection Orthopoxviruses Using Laboratory-Grown Viruses and Animal's Clinical Specimens. Viruses 2022; 14:v14112580. [PMID: 36423189 PMCID: PMC9697496 DOI: 10.3390/v14112580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The aim of the work was an experimental evaluation of the characteristics of the kit for the rapid immunochemical detection of orthopoxviruses (OPV). The kit is based on the method of one-stage dot-immunoassay on flat protein arrays using gold conjugates and a silver developer. Rabbit polyclonal antibodies against the vaccinia virus were used as capture and detection reagents. The sensitivity of detection of OPV and the specificity of the analysis were assessed using culture crude preparations (monkeypox virus, vaccinia virus, rabbitpox virus, cowpox virus, and ectromelia virus), a suspension from a crust from a human vaccination site as well as blood and tissue suspensions of infected rabbits. It has been shown that the assay using the kit makes it possible to detect OPV within 36 min at a temperature of 18-40 °C in unpurified culture samples of the virus and clinical samples in the range of 103-104 PFU/mL. Tests of the kit did not reveal cross-reactivity with uninfected cell cultures and viral pathogens of exanthematous infections (measles, rubella and chicken pox). The kit can be used to detect or exclude the presence of a virus threat in samples and can be useful in various aspects of biosecurity. The simplicity of analysis, the possibility of visual accounting the and interpretation of the results make it possible to use the test in laboratories with a high level of biological protection and in out-of-laboratory conditions.
Collapse
|
47
|
White J, Rivero MJ, Mohamed AI, Thomas J, Muthigi A, Rahman F, Ory J, Petrella F, Ramasamy R. Male Sexual Health Implications of the 2022 Global Monkeypox Outbreak. Res Rep Urol 2022; 14:415-421. [PMID: 36438435 PMCID: PMC9698321 DOI: 10.2147/rru.s381191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/16/2022] [Indexed: 05/12/2025] Open
Abstract
The 2022 global monkeypox (MPX) outbreak is the largest in history to occur outside of endemic African regions. Disease spread during this outbreak has been primarily through human-to-human transmission, with sexual contact being of particular concern. Clinical presentations have commonly featured genital, perianal, and oral lesions associated with sexual activity among men who have sex with men (MSM), who compose the vast majority of MPX cases. This review discusses the epidemiology, clinical features, and evaluation of MPX with regards to men's sexual health. Comparisons were made between MPX and its relative from the Orthopoxvirus genus, smallpox, in order to make informed inferences on the potential effects of MPX on men's sexual health. This review also discusses the role of men's health specialists and urologists in addressing the current outbreak.
Collapse
Affiliation(s)
- Joshua White
- Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Marco-Jose Rivero
- Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Arslan I Mohamed
- Undergraduate Medical Education, City University of New York, New York, NY, USA
| | - Jamie Thomas
- Undergraduate Medical Education, Nova Southeastern University Kiran Patel College of Osteopathic Medicine, Fort Lauderdale, FL, USA
| | - Akhil Muthigi
- Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Farah Rahman
- Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jesse Ory
- Urology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Ranjith Ramasamy
- Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
48
|
Huang Y, Mu L, Wang W. Monkeypox: epidemiology, pathogenesis, treatment and prevention. Signal Transduct Target Ther 2022; 7:373. [PMID: 36319633 PMCID: PMC9626568 DOI: 10.1038/s41392-022-01215-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022] Open
Abstract
Monkeypox is a zoonotic disease that was once endemic in west and central Africa caused by monkeypox virus. However, cases recently have been confirmed in many nonendemic countries outside of Africa. WHO declared the ongoing monkeypox outbreak to be a public health emergency of international concern on July 23, 2022, in the context of the COVID-19 pandemic. The rapidly increasing number of confirmed cases could pose a threat to the international community. Here, we review the epidemiology of monkeypox, monkeypox virus reservoirs, novel transmission patterns, mutations and mechanisms of viral infection, clinical characteristics, laboratory diagnosis and treatment measures. In addition, strategies for the prevention, such as vaccination of smallpox vaccine, is also included. Current epidemiological data indicate that high frequency of human-to-human transmission could lead to further outbreaks, especially among men who have sex with men. The development of antiviral drugs and vaccines against monkeypox virus is urgently needed, despite some therapeutic effects of currently used drugs in the clinic. We provide useful information to improve the understanding of monkeypox virus and give guidance for the government and relative agency to prevent and control the further spread of monkeypox virus.
Collapse
Affiliation(s)
- Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Mu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
49
|
RACK1 Regulates Poxvirus Protein Synthesis Independently of Its Role in Ribosome-Based Stress Signaling. J Virol 2022; 96:e0109322. [PMID: 36098514 PMCID: PMC9517738 DOI: 10.1128/jvi.01093-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Receptor for activated C kinase 1 (RACK1) is a small ribosomal subunit protein that is phosphorylated by vaccinia virus (VacV) to maximize translation of postreplicative (PR) mRNAs that harbor 5' polyA leaders. However, RACK1 is a multifunctional protein that both controls translation directly and acts as a scaffold for signaling to and from the ribosome. This includes stress signaling that is activated by ribosome-associated quality control (RQC) and ribotoxic stress response (RSR) pathways. As VacV infection activates RQC and stress signaling, whether RACK1 influences viral protein synthesis through its effects on translation, signaling, or both remains unclear. Examining the effects of genetic knockout of RACK1 on the phosphorylation of key mitogenic and stress-related kinases, we reveal that loss of RACK1 specifically blunts the activation of c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) at late stages of infection. However, RACK1 was not required for JNK recruitment to ribosomes, and unlike RACK1 knockout, JNK inhibitors had no effect on viral protein synthesis. Moreover, reduced JNK activity during infection in RACK1 knockout cells contrasted with the absolute requirement for RACK1 in RSR-induced JNK phosphorylation. Comparing the effects of RACK1 knockout alongside inhibitors of late stage replication, our data suggest that JNK activation is only indirectly affected by the absence of RACK1 due to reduced viral protein accumulation. Cumulatively, our findings in the context of infection add further support for a model whereby RACK1 plays a specific and direct role in controlling translation of PR viral mRNAs that is independent of its role in ribosome-based stress signaling. IMPORTANCE Receptor for activated C kinase 1 (RACK1) is a multifunctional ribosomal protein that regulates translation directly and mediates signaling to and from the ribosome. While recent work has shown that RACK1 is phosphorylated by vaccinia virus (VacV) to stimulate translation of postreplicative viral mRNAs, whether RACK1 also contributes to VacV replication through its roles in ribosome-based stress signaling remains unclear. Here, we characterize the role of RACK1 in infected cells. In doing so, we find that RACK1 is essential for stress signal activation by ribotoxic stress responses but not by VacV infection. Moreover, although the loss of RACK1 reduces the level of stress-associated JNK activation in infected cells, this is an indirect consequence of RACK1's specific requirement for the synthesis of postreplicative viral proteins, the accumulation of which determines the level of cellular stress. Our findings reveal both the specific role of RACK1 and the complex downstream effects of its control of viral protein synthesis in the context of infection.
Collapse
|
50
|
Iyer RF, Edwards DM, Kolb P, Raué HP, Nelson CA, Epperson ML, Slifka MK, Nolz JC, Hengel H, Fremont DH, Früh K. The secreted protein Cowpox Virus 14 contributes to viral virulence and immune evasion by engaging Fc-gamma-receptors. PLoS Pathog 2022; 18:e1010783. [PMID: 36121874 PMCID: PMC9521928 DOI: 10.1371/journal.ppat.1010783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 09/29/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
The genome of cowpoxvirus (CPXV) could be considered prototypical for orthopoxviridae (OXPV) since it contains many open reading frames (ORFs) absent or lost in other OPXV, including vaccinia virus (VACV). These additional ORFs are non-essential for growth in vitro but are expected to contribute to the broad host range, virulence and immune evasion characteristics of CPXV. For instance, unlike VACV, CPXV encodes proteins that interfere with T cell stimulation, either directly or by preventing antigen presentation or co-stimulation. When studying the priming of naïve T cells, we discovered that CPXV, but not VACV, encodes a secreted factor that interferes with activation and proliferation of naïve CD8+ and CD4+ T cells, respectively, in response to anti-CD3 antibodies, but not to other stimuli. Deletion mapping revealed that the inhibitory protein is encoded by CPXV14, a small secreted glycoprotein belonging to the poxvirus immune evasion (PIE) family and containing a smallpoxvirus encoded chemokine receptor (SECRET) domain that mediates binding to chemokines. We demonstrate that CPXV14 inhibition of antibody-mediated T cell activation depends on the presence of Fc-gamma receptors (FcγRs) on bystander cells. In vitro, CPXV14 inhibits FcγR-activation by antigen/antibody complexes by binding to FcγRs with high affinity and immobilized CPXV14 can trigger signaling through FcγRs, particularly the inhibitory FcγRIIB. In vivo, CPXV14-deleted virus showed reduced viremia and virulence resulting in reduced weight loss and death compared to wildtype virus whereas both antibody and CD8+ T cell responses were increased in the absence of CPXV14. Furthermore, no impact of CPXV14-deletion on virulence was observed in mice lacking the inhibitory FcγRIIB. Taken together our results suggest that CPXV14 contributes to virulence and immune evasion by binding to host FcγRs.
Collapse
Affiliation(s)
- Ravi F. Iyer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - David M. Edwards
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Peter Raué
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Chris A. Nelson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Megan L. Epperson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| |
Collapse
|