1
|
Baker CN, Duso D, Kothapalli N, Hart T, Casey S, Cookenham T, Kummer L, Hvizdos J, Lanzer K, Vats P, Shanbhag P, Bell I, Tighe M, Travis K, Szaba F, Harder JM, Bedard O, Oberding N, Ward JM, Adams MD, Lutz C, Bradrick SS, Reiley WW, Rosenthal NA. Characterization of Collaborative Cross mouse founder strain CAST/EiJ as a novel model for lethal COVID-19. Sci Rep 2024; 14:25147. [PMID: 39448712 PMCID: PMC11502910 DOI: 10.1038/s41598-024-77087-1] [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: 07/02/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Mutations in SARS-CoV-2 variants of concern (VOCs) have expanded the viral host range beyond primates, and a few other mammals, to mice, affording the opportunity to exploit genetically diverse mouse panels to model the broad spectrum of responses to infection in patient populations. Here we surveyed responses to VOC infection in genetically diverse Collaborative Cross (CC) founder strains. Infection of wild-derived CC founder strains produced a broad range of viral burden, disease susceptibility and survival, whereas most other strains were resistant to disease despite measurable lung viral titers. In particular, CAST/EiJ, a wild-derived strain, developed high lung viral burdens, more severe lung pathology than seen in other CC strains, and a dysregulated cytokine profile resulting in morbidity and mortality. These inbred mouse strains may serve as a valuable platform to evaluate therapeutic countermeasures against severe COVID-19 and other coronavirus pandemics in the future.
Collapse
Affiliation(s)
| | - Debra Duso
- Trudeau Institute, Saranac Lake, NY, USA
| | | | | | - Sean Casey
- Trudeau Institute, Saranac Lake, NY, USA
| | | | | | | | | | - Purva Vats
- The Jackson Laboratory, Farmington, CT, USA
| | | | - Isaac Bell
- The Jackson Laboratory, Farmington, CT, USA
| | - Mike Tighe
- Trudeau Institute, Saranac Lake, NY, USA
| | | | | | | | | | | | | | | | | | | | | | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
2
|
Fang D, Liu Y, Dou D, Su B. The unique immune evasion mechanisms of the mpox virus and their implication for developing new vaccines and immunotherapies. Virol Sin 2024:S1995-820X(24)00135-4. [PMID: 39181538 DOI: 10.1016/j.virs.2024.08.008] [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: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Mpox is an infectious and contagious zoonotic disease caused by the mpox virus (MPXV), which belongs to the genus Orthopoxvirus. Since 2022, MPXV has posed a significant threat to global public health. The emergence of thousands of cases across the Western Hemisphere prompted the World Health Organization to declare an emergency. The extensive coevolutionary history of poxviruses with humans has enabled these viruses to develop sophisticated mechanisms to counter the human immune system. Specifically, MPXV employs unique immune evasion strategies against a wide range of immunological elements, presenting a considerable challenge for treatment, especially following the discontinuation of routine smallpox vaccination among the general population. In this review, we start by discussing the entry of the mpox virus and the onset of early infection, followed by an introduction to the mechanisms by which the mpox virus can evade the innate and adaptive immune responses. Two caspase-1 inhibitory proteins and a PKR escape-related protein have been identified as phylogenomic hubs involved in modulating the immune environment during the MPXV infection. With respect to adaptive immunity, mpox viruses exhibit unique and exceptional T-cell inhibition capabilities, thereby comprehensively remodeling the host immune environment. The viral envelope also poses challenges for the neutralizing effects of antibodies and the complement system. The unique immune evasion mechanisms employed by MPXV make novel multi-epitope and nucleic acid-based vaccines highly promising research directions worth investigating. Finally, we briefly discuss the impact of MPXV infection on immunosuppressed patients and the current status of MPXV vaccine development. This review may provide valuable information for the development of new immunological treatments for mpox.
Collapse
Affiliation(s)
- Dong Fang
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China
| | - Yan Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Dou Dou
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China
| | - Bin Su
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Central Laboratory, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
3
|
Baker CN, Duso D, Kothapalli N, Hart T, Casey S, Cookenham T, Kummer L, Hvizdos J, Lanzer K, Vats P, Shanbhag P, Bell I, Tighe M, Travis K, Szaba F, Bedard O, Oberding N, Ward JM, Adams MD, Lutz C, Bradrick SS, Reiley WW, Rosenthal N. Characterization of Collaborative Cross mouse founder strain CAST/EiJ as a novel model for lethal COVID-19. RESEARCH SQUARE 2024:rs.3.rs-4675061. [PMID: 39149485 PMCID: PMC11326417 DOI: 10.21203/rs.3.rs-4675061/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Mutations in SARS-CoV-2 variants of concern (VOCs) have expanded the viral host range beyond primates, and a limited range of other mammals, to mice, affording the opportunity to exploit genetically diverse mouse panels to model the broad range of responses to infection in patient populations. Here we surveyed responses to VOC infection in genetically diverse Collaborative Cross (CC) founder strains. Infection of wild-derived CC founder strains produced a broad range of viral burden, disease susceptibility and survival, whereas most other strains were resistant to disease despite measurable lung viral titers. In particular, CAST/EiJ, a wild-derived strain, developed high lung viral burdens, more severe lung pathology than seen in other CC strains, and a dysregulated cytokine profile resulting in morbidity and mortality. These inbred mouse strains may serve as a valuable platform to evaluate therapeutic countermeasures against severe COVID-19 and other coronavirus pandemics in the future.
Collapse
|
4
|
Li J, Li X, Dong J, Wei J, Guo X, Wang G, Xu M, Zhao A. Enhanced Immune Responses in Mice by Combining the Mpox Virus B6R-Protein and Aluminum Hydroxide-CpG Vaccine Adjuvants. Vaccines (Basel) 2024; 12:776. [PMID: 39066415 PMCID: PMC11281346 DOI: 10.3390/vaccines12070776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Novel adjuvants and innovative combinations of adjuvants (Adjuvant Systems) have facilitated the development of enhanced and new vaccines against re-emerging and challenging pathogenic microorganisms. Nonetheless, the efficacy of adjuvants is influenced by various factors, and the same adjuvant may generate entirely different immune responses when paired with different antigens. Herein, we combined the MPXV-B6R antigen with BC02, a novel adjuvant with proprietary technology, to assess its capability to induce both cellular and humoral immunity in mouse models. Mice received two intramuscular injections of B6R-BC02, which resulted in the production of MPXV-specific IgG, IgG1, and IgG2a antibodies. Additionally, it elicited strong MPXV-specific Th1-oriented cellular immunity and persistent effector memory B-cell responses. The advantages of BC02 were further validated, including rapid initiation of the immune response, robust recall memory, and sustained immune response induction. Although the potential of immunized mice to produce serum-neutralizing antibodies against the vaccinia virus requires further improvement, the exceptional performance of BC02 as an adjuvant for the MPXV-B6R antigen has been consistently demonstrated.
Collapse
Affiliation(s)
- Junli Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Xiaochi Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Jiaxin Dong
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Jiazheng Wei
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- College of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang 117004, China
| | - Xiaonan Guo
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Guozhi Wang
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Miao Xu
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| | - Aihua Zhao
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing 102629, China; (J.L.); (X.L.); (J.D.); (J.W.); (X.G.); (G.W.); (M.X.)
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing 102629, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing 102629, China
| |
Collapse
|
5
|
da Silva GB, de Carvalho Braga G, Simões JLB, Kempka AP, Bagatini MD. Cytokine storm in human monkeypox: A possible involvement of purinergic signaling. Cytokine 2024; 177:156560. [PMID: 38447385 DOI: 10.1016/j.cyto.2024.156560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
Some evidence has indicated that monkeypox can induce a cytokine storm. Purinergic signaling is a cell pathway related to the cytokine storm. However, the precise mechanisms that lead to cytokine storms in monkeypox infections and the possible involvement of purinergic signaling in the immune response to this virus remain unknown. In this review article, we aimed to highlight a body of scientific evidence that consolidates the role of the cytokine storm in monkeypox infection and proposes a new hypothesis regarding the roles of purinergic signaling in this immune-mediated mechanism. We further suggested some purinergic signaling modulators to mitigate the deleterious and aggravating effects of immune dysregulation in human monkeypox virus infection by inhibiting P2X3, P2X7, P2Y2, and P2Y12, reducing inflammation, and activating A1 and A2A receptors to promote an anti-inflammatory response.
Collapse
Affiliation(s)
- Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil.
| | | | | | - Aniela Pinto Kempka
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Margarete Dulce Bagatini
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil; Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
6
|
Tamir H, Noy-Porat T, Melamed S, Cherry-Mimran L, Barlev-Gross M, Alcalay R, Yahalom-Ronen Y, Achdout H, Politi B, Erez N, Weiss S, Rosenfeld R, Epstein E, Mazor O, Makdasi E, Paran N, Israely T. Synergistic effect of two human-like monoclonal antibodies confers protection against orthopoxvirus infection. Nat Commun 2024; 15:3265. [PMID: 38627363 PMCID: PMC11021552 DOI: 10.1038/s41467-024-47328-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
The eradication of smallpox was officially declared by the WHO in 1980, leading to discontinuation of the vaccination campaign against the virus. Consequently, immunity against smallpox and related orthopoxviruses like Monkeypox virus gradually declines, highlighting the need for efficient countermeasures not only for the prevention, but also for the treatment of already exposed individuals. We have recently developed human-like monoclonal antibodies (mAbs) from vaccinia virus-immunized non-human primates. Two mAbs, MV33 and EV42, targeting the two infectious forms of the virus, were selected for in vivo evaluation, based on their in vitro neutralization potency. A single dose of either MV33 or EV42 administered three days post-infection (dpi) to BALB/c female mice provides full protection against lethal ectromelia virus challenge. Importantly, a combination of both mAbs confers full protection even when provided five dpi. Whole-body bioimaging and viral load analysis reveal that combination of the two mAbs allows for faster and more efficient clearance of the virus from target organs compared to either MV33 or EV42 separately. The combined mAbs treatment further confers post-exposure protection against the currently circulating Monkeypox virus in Cast/EiJ female mice, highlighting their therapeutic potential against other orthopoxviruses.
Collapse
Affiliation(s)
- Hadas Tamir
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tal Noy-Porat
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Israel Institute for Biological Research, Ness Ziona, Israel
| | | | | | - Ron Alcalay
- Israel Institute for Biological Research, Ness Ziona, Israel
| | | | - Hagit Achdout
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Noam Erez
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shay Weiss
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ronit Rosenfeld
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Eyal Epstein
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ohad Mazor
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Efi Makdasi
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Nir Paran
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tomer Israely
- Israel Institute for Biological Research, Ness Ziona, Israel.
| |
Collapse
|
7
|
Zuiani A, Dulberger CL, De Silva NS, Marquette M, Lu YJ, Palowitch GM, Dokic A, Sanchez-Velazquez R, Schlatterer K, Sarkar S, Kar S, Chawla B, Galeev A, Lindemann C, Rothenberg DA, Diao H, Walls AC, Addona TA, Mensa F, Vogel AB, Stuart LM, van der Most R, Srouji JR, Türeci Ö, Gaynor RB, Şahin U, Poran A. A multivalent mRNA monkeypox virus vaccine (BNT166) protects mice and macaques from orthopoxvirus disease. Cell 2024; 187:1363-1373.e12. [PMID: 38366591 DOI: 10.1016/j.cell.2024.01.017] [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: 10/04/2023] [Revised: 11/13/2023] [Accepted: 01/12/2024] [Indexed: 02/18/2024]
Abstract
In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Özlem Türeci
- BioNTech SE, Mainz, Germany; HI-TRON - Helmholtz Institute for Translational Oncology Mainz by DKFZ, Mainz, Germany
| | | | - Uğur Şahin
- BioNTech SE, Mainz, Germany; TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | | |
Collapse
|
8
|
Alakunle E, Kolawole D, Diaz-Cánova D, Alele F, Adegboye O, Moens U, Okeke MI. A comprehensive review of monkeypox virus and mpox characteristics. Front Cell Infect Microbiol 2024; 14:1360586. [PMID: 38510963 PMCID: PMC10952103 DOI: 10.3389/fcimb.2024.1360586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Monkeypox virus (MPXV) is the etiological agent of monkeypox (mpox), a zoonotic disease. MPXV is endemic in the forested regions of West and Central Africa, but the virus has recently spread globally, causing outbreaks in multiple non-endemic countries. In this paper, we review the characteristics of the virus, including its ecology, genomics, infection biology, and evolution. We estimate by phylogenomic molecular clock that the B.1 lineage responsible for the 2022 mpox outbreaks has been in circulation since 2016. We interrogate the host-virus interactions that modulate the virus infection biology, signal transduction, pathogenesis, and host immune responses. We highlight the changing pathophysiology and epidemiology of MPXV and summarize recent advances in the prevention and treatment of mpox. In addition, this review identifies knowledge gaps with respect to the virus and the disease, suggests future research directions to address the knowledge gaps, and proposes a One Health approach as an effective strategy to prevent current and future epidemics of mpox.
Collapse
Affiliation(s)
- Emmanuel Alakunle
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| | - Daniel Kolawole
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| | - Diana Diaz-Cánova
- Department of Medical Biology, UIT – The Arctic University of Norway, Tromsø, Norway
| | - Faith Alele
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | - Oyelola Adegboye
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Ugo Moens
- Department of Medical Biology, UIT – The Arctic University of Norway, Tromsø, Norway
| | - Malachy Ifeanyi Okeke
- Department of Natural and Environmental Sciences, American University of Nigeria, Yola, Nigeria
| |
Collapse
|
9
|
Atasoy MO, Naggar RFE, Rohaim MA, Munir M. Zoonotic and Zooanthroponotic Potential of Monkeypox. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:75-90. [PMID: 38801572 DOI: 10.1007/978-3-031-57165-7_5] [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
The current multicounty outbreak of monkeypox virus (MPXV) posed an emerging and continued challenge to already strained public healthcare sector, around the globe. Since its first identification, monkeypox disease (mpox) remained enzootic in Central and West African countries where reports of human cases are sporadically described. Recent trends in mpox spread outside the Africa have highlighted increased incidence of spillover of the MPXV from animal to humans. While nature of established animal reservoirs remained undefined, several small mammals including rodents, carnivores, lagomorphs, insectivores, non-human primates, domestic/farm animals, and several species of wildlife are proposed to be carrier of the MPXV infection. There are established records of animal-to-human (zoonotic) spread of MPXV through close interaction of humans with animals by eating bushmeat, contracting bodily fluids or trading possibly infected animals. In contrast, there are reports and increasing possibilities of human-to-animal (zooanthroponotic) spread of the MPXV through petting and close interaction with pet owners and animal care workers. We describe here the rationales and molecular factors which predispose the spread of MPXV not only amongst humans but also from animals to humans. A range of continuing opportunities for the spread and evolution of MPXV are discussed to consider risks beyond the currently identified groups. With the possibility of MPXV establishing itself in animal reservoirs, continued and broad surveillance, investigation into unconventional transmissions, and exploration of spillover events are warranted.
Collapse
Affiliation(s)
- Mustafa O Atasoy
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Rania F El Naggar
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Mohammed A Rohaim
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK.
| |
Collapse
|
10
|
Mungmunpuntipantip R, Wiwanitkit V. Annual ivermectin treatment, interferon-gamma, and responsiveness to monkeypox infection. Indian J Pharmacol 2023; 55:341-342. [PMID: 37929416 PMCID: PMC10751528 DOI: 10.4103/ijp.ijp_433_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/20/2023] [Accepted: 08/29/2023] [Indexed: 11/07/2023] Open
Affiliation(s)
| | - Viroj Wiwanitkit
- Department of Biological Science, Joseph Ayobaalola University, Ikeji-Arakeji, Nigeria
- Department of Community Medicine, Dr. DY Patil Vidhyapeeth, Pune, Maharashtra, India
- Department of Medical Science, Faculty of Medicine, University of Nis, Nis, Serbia
- Department of Tropical Medicine, Hainan Medical University, Haikou, China
- Research Center, Chandigarh University, Punjab, India
| |
Collapse
|
11
|
Xia H, He YR, Zhan XY, Zha GF. Mpox virus mRNA-lipid nanoparticle vaccine candidates evoke antibody responses and drive protection against the Vaccinia virus challenge in mice. Antiviral Res 2023; 216:105668. [PMID: 37429529 DOI: 10.1016/j.antiviral.2023.105668] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
In response to the human Mpox (hMPX) epidemic that began in 2022, there is an urgent need for a monkeypox vaccine. Here, we have developed a series of mRNA-lipid nanoparticle (mRNA-LNP)-based vaccine candidates that encode a collection of four highly conserved Mpox virus (MPXV) surface proteins involved in virus attachment, entry, and transmission, namely A29L, A35R, B6R, and M1R, which are homologs to Vaccinia virus (VACV) A27, A33, B5, and L1, respectively. Despite possible differences in immunogenicity among the four antigenic mRNA-LNPs, administering these antigenic mRNA-LNPs individually (5 μg each) or an average mixture of these mRNA-LNPs at a low dose (0.5 μg each) twice elicited MPXV-specific IgG antibodies and potent VACV-specific neutralizing antibodies. Furthermore, two doses of 5 μg of A27, B5, and L1 mRNA-LNPs or a 2 μg average mixture of the four antigenic mRNA-LNPs protected mice against weight loss and death after the VACV challenge. Overall, our data suggest that these antigenic mRNA-LNP vaccine candidates are both safe and efficacious against MPXV, as well as diseases caused by other orthopoxviruses.
Collapse
Affiliation(s)
- Heng Xia
- The Seventh Affiliated Hospital, Sun Yat-sen University, China
| | - Yun-Ru He
- The Seventh Affiliated Hospital, Sun Yat-sen University, China
| | - Xiao-Yong Zhan
- The Seventh Affiliated Hospital, Sun Yat-sen University, China.
| | - Gao-Feng Zha
- The Seventh Affiliated Hospital, Sun Yat-sen University, China.
| |
Collapse
|
12
|
Saadh MJ, Ghadimkhani T, Soltani N, Abbassioun A, Daniel Cosme Pecho R, Taha A, Jwad Kazem T, Yasamineh S, Gholizadeh O. Progress and prospects on vaccine development against monkeypox infection. Microb Pathog 2023; 180:106156. [PMID: 37201635 PMCID: PMC10186953 DOI: 10.1016/j.micpath.2023.106156] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
The monkeypox virus (MPOX) is an uncommon zoonotic illness brought on by an orthopoxvirus (OPXV). MPOX can occur with symptoms similar to smallpox. Since April 25, 2023, 110 nations have reported 87,113 confirmed cases and 111 fatalities. Moreover, the outspread prevalence of MPOX in Africa and a current outbreak of MPOX in the U.S. have made it clear that naturally occurring zoonotic OPXV infections remain a public health concern. Existing vaccines, though they provide cross-protection to MPOX, are not specific for the causative virus, and their effectiveness in the light of the current multi-country outbreak is still to be verified. Furthermore, as a sequel of the eradication and cessation of smallpox vaccination for four decades, MPOX found a possibility to re-emerge, but with distinct characteristics. The World Health Organization (WHO) suggested that nations use affordable MPOX vaccines within a framework of coordinated clinical effectiveness and safety evaluations. Vaccines administered in the smallpox control program and conferred immunity against MPOX. Currently, vaccines approved by WHO for use against MPOX are replicating (ACAM2000), low replicating (LC16m8), and non-replicating (MVA-BN). Although vaccines are accessible, investigations have demonstrated that smallpox vaccination is approximately 85% efficient in inhibiting MPOX. In addition, developing new vaccine methods against MPOX can help prevent this infection. To recognize the most efficient vaccine, it is essential to assess effects, including reactogenicity, safety, cytotoxicity effect, and vaccine-associated side effects, especially for high-risk and vulnerable people. Recently, several orthopoxvirus vaccines have been produced and are being evaluated. Hence, this review aims to provide an overview of the efforts dedicated to several types of vaccine candidates with different strategies for MPOX, including inactivated, live-attenuated, virus-like particles (VLPs), recombinant protein, nucleic acid, and nanoparticle-based vaccines, which are being developed and launched.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | | | - Narges Soltani
- School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Arian Abbassioun
- Department of Virology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Ali Taha
- Medical Technical College, Al-Farahidi University, Iraq
| | - Tareq Jwad Kazem
- Scientific Affairs Department, Al-Mustaqbal University, 51001, Hillah, Babylon, Iraq
| | - Saman Yasamineh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Gholizadeh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Tang D, Liu X, Lu J, Fan H, Xu X, Sun K, Wang R, Li C, Dan D, Du H, Wang Z, Li X, Yang X. Recombinant proteins A29L, M1R, A35R, and B6R vaccination protects mice from mpox virus challenge. Front Immunol 2023; 14:1203410. [PMID: 37435062 PMCID: PMC10331816 DOI: 10.3389/fimmu.2023.1203410] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
Since May 2022, mutant strains of mpox (formerly monkeypox) virus (MPXV) have been rapidly spreading among individuals who have not traveled to endemic areas in multiple locations, including Europe and the United States. Both intracellular and extracellular forms of mpox virus have multiple outer membrane proteins that can stimulate immune response. Here, we investigated the immunogenicity of MPXV structural proteins such as A29L, M1R, A35R, and B6R as a combination vaccine, and the protective effect against the 2022 mpox mutant strain was also evaluated in BALB/c mice. After mixed 15 μg QS-21 adjuvant, all four virus structural proteins were administered subcutaneously to mice. Antibody titers in mouse sera rose sharply after the initial boost, along with an increased capacity of immune cells to produce IFN-γ alongside an elevated level of cellular immunity mediated by Th1 cells. The vaccine-induced neutralizing antibodies significantly inhibited the replication of MPXV in mice and reduced the pathological damage of organs. This study demonstrates the feasibility of a multiple recombinant vaccine for MPXV variant strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zejun Wang
- *Correspondence: Zejun Wang, ; Xinguo Li, ; Xiaoming Yang,
| | - Xinguo Li
- *Correspondence: Zejun Wang, ; Xinguo Li, ; Xiaoming Yang,
| | - Xiaoming Yang
- *Correspondence: Zejun Wang, ; Xinguo Li, ; Xiaoming Yang,
| |
Collapse
|
14
|
Banuet-Martinez M, Yang Y, Jafari B, Kaur A, Butt ZA, Chen HH, Yanushkevich S, Moyles IR, Heffernan JM, Korosec CS. Monkeypox: a review of epidemiological modelling studies and how modelling has led to mechanistic insight. Epidemiol Infect 2023; 151:e121. [PMID: 37218612 PMCID: PMC10468816 DOI: 10.1017/s0950268823000791] [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: 02/13/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
Human monkeypox (mpox) virus is a viral zoonosis that belongs to the Orthopoxvirus genus of the Poxviridae family, which presents with similar symptoms as those seen in human smallpox patients. Mpox is an increasing concern globally, with over 80,000 cases in non-endemic countries as of December 2022. In this review, we provide a brief history and ecology of mpox, its basic virology, and the key differences in mpox viral fitness traits before and after 2022. We summarize and critique current knowledge from epidemiological mathematical models, within-host models, and between-host transmission models using the One Health approach, where we distinguish between models that focus on immunity from vaccination, geography, climatic variables, as well as animal models. We report various epidemiological parameters, such as the reproduction number, R0, in a condensed format to facilitate comparison between studies. We focus on how mathematical modelling studies have led to novel mechanistic insight into mpox transmission and pathogenesis. As mpox is predicted to lead to further infection peaks in many historically non-endemic countries, mathematical modelling studies of mpox can provide rapid actionable insights into viral dynamics to guide public health measures and mitigation strategies.
Collapse
Affiliation(s)
- Marina Banuet-Martinez
- Climate Change and Global Health Research Group, School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Yang Yang
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Behnaz Jafari
- Mathematics and Statistics Department, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Avneet Kaur
- Irving K. Barber School of Arts and Sciences, Department of Computer Science, Mathematics, Physics and Statistics, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Zahid A. Butt
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Helen H. Chen
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Svetlana Yanushkevich
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Iain R. Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Jane M. Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, Toronto, ON, Canada
| |
Collapse
|
15
|
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:v15040905. [PMID: 37112885 PMCID: PMC10142277 DOI: 10.3390/v15040905] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [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
|
16
|
Alcamí A. Pathogenesis of the circulating mpox virus and its adaptation to humans. Proc Natl Acad Sci U S A 2023; 120:e2301662120. [PMID: 36940331 PMCID: PMC10068839 DOI: 10.1073/pnas.2301662120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
Affiliation(s)
- Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), 28049Madrid, Spain
| |
Collapse
|
17
|
Anand A, Das AK, Bhardwaj S, Singh SK. A brief review of the monkeypox virus and emerging concerns for neuroinvasiveness. Surg Neurol Int 2023; 14:78. [PMID: 37025545 PMCID: PMC10070311 DOI: 10.25259/sni_1176_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Background:
Amidst the ongoing COVID-19 pandemic, monkeypox virus (MPXV) disease has been recognized as another disease of pandemic nature by the World Health Organization. Nearly four decades after the eradication of smallpox, as half of the world population is naïve to ortho-pox viruses (supposedly due to lack of immunity by vaccination), MPXV remains the most pathogenic species of the family of poxviruses.
Methods:
The articles on MPXV were searched on PubMed/Medline and data were retrieved and analyzed.
Results:
Although reported as a disease of milder exanthem and lower mortality as compared to smallpox, the MPXV disease tends to be neuroinvasive. This article highlights the neurological signs and symptoms of MPXV disease and discusses, in brief, the management strategies.
Conclusion:
Neuroinvasive properties of the virus as demonstrated in in vitro studies, and further verified by neurological illnesses in patients, present a special threat to mankind. Clinicians must be prepared to recognize and treat these neurological complications and start treatment to limit long-lasting brain injury as seen in patients with COVID-19.
Collapse
Affiliation(s)
- Atul Anand
- Department of General Surgery, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Anand Kumar Das
- Department of Neurosurgery, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Sona Bhardwaj
- Department of Microbiology, ESIC Hospital, Patna, Bihar, India
| | - Saraj Kumar Singh
- Department of Neurosurgery, All India Institute of Medical Sciences, Patna, Bihar, India
| |
Collapse
|
18
|
Americo JL, Earl PL, Moss B. Virulence differences of mpox (monkeypox) virus clades I, IIa, and IIb.1 in a small animal model. Proc Natl Acad Sci U S A 2023; 120:e2220415120. [PMID: 36787354 PMCID: PMC9974501 DOI: 10.1073/pnas.2220415120] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/19/2023] [Indexed: 02/15/2023] Open
Abstract
Human mpox (monkeypox), a disease with similarities to smallpox, is endemic in Africa where it has persisted as a zoonosis with limited human-to-human spread. Unexpectedly, the disease expanded globally in 2022 driven by human-to-human transmission outside of Africa. It is not yet known whether the latter is due solely to behavioral and environmental factors or whether the mpox virus is adapting to a new host. Genome sequencing has revealed differences between the current outbreak strains, classified as clade IIb, and the prior clade IIa and clade I viruses, but whether these differences contribute to virulence or transmission has not been determined. We demonstrate that the wild-derived inbred castaneous mouse provides an exceptional animal model for investigating clade differences in mpox virus virulence and show that the order is clade I > clade IIa > clade IIb.1. The greatly reduced replication of the clade IIb.1 major outbreak strain in mice and absence of lethality at 100 times the lethal dose of a closely related clade IIa virus, despite similar multiplication in cell culture, suggest that clade IIb is evolving diminished virulence or adapting to other species.
Collapse
Affiliation(s)
- Jeffrey L. Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20814
| | - Patricia L. Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20814
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20814
| |
Collapse
|
19
|
Rosa RB, Ferreira de Castro E, Vieira da Silva M, Paiva Ferreira DC, Jardim ACG, Santos IA, Marinho MDS, Ferreira França FB, Pena LJ. In vitro and in vivo models for monkeypox. iScience 2023; 26:105702. [PMID: 36471873 PMCID: PMC9712139 DOI: 10.1016/j.isci.2022.105702] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The emergence and rapid spread outside of monkeypox virus (MPXV) to non-endemic areas has led to another global health emergency in the midst of the COVID-19 pandemic. The scientific community has sought to rapidly develop in vitro and in vivo models that could be applied in research with MPXV. In vitro models include two-dimensional (2D) cultures of immortalized cell lines or primary cells and three-dimensional (3D) cultures. In vitro models are considered cost-effective and can be done in highly controlled conditions; however, they do not always resemble physiological conditions. In this way, several in vivo models are being characterized to meet the growing demand for new studies related to MPXV. In this review, we summarize the main MPXV models that have already been developed and discuss how they can contribute to advance the understanding of its pathogenesis, replication, and transmission, as well as identifying antivirals to treat infected patients.
Collapse
Affiliation(s)
- Rafael Borges Rosa
- Department of Virology and Experimental Therapy (LAVITE), Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil
- Rodents Animal Facilities Complex, Federal University of Uberlândia (REBIR-UFU), Uberlândia 38400-902, Brazil
| | - Emilene Ferreira de Castro
- Rodents Animal Facilities Complex, Federal University of Uberlândia (REBIR-UFU), Uberlândia 38400-902, Brazil
| | - Murilo Vieira da Silva
- Rodents Animal Facilities Complex, Federal University of Uberlândia (REBIR-UFU), Uberlândia 38400-902, Brazil
| | | | | | - Igor Andrade Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38405-302, Brazil
| | | | | | - Lindomar José Pena
- Department of Virology and Experimental Therapy (LAVITE), Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil
| |
Collapse
|
20
|
Monkeypox virus from neurological complications to neuroinvasive properties: current status and future perspectives. J Neurol 2023; 270:101-108. [PMID: 35989372 PMCID: PMC9393054 DOI: 10.1007/s00415-022-11339-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 01/07/2023]
Abstract
Cases of monkeypox (MPV) are sharply rising around the world. While most efforts are being focused on the management of the first symptoms of monkeypox, such as cutaneous lesions and flu-like symptoms, the effect of the monkeypox virus (MPXV) on multiple organs still remains unclear. Recently, several neurological manifestations, such as headache, myalgia, malaise, fatigue, altered consciousness, agitation, anorexia, nausea, and vomiting, have been reported in patients with MPV. In addition, data from experimental studies have indicated that MPXV can gain access to the central nervous system (CNS) through the olfactory epithelium and infected circulatory monocytes/macrophages as two probable neuroinvasive mechanisms. Therefore, there are growing concerns about the long-term effect of MPXV on the CNS and subsequent neurological complications. This paper highlights the importance of the neuroinvasive potential of MPXV, coupled with neurological manifestations.
Collapse
|
21
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Ahmad S, Hasan H, Ahmad Suhaimi NA, Albakri KA, Abedalbaset Alzyoud A, Kadir R, Mohamud R. Comprehensive literature review of monkeypox. Emerg Microbes Infect 2022; 11:2600-2631. [PMID: 36263798 PMCID: PMC9627636 DOI: 10.1080/22221751.2022.2132882] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/02/2022] [Indexed: 11/03/2022]
Abstract
The current outbreak of monkeypox (MPX) infection has emerged as a global matter of concern in the last few months. MPX is a zoonosis caused by the MPX virus (MPXV), which is one of the Orthopoxvirus species. Thus, it is similar to smallpox caused by the variola virus, and smallpox vaccines and drugs have been shown to be protective against MPX. Although MPX is not a new disease and is rarely fatal, the current multi-country MPX outbreak is unusual because it is occurring in countries that are not endemic for MPXV. In this work, we reviewed the extensive literature available on MPXV to summarize the available data on the major biological, clinical and epidemiological aspects of the virus and the important scientific findings. This review may be helpful in raising awareness of MPXV transmission, symptoms and signs, prevention and protective measures. It may also be of interest as a basis for performance of studies to further understand MPXV, with the goal of combating the current outbreak and boosting healthcare services and hygiene practices.Trial registration: ClinicalTrials.gov identifier: NCT02977715..Trial registration: ClinicalTrials.gov identifier: NCT03745131..Trial registration: ClinicalTrials.gov identifier: NCT00728689..Trial registration: ClinicalTrials.gov identifier: NCT02080767..
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | | | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | | | | | | | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
22
|
Chen JM, Chen RX, Gong HY, Zhao MM, Ji YF, Sun MH, Li GH, Tan SM, Zhang GH, Chen JW. Epidemiology-based analysis of the risks and elimination strategies of the monkeypox outbreak in 2022. Front Vet Sci 2022; 9:1064766. [PMID: 36532347 PMCID: PMC9748476 DOI: 10.3389/fvets.2022.1064766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/14/2022] [Indexed: 08/30/2023] Open
Abstract
Human monkeypox, caused by monkeypox virus, has spread unprecedentedly to more than 100 countries since May 2022. Here we summarized the epidemiology of monkeypox through a literature review and elucidated the risks and elimination strategies of this outbreak mainly based on the summarized epidemiology. We demonstrated that monkeypox virus became more contagious and less virulent in 2022, which could result from the fact that the virus entered a special transmission network favoring close contacts (i.e., sexual behaviors of men who have sex with men outside Africa) and the possibility that the virus accumulated a few adaptive mutations. We gave the reasons to investigate whether cattle, goats, sheep, and pigs are susceptible to monkeypox virus and whether infection with monkeypox virus could be latent in some primates. We listed six potential scenarios for the future of the outbreak (e.g., the outbreak could lead to endemicity outside Africa with increased transmissibility or virulence). We also listed multiple factors aiding or impeding the elimination of the outbreak. We showed that the control measures strengthened worldwide after the World Health Organization declared the outbreak a public health emergency of international concern (PHEIC) could eliminate the outbreak in 2022. We clarified eight strategies, i.e., publicity and education, case isolation, vaccine stockpiling, risk-based vaccination or ring vaccination, importation quarantine, international collaboration, and laboratory management, for the elimination of the outbreak.
Collapse
Affiliation(s)
- Ji-Ming Chen
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Rui-Xu Chen
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Huan-Yu Gong
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Meng-Meng Zhao
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yu-Fei Ji
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Ming-Hui Sun
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Guo-Hui Li
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Su-Mei Tan
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Gui-Hong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ji-Wang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
23
|
Al-Musa A, Chou J, LaBere B. The resurgence of a neglected orthopoxvirus: Immunologic and clinical aspects of monkeypox virus infections over the past six decades. Clin Immunol 2022; 243:109108. [PMID: 36067982 PMCID: PMC9628774 DOI: 10.1016/j.clim.2022.109108] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022]
Abstract
Monkeypox is a zoonotic Orthopoxvirus which has predominantly affected humans living in western and central Africa since the 1970s. Type I and II interferon signaling, NK cell function, and serologic immunity are critical for host immunity against monkeypox. Monkeypox can evade host viral recognition and block interferon signaling, leading to overall case fatality rates of up to 11%. The incidence of monkeypox has increased since cessation of smallpox vaccination. In 2022, a global outbreak emerged, predominantly affecting males, with exclusive human-to-human transmission and more phenotypic variability than earlier outbreaks. Available vaccines are safe and effective tools for prevention of severe disease, but supply is limited. Now considered a public health emergency, more studies are needed to better characterize at-risk populations and to develop new anti-viral therapies.
Collapse
Affiliation(s)
- Amer Al-Musa
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA..
| | - Brenna LaBere
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA..
| |
Collapse
|
24
|
Panda K, Mukherjee A. Is monkeypox a threat to another pandemic? Front Microbiol 2022; 13:983076. [PMID: 36118218 PMCID: PMC9470849 DOI: 10.3389/fmicb.2022.983076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
| | - Anupam Mukherjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune, MH, India
- *Correspondence: Anupam Mukherjee ;
| |
Collapse
|
25
|
Chakraborty S, Chandran D, Mohapatra RK, Alagawany M, El-Shall NA, Sharma AK, Chakraborty C, Dhama K. Clinical management, antiviral drugs and immunotherapeutics for treating monkeypox. An update on current knowledge and futuristic prospects. Int J Surg 2022; 105:106847. [PMID: 35995352 PMCID: PMC9533875 DOI: 10.1016/j.ijsu.2022.106847] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, R.K. Nagar, West Tripura, Tripura, 799008, India
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, 642109, Tamil Nadu, India
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, 758002, Odisha, India.
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Nahed A El-Shall
- Department of Poultry and Fish Diseases, Faculty of Veterinary Medicine, Alexandria University, Edfina, El-Beheira, 22758, Egypt
| | - Anil K Sharma
- Department of Biotechnology, Maharishi Markandeshwar University (Deemed to be University) Mullana-Ambala, 133207, Haryana, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| |
Collapse
|
26
|
MacNeill AL. Comparative Pathology of Zoonotic Orthopoxviruses. Pathogens 2022; 11:pathogens11080892. [PMID: 36015017 PMCID: PMC9412692 DOI: 10.3390/pathogens11080892] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
This review provides a brief history of the impacts that a human-specific Orthopoxvirus (OPXV), Variola virus, had on mankind, recalls how critical vaccination was for the eradication of this disease, and discusses the consequences of discontinuing vaccination against OPXV. One of these consequences is the emergence of zoonotic OPXV diseases, including Monkeypox virus (MPXV). The focus of this manuscript is to compare pathology associated with zoonotic OPXV infection in veterinary species and in humans. Efficient recognition of poxvirus lesions and other, more subtle signs of disease in multiple species is critical to prevent further spread of poxvirus infections. Additionally included are a synopsis of the pathology observed in animal models of MPXV infection, the recent spread of MPXV among humans, and a discussion of the potential for this virus to persist in Europe and the Americas.
Collapse
Affiliation(s)
- Amy L MacNeill
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
27
|
Parker S, D'Angelo J, Buller RM, Smee DF, Lantto J, Nielsen H, Jensen A, Prichard M, George SL. A human recombinant analogue to plasma-derived vaccinia immunoglobulin prophylactically and therapeutically protects against lethal orthopoxvirus challenge. Antiviral Res 2021; 195:105179. [PMID: 34530009 PMCID: PMC9628779 DOI: 10.1016/j.antiviral.2021.105179] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/22/2022]
Abstract
Orthopoxviruses such as variola and monkeypox viruses continue to threaten the human population. Monkeypox virus is endemic in central and western Africa and outbreaks have reached as far as the U.S. Although variola virus, the etiologic agent of smallpox, has been eradicated by a successful vaccination program, official and likely clandestine stocks of the virus exist. Moreover, studies with ectromelia virus (the etiological agent of mousepox) have revealed that IL-4 recombinant viruses are significantly more virulent than wild-type viruses even in mice treated with vaccines and/or antivirals. For these reasons, it is critical that antiviral modalities are developed to treat these viruses should outbreaks, or deliberate dissemination, occur. Currently, 2 antivirals (brincidofovir and tecovirimat) are in the U.S. stockpile allowing for emergency use of the drugs to treat smallpox. Both antivirals have advantages and disadvantages in a clinical and emergency setting. Here we report on the efficacy of a recombinant immunoglobulin (rVIG) that demonstrated efficacy against several orthopoxviruses in vitro and in vivo in both a prophylactic and therapeutic fashion. A single intraperitoneal injection of rVIG significantly protected mice when given up to 14 days before or as late as 6 days post challenge. Moreover, rVIG reduced morbidity, as measured by weight-change, as well as several previously established biomarkers of disease. In rVIG treated mice, we found that vDNA levels in blood were significantly reduced, as was ALT (a marker of liver damage) and infectious virus levels in the liver. No apparent adverse events were observed in rVIG treated mice, suggesting the immunoglobulin is well tolerated. These findings suggest that recombinant immunoglobulins could be candidates for further evaluation and possible licensure under the FDA Animal Rule.
Collapse
Affiliation(s)
- Scott Parker
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA
| | - June D'Angelo
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA
| | - R Mark Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, 63104, USA
| | - Donald F Smee
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Johan Lantto
- Symphogen, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | | | - Allan Jensen
- Symphogen, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Mark Prichard
- Department of Pediatrics, University of Alabama, Birmingham, AL, 35233, USA
| | - Sarah L George
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA.
| |
Collapse
|
28
|
Yang HS, Onos KD, Choi K, Keezer KJ, Skelly DA, Carter GW, Howell GR. Natural genetic variation determines microglia heterogeneity in wild-derived mouse models of Alzheimer's disease. Cell Rep 2021; 34:108739. [PMID: 33567283 PMCID: PMC7937391 DOI: 10.1016/j.celrep.2021.108739] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/09/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Genetic and genome-wide association studies suggest a central role for microglia in Alzheimer’s disease (AD). However, single-cell RNA sequencing (scRNA-seq) of microglia in mice, a key preclinical model, has shown mixed results regarding translatability to human studies. To address this, scRNA-seq of microglia from C57BL/6J (B6) and wild-derived strains (WSB/EiJ, CAST/EiJ, and PWK/PhJ) with and without APP/PS1 demonstrates that genetic diversity significantly alters features and dynamics of microglia in baseline neuroimmune functions and in response to amyloidosis. Results show significant variation in the abundance of microglial subtypes or states, including numbers of previously identified disease-associated and interferon-responding microglia, across the strains. For each subtype, significant differences in the expression of many genes are observed in wild-derived strains relative to B6, including 19 genes previously associated with human AD including Apoe, Trem2, and Sorl1. This resource is critical in the development of appropriately targeted therapeutics for AD and other neurological diseases. Neuroinflammation is a key component of Alzheimer’s disease. Yang et al. perform single-cell sequencing of microglia in wild-derived mouse strains that carry amyloid and show that these strains differ from the commonly used strains, exhibiting significant variation in abundance of microglial subtypes, including numbers of disease-associated and interferon-responding microglia.
Collapse
Affiliation(s)
| | | | | | | | | | - Gregory W Carter
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
29
|
Morgan CN, Matheny AM, Nakazawa YJ, Kling C, Gallardo-Romero N, Seigler L, Barbosa Costa G, Hutson C, Maghlakelidze G, Olson V, Doty JB. Laboratory Infection of Novel Akhmeta Virus in CAST/EiJ Mice. Viruses 2020; 12:v12121416. [PMID: 33317132 PMCID: PMC7763702 DOI: 10.3390/v12121416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
Akhmeta virus is a zoonotic Orthopoxvirus first identified in 2013 in the country of Georgia. Subsequent ecological investigations in Georgia have found evidence that this virus is widespread in its geographic distribution within the country and in its host-range, with rodents likely involved in its circulation in the wild. Yet, little is known about the pathogenicity of this virus in rodents. We conducted the first laboratory infection of Akhmeta virus in CAST/EiJ Mus musculus to further characterize this novel virus. We found a dose-dependent effect on mortality and weight loss (p < 0.05). Anti-orthopoxvirus antibodies were detected in the second- and third-highest dose groups (5 × 104 pfu and 3 × 102 pfu) at euthanasia by day 10, and day 14 post-infection, respectively. Anti-orthopoxvirus antibodies were not detected in the highest dose group (3 × 106 pfu), which were euthanized at day 7 post-infection and had high viral load in tissues, suggesting they succumbed to disease prior to mounting an effective immune response. In order of highest burden, viable virus was detected in the nostril, lung, tail, liver and spleen. All individuals tested in the highest dose groups were DNAemic. Akhmeta virus was highly pathogenic in CAST/EiJ Mus musculus, causing 100% mortality when ≥3 × 102 pfu was administered.
Collapse
Affiliation(s)
- Clint N. Morgan
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
- Correspondence: ; Tel.: +1-404-639-0844
| | - Audrey M. Matheny
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
- Oak Ridge Institute for Science and Education, CDC Fellowship Program, Oak Ridge, TN 37830, USA
| | - Yoshinori J. Nakazawa
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
| | - Chantal Kling
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
- Oak Ridge Institute for Science and Education, CDC Fellowship Program, Oak Ridge, TN 37830, USA
| | - Nadia Gallardo-Romero
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
| | - Laurie Seigler
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
- Kāpili Services, LLC, An Alaka’ina Foundation Company, Honolulu, HI 96814, USA
| | - Galileu Barbosa Costa
- Núcleo de Epidemiologia e Bioestatística, Centro de Pesquisas Gonçalo Moniz, Fiocruz, Bahia 40296-710, Brazil;
| | - Christina Hutson
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
| | - Giorgi Maghlakelidze
- U.S. Centers for Disease Control and Prevention, South Caucuses Office, Tbilisi 0177, Georgia;
| | - Victoria Olson
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
| | - Jeffrey B. Doty
- Poxvirus & Rabies Branch, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Division of High-Consequence Pathogens & Pathology, Atlanta, GA 30329, USA; (A.M.M.); (Y.J.N.); (C.K.); (N.G.-R.); (L.S.); (C.H.); (V.O.); (J.B.D.)
| |
Collapse
|
30
|
Alakunle E, Moens U, Nchinda G, Okeke MI. Monkeypox Virus in Nigeria: Infection Biology, Epidemiology, and Evolution. Viruses 2020; 12:E1257. [PMID: 33167496 PMCID: PMC7694534 DOI: 10.3390/v12111257] [Citation(s) in RCA: 370] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/22/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Monkeypox is a zoonotic disease caused by monkeypox virus (MPXV), which is a member of orthopoxvirus genus. The reemergence of MPXV in 2017 (at Bayelsa state) after 39 years of no reported case in Nigeria, and the export of travelers' monkeypox (MPX) from Nigeria to other parts of the world, in 2018 and 2019, respectively, have raised concern that MPXV may have emerged to occupy the ecological and immunological niche vacated by smallpox virus. This review X-rays the current state of knowledge pertaining the infection biology, epidemiology, and evolution of MPXV in Nigeria and worldwide, especially with regard to the human, cellular, and viral factors that modulate the virus transmission dynamics, infection, and its maintenance in nature. This paper also elucidates the role of recombination, gene loss and gene gain in MPXV evolution, chronicles the role of signaling in MPXV infection, and reviews the current therapeutic options available for the treatment and prevention of MPX. Additionally, genome-wide phylogenetic analysis was undertaken, and we show that MPXV isolates from recent 2017 outbreak in Nigeria were monophyletic with the isolate exported to Israel from Nigeria but do not share the most recent common ancestor with isolates obtained from earlier outbreaks, in 1971 and 1978, respectively. Finally, the review highlighted gaps in knowledge particularly the non-identification of a definitive reservoir host animal for MPXV and proposed future research endeavors to address the unresolved questions.
Collapse
Affiliation(s)
- Emmanuel Alakunle
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, School of Arts and Sciences, American University of Nigeria, 98 Lamido Zubairu Way, PMB 2250 Yola, Nigeria;
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, University i Tromsø (UIT)—The Arctic University of Norway, N-9037 Tromsø, Norway;
| | - Godwin Nchinda
- Laboratory of Vaccinology and Immunology, The Chantal Biya International Reference Center for Research on the Prevention and Management HIV/AIDS (CIRCB), P.O Box 3077 Yaoundé-Messa, Cameroon;
- Department of Pharmaceutical Microbiology & Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, P.O Box 420110 Awka, Nigeria
| | - Malachy Ifeanyi Okeke
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, School of Arts and Sciences, American University of Nigeria, 98 Lamido Zubairu Way, PMB 2250 Yola, Nigeria;
| |
Collapse
|
31
|
Earl PL, Americo JL, Moss B. Natural killer cells expanded in vivo or ex vivo with IL-15 overcomes the inherent susceptibility of CAST mice to lethal infection with orthopoxviruses. PLoS Pathog 2020; 16:e1008505. [PMID: 32320436 PMCID: PMC7197867 DOI: 10.1371/journal.ppat.1008505] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 05/04/2020] [Accepted: 03/26/2020] [Indexed: 02/05/2023] Open
Abstract
The wild-derived inbred CAST/EiJ mouse, one of eight founder strains in the Collaborative Cross panel, is an exceptional model for studying monkeypox virus (MPXV), an emerging human pathogen, and other orthopoxviruses including vaccinia virus (VACV). Previous studies suggested that the extreme susceptibility of the CAST mouse to orthopoxviruses is due to an insufficient innate immune response. Here, we focused on the low number of natural killer (NK) cells in the naïve CAST mouse as a contributing factor to this condition. Administration of IL-15 to CAST mice transiently increased NK and CD8+ T cells that could express IFN-γ, indicating that the progenitor cells were capable of responding to cytokines. However, the number of NK cells rapidly declined indicating a defect in their homeostasis. Furthermore, IL-15-treated mice were protected from an otherwise lethal challenge with VACV or MPXV. IL-15 decreased virus spread and delayed death even when CD4+/CD8+ T cells were depleted with antibody, supporting an early protective role of the expanded NK cells. Purified splenic NK cells from CAST mice proliferated in vitro in response to IL-15 and could be activated with IL-12/IL-18 to secrete interferon-γ. Passive transfer of non-activated or activated CAST NK cells reduced VACV spread but only the latter completely prevented death at the virus dose used. Moreover, antibodies to interferon-γ abrogated the protection by activated NK cells. Thus, the inherent susceptibility of CAST mice to orthopoxviruses can be explained by a low level of NK cells and this vulnerability can be overcome either by expanding their NK cells in vivo with IL-15 or by passive transfer of purified NK cells that were expanded and activated in vitro. With the eradication of smallpox, monkeypox virus (MPXV) remains the only poxvirus causing significant mortality in humans. Although endemic in parts of Africa, human infections have occurred in the United States, the United Kingdom and Israel due to travelers or imported animals. Contrary to its name, MPXV primarily infects rodents and secondarily infects humans and other primates. The wild-derived CAST mouse is an excellent small animal model for studying the pathogenicity of MPXV and related orthopoxviruses including vaccinia virus (VACV) and for evaluating therapeutics. We previously found that the susceptibility of CAST mice is correlated with low numbers of natural killer (NK) cells and a delayed interferon-γ response. Here we showed that in vivo administration of the cytokine IL-15 transiently raised NK cell numbers and protected CAST mice from systemic infections with VACV and MPXV. CAST mouse NK cells that were purified and expanded in vitro with IL-15 also provided protection, further demonstrating the important role of NK cells. The rapid decline in NK cell numbers following cessation of IL-15 administration or NK cell transfer suggests that a low level of NK cell homeostasis contributes to the susceptibility of CAST mice to virus infection.
Collapse
Affiliation(s)
- Patricia L. Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey L. Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
Forty years after the last endemic smallpox case, variola virus (VARV) is still considered a major threat to humans due to its possible use as a bioterrorism agent. For many years, the risk of disease reemergence was thought to solely be through deliberate misuse of VARV strains kept in clandestine laboratories. However, recent experiments using synthetic biology have proven the feasibility of recreating a poxvirus de novo, implying that VARV could, in theory, be resurrected. Because of this new perspective, the WHO Advisory Committee on VARV Research released new recommendations concerning research on poxviruses that strongly encourages pursuing the development of new antiviral drugs against orthopoxviruses. In 2018, the U.S. FDA advised in favor of two molecules for smallpox treatment, tecovirimat and brincidofovir. This review highlights the difficulties to develop new drugs targeting an eradicated disease, especially as it requires working under the FDA "animal efficacy rule" with the few, and imperfect, animal models available.
Collapse
|
33
|
Abstract
The variable outcome of Mycobacterium tuberculosis infection observed in natural populations is difficult to model in genetically homogeneous small-animal models. The newly developed Collaborative Cross (CC) represents a reproducible panel of genetically diverse mice that display a broad range of phenotypic responses to infection. We explored the genetic basis of this variation, focusing on a CC line that is highly susceptible to M. tuberculosis infection. This study identified multiple quantitative trait loci associated with bacterial control and cytokine production, including one that is caused by a novel loss-of-function mutation in the Itgal gene, which is necessary for T cell recruitment to the infected lung. These studies verify the multigenic control of mycobacterial disease in the CC panel, identify genetic loci controlling diverse aspects of pathogenesis, and highlight the utility of the CC resource. Host genetics plays an important role in determining the outcome of Mycobacterium tuberculosis
infection. We previously found that Collaborative Cross (CC) mouse strains differ in their susceptibility to M. tuberculosis and that the CC042/GeniUnc (CC042) strain suffered from a rapidly progressive disease and failed to produce the protective cytokine gamma interferon (IFN-γ) in the lung. Here, we used parallel genetic and immunological approaches to investigate the basis of CC042 mouse susceptibility. Using a population derived from a CC001/Unc (CC001) × CC042 intercross, we mapped four quantitative trait loci (QTL) underlying tuberculosis immunophenotypes (Tip1 to Tip4). These included QTL that were associated with bacterial burden, IFN-γ production following infection, and an IFN-γ-independent mechanism of bacterial control. Further immunological characterization revealed that CC042 animals recruited relatively few antigen-specific T cells to the lung and that these T cells failed to express the integrin alpha L (αL; i.e., CD11a), which contributes to T cell activation and migration. These defects could be explained by a CC042 private variant in the Itgal gene, which encodes CD11a and is found within the Tip2 interval. This 15-bp deletion leads to aberrant mRNA splicing and is predicted to result in a truncated protein product. The ItgalCC042 genotype was associated with all measured disease traits, indicating that this variant is a major determinant of susceptibility in CC042 mice. The combined effect of functionally distinct Tip variants likely explains the profound susceptibility of CC042 mice and highlights the multigenic nature of tuberculosis control in the Collaborative Cross.
Collapse
|
34
|
Gallardo-Romero NF, Hutson CL, Carroll D, Kondas AV, Salzer JS, Dietz-Ostergaard S, Smith S, Hudson P, Olson V, Damon I. Use of live Variola virus to determine whether CAST/EiJ mice are a suitable surrogate animal model for human smallpox. Virus Res 2019; 275:197772. [PMID: 31593747 PMCID: PMC9533991 DOI: 10.1016/j.virusres.2019.197772] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 11/19/2022]
Abstract
Numerous animal models of systemic orthopoxvirus disease have been developed to evaluate therapeutics against variola virus (VARV), the causative agent of smallpox. These animal models do not resemble the disease presentation in human smallpox and most used surrogate Orthopoxviruses. A rodent model using VARV has a multitude of advantages, and previous investigations identified the CAST/EiJ mouse as highly susceptible to monkeypox virus infection, making it of interest to determine if these rodents are also susceptible to VARV infection. In this study, we inoculated CAST/EiJ mice with a range of VARV doses (102-106 plaque forming units). Some animals had detectable viable VARV from the oropharynx between days 3 and 12 post inoculation. Despite evidence of disease, the CAST/EiJ mouse does not provide a model for clinical smallpox due to mild signs of morbidity and limited skin lesions. However, in contrast to previous rodent models using VARV challenge (i.e. prairie dogs and SCID mice), a robust immune response was observed in the CAST/EiJ mice (measured by Immunoglobulin G enzyme-linked immunosorbent assay). This is an advantage of this model for the study of VARV and presents a unique potential for the study of the immunomodulatory pathways following VARV infection.
Collapse
Affiliation(s)
- Nadia F Gallardo-Romero
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Christina L Hutson
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Darin Carroll
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Ashley V Kondas
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Johanna S Salzer
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Sharon Dietz-Ostergaard
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of Scientific resources, Comparative Medicine Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Scott Smith
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Paul Hudson
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Victoria Olson
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| | - Inger Damon
- Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA.
| |
Collapse
|
35
|
Reynolds MG, Doty JB, McCollum AM, Olson VA, Nakazawa Y. Monkeypox re-emergence in Africa: a call to expand the concept and practice of One Health. Expert Rev Anti Infect Ther 2019; 17:129-139. [PMID: 30625020 PMCID: PMC6438170 DOI: 10.1080/14787210.2019.1567330] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/03/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Monkeypox is a re-emerging viral zoonosis that occurs naturally in heavily forested regions of West and Central Africa. Inter-human transmission of monkeypox virus, although limited, drives outbreaks, particularly in household and health-care settings. But the available evidence suggests that without repeated zoonotic introductions, human infections would eventually cease to occur. Therefore, interrupting virus transmission from animals to humans is key to combating this disease. Areas covered: Herein we review laboratory and field studies examining the susceptibility of various animal taxa to monkeypox virus infection, and note the competence of various species to serve as reservoirs or transmission hosts. In addition, we discuss early socio-ecologic theories of monkeypox virus transmission in rural settings and review current modes of ecologic investigation - including ecologic niche modeling, and ecologic sampling - in light of their potential to identify specific animal species and features of the environment that are associated with heightened risk for human disease. Expert opinion: The role of disease ecology and scientific research in ongoing disease prevention efforts should be reinforced, particularly for wildlife-associated zoonoses such as monkeypox. Such efforts alongside those aimed at nurturing 'One Health' collaborations may ultimately hold the greatest promise for reducing human infections with this pathogen.
Collapse
Affiliation(s)
- Mary G. Reynolds
- US Centers for Disease Control and Prevention, Poxvirus and Rabies Branch, Atlanta, GA, USA
| | - Jeffry B. Doty
- US Centers for Disease Control and Prevention, Poxvirus and Rabies Branch, Atlanta, GA, USA
| | - Andrea M. McCollum
- US Centers for Disease Control and Prevention, Poxvirus and Rabies Branch, Atlanta, GA, USA
| | - Victoria A. Olson
- US Centers for Disease Control and Prevention, Poxvirus and Rabies Branch, Atlanta, GA, USA
| | - Yoshinori Nakazawa
- US Centers for Disease Control and Prevention, Poxvirus and Rabies Branch, Atlanta, GA, USA
| |
Collapse
|
36
|
Poltorak A, Apalko S, Sherbak S. Wild-derived mice: from genetic diversity to variation in immune responses. Mamm Genome 2018; 29:577-584. [PMID: 30056578 DOI: 10.1007/s00335-018-9766-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/23/2018] [Indexed: 10/28/2022]
Abstract
Classical inbred mouse strains have historically been instrumental in mapping immunological traits. However, most of the classical strains originate from a relatively limited number of founder animals, largely within the Mus musculus domesticus subspecies. Therefore, their genetic diversity is ultimately limited. For this reason, it is not feasible to use these mice for exhaustive interrogation of immune signaling pathways. In order to investigate networks through forward genetic analysis, larger genetic diversity is required than is introduced under laboratory conditions. Recently, inbred strains from other mouse subspecies were established such as Mus musculus castaneus and Mus musculus musculus, which diverged from a shared common ancestor with Mus musculus domesticus more than one million years ago. A direct genomic comparison clearly demonstrates the evolutionary divergence that has occurred between wild-derived mice and the classical inbred strains. When compared to classical inbred strains, wild-derived mice exhibit polymorphisms every 100-200 base pairs. Studying the molecular basis of these traits provides us with insight into how the immune system can evolve regulatory features to accommodate environment-specific constraints. Because most wild-derived strains are able to breed with classical inbred mice, they represent a rich source of evolutionarily significant diversity for forward genetic studies. These organisms are an emerging, though still largely unexplored, model for the identification and study of novel immunological genes.
Collapse
Affiliation(s)
- Alexander Poltorak
- Department of Immunology, Tufts University, Boston, MA, 02111, USA. .,Petrozavodsk State University, Karelia, Russian Federation.
| | | | - Sergei Sherbak
- City Hospital, 40, St. Petersburg, Russian Federation.,St. Petersburg State University, St. Petersburg, Russian Federation
| |
Collapse
|
37
|
Mucker EM, Wollen-Roberts SE, Kimmel A, Shamblin J, Sampey D, Hooper JW. Intranasal monkeypox marmoset model: Prophylactic antibody treatment provides benefit against severe monkeypox virus disease. PLoS Negl Trop Dis 2018; 12:e0006581. [PMID: 29927927 PMCID: PMC6029809 DOI: 10.1371/journal.pntd.0006581] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 07/03/2018] [Accepted: 06/04/2018] [Indexed: 02/08/2023] Open
Abstract
Concerns regarding outbreaks of human monkeypox or the potential reintroduction of smallpox into an immunological naïve population have prompted the development of animal models and countermeasures. Here we present a marmoset model of monkeypox and smallpox disease utilizing a relevant poxvirus via a natural exposure route. We found that 1000 plaque forming units (PFU) of Monkeypox virus was sufficient to recapitulate smallpox disease, to include an incubation period of approximately 13 days, followed by the onset of rash, and death between 15 and 17 days. Temporally accurate manifestation of viremia and oral shedding were also features. The number of lesions ranged from no lesions to 299, the most reported in a marmoset exposed to a poxvirus. To both evaluate the efficacy of our antibodies and the applicability of the model system, marmosets were prophylactically treated with two monoclonal antibodies, c7D11 and c8A. Of three marmosets, two were completely free of disease and a single marmoset died 8 days after the mock (n = 1) or PBS control(s) (n = 2). Evaluation of the serum levels of the three animals provided a possible explanation to the animal succumbing to disease. Interestingly, more females had lesions (and a greater number of lesions) and lower viral burden (viremia and oral shedding) than males in our studies, suggesting a possible gender effect.
Collapse
Affiliation(s)
- Eric M. Mucker
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Suzanne E. Wollen-Roberts
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Adrienne Kimmel
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Josh Shamblin
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Darryl Sampey
- BioFactura, Inc, Frederick, Maryland, United States of America
| | - Jay W. Hooper
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| |
Collapse
|
38
|
Bayesian Diallel Analysis Reveals Mx1-Dependent and Mx1-Independent Effects on Response to Influenza A Virus in Mice. G3-GENES GENOMES GENETICS 2018; 8:427-445. [PMID: 29187420 PMCID: PMC5919740 DOI: 10.1534/g3.117.300438] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Influenza A virus (IAV) is a respiratory pathogen that causes substantial morbidity and mortality during both seasonal and pandemic outbreaks. Infection outcomes in unexposed populations are affected by host genetics, but the host genetic architecture is not well understood. Here, we obtain a broad view of how heritable factors affect a mouse model of response to IAV infection using an 8 × 8 diallel of the eight inbred founder strains of the Collaborative Cross (CC). Expanding on a prior statistical framework for modeling treatment response in diallels, we explore how a range of heritable effects modify acute host response to IAV through 4 d postinfection. Heritable effects in aggregate explained ∼57% of the variance in IAV-induced weight loss. Much of this was attributable to a pattern of additive effects that became more prominent through day 4 postinfection and was consistent with previous reports of antiinfluenza myxovirus resistance 1 (Mx1) polymorphisms segregating between these strains; these additive effects largely recapitulated haplotype effects observed at the Mx1 locus in a previous study of the incipient CC, and are also replicated here in a CC recombinant intercross population. Genetic dominance of protective Mx1 haplotypes was observed to differ by subspecies of origin: relative to the domesticus null Mx1 allele, musculus acts dominantly whereas castaneus acts additively. After controlling for Mx1, heritable effects, though less distinct, accounted for ∼34% of the phenotypic variance. Implications for future mapping studies are discussed.
Collapse
|
39
|
Insufficient Innate Immunity Contributes to the Susceptibility of the Castaneous Mouse to Orthopoxvirus Infection. J Virol 2017; 91:JVI.01042-17. [PMID: 28747505 DOI: 10.1128/jvi.01042-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/19/2017] [Indexed: 11/20/2022] Open
Abstract
The castaneous (CAST) mouse, a wild-derived inbred strain, is highly susceptible to orthopoxvirus infection by intranasal and systemic routes. The 50% lethal intraperitoneal dose of vaccinia virus (VACV) was 3 PFU for CAST mice, whereas BALB/c mice survived 106 PFU. At all times and in all organs analyzed, virus titers were higher in CAST than in BALB/c mice. In individual CAST mice, luciferase-expressing VACV was seen to replicate rapidly leading to death, whereas virus levels increased for a few days and then declined in BALB/c mice. Increases in gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α) were delayed and low in CAST mice compared to BALB/c mice following VACV infection or poly(I-C) inoculation, consistent with differences in innate immune responses. In addition, naive CAST mice had considerably lower numbers of NK and T cells than BALB/c mice. The percentage of IFN-γ-producing CD4+ and CD8+ T cells increased following infection of CAST mice only after considerable virus spread, and the absolute cell numbers remained low. Administration of exogenous IFN-γ or -α to CAST mice before or during the first days of infection suppressed virus replication and prolonged survival, allowing the mice to make adaptive CD4+ and CD8+ T cell responses that were necessary to clear the virus after cessation of interferon treatment. Thus, insufficient innate cytokine and cellular immune responses contribute to the unique susceptibility of CAST mice to VACV, whereas the adaptive immune response can be protective only if virus replication is suppressed during the first several days of infection.IMPORTANCE Most inbred mouse strains are relatively resistant to orthopoxviruses. The castaneous (CAST) mouse is a notable exception, exhibiting extreme vulnerability to monkeypox virus, cowpox virus, and vaccinia virus and thus providing a unique model for studying pathogenicity, immunity, vaccines, and antiviral drugs. To fully utilize the CAST mouse for such purposes, it is necessary to understand the basis for virus susceptibility. We showed that naive CAST mice make low IFN-γ and TNF-α responses and have low levels of NK cells and CD4+ and CD8+ T cells compared to a resistant classical inbred mouse strain. Attenuating virus replication with one or more doses of exogenous IFN-α or -γ before or during the first few days of infection enabled the development of adaptive cellular immunity and clearance of virus. Further genetic studies may reveal the basis for the low innate immunity.
Collapse
|
40
|
Arriero E, Wanelik KM, Birtles RJ, Bradley JE, Jackson JA, Paterson S, Begon M. From the animal house to the field: Are there consistent individual differences in immunological profile in wild populations of field voles (Microtus agrestis)? PLoS One 2017; 12:e0183450. [PMID: 28817724 PMCID: PMC5560671 DOI: 10.1371/journal.pone.0183450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/06/2017] [Indexed: 12/20/2022] Open
Abstract
Inbred mouse strains, living in simple laboratory environments far removed from nature, have been shown to vary consistently in their immune response. However, wildlife populations are typically outbreeding and face a multiplicity of challenges, parasitological and otherwise. In this study we seek evidence of consistent difference in immunological profile amongst individuals in the wild. We apply a novel method in this context, using longitudinal (repeated capture) data from natural populations of field voles, Microtus agrestis, on a range of life history and infection metrics, and on gene expression levels. We focus on three immune genes, IFN-γ, Gata3, and IL-10, representing respectively the Th1, Th2 and regulatory elements of the immune response. Our results show that there was clear evidence of consistent differences between individuals in their typical level of expression of at least one immune gene, and at most all three immune genes, after other measured sources of variation had been taken into account. Furthermore, individuals that responded to changing circumstances by increasing expression levels of Gata3 had a correlated increase in expression levels of IFN-γ. Our work stresses the importance of acknowledging immunological variation amongst individuals in studies of parasitological and infectious disease risk in wildlife populations.
Collapse
Affiliation(s)
- Elena Arriero
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.,Department of Zoology and Physical Anthropology, University Complutense of Madrid, Madrid, Spain
| | - Klara M Wanelik
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Richard J Birtles
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Janette E Bradley
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Joseph A Jackson
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Steve Paterson
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Mike Begon
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Meiotic Consequences of Genetic Divergence Across the Murine Pseudoautosomal Region. Genetics 2017; 205:1089-1100. [PMID: 28100589 PMCID: PMC5340325 DOI: 10.1534/genetics.116.189092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023] Open
Abstract
The production of haploid gametes during meiosis is dependent on the homology-driven processes of pairing, synapsis, and recombination. On the mammalian heterogametic sex chromosomes, these key meiotic activities are confined to the pseudoautosomal region (PAR), a short region of near-perfect sequence homology between the X and Y chromosomes. Despite its established importance for meiosis, the PAR is rapidly evolving, raising the question of how proper X/Y segregation is buffered against the accumulation of homology-disrupting mutations. Here, I investigate the interplay of PAR evolution and function in two interfertile house mouse subspecies characterized by structurally divergent PARs, Mus musculus domesticus and M. m. castaneus. Using cytogenetic methods to visualize the sex chromosomes at meiosis, I show that intersubspecific F1 hybrids harbor an increased frequency of pachytene spermatocytes with unsynapsed sex chromosomes. This high rate of asynapsis is due, in part, to the premature release of synaptic associations prior to completion of prophase I. Further, I show that when sex chromosomes do synapse in intersubspecific hybrids, recombination is reduced across the paired region. Together, these meiotic defects afflict ∼50% of spermatocytes from F1 hybrids and lead to increased apoptosis in meiotically dividing cells. Despite flagrant disruption of the meiotic program, a subset of spermatocytes complete meiosis and intersubspecific F1 males remain fertile. These findings cast light on the meiotic constraints that shape sex chromosome evolution and offer initial clues to resolve the paradox raised by the rapid evolution of this functionally significant locus.
Collapse
|
42
|
Sergeev AA, Kabanov AS, Bulychev LE, Sergeev AA, Pyankov OV, Bodnev SA, Galahova DO, Zamedyanskaya AS, Titova KA, Glotov AG, Taranov OS, Omigov VV, Shishkina LN, Agafonov AP, Sergeev AN. The Possibility of Using the ICR Mouse as an Animal Model to Assess Antimonkeypox Drug Efficacy. Transbound Emerg Dis 2016; 63:e419-30. [PMID: 25597343 DOI: 10.1111/tbed.12323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Indexed: 12/30/2022]
Abstract
As a result of the conducted experimental studies on intranasal challenge of ICR mice, rabbits and miniature pigs (even in the maximum variant) with the doses of 4.0-5.5 lg PFU of monkeypox virus (MPXV), some clinical signs such as purulent conjunctivitis, blepharitis and ruffled fur were found only in mice. The 50% infective dose (C ID50 ) of MPXV for these animals estimated by the presence of external clinical signs was 4.8 lg PFU, and L ID50 estimated by the virus presence in the lungs of mice 7 days post-infection taking into account its 10% application in the animal respiratory tract was 1.4 lg PFU. When studying the dynamics of MPXV propagation in mice challenged intranasally with 25 L ID50 of MPXV, the maximum pathogen accumulation was revealed in nasal cavity, lungs and brain: 5.7 ± 0.1, 5.5 ± 0.1 and 5.3 ± 0.3 lg PFU/ml, respectively. The pathomorphological examination of these animals revealed the presence and replication of the pathogen in the traditional primary target cells for MPXV (mononuclear phagocyte system cells and respiratory tract epitheliocytes) as well as in some other types of cells (endothelial cells, reticular cells, connective tissue cells). Our use of these animals to assess the antiviral efficacy of some drugs demonstrated the agreement of the results (a significant positive effect of NIOCH-14 and ST-246) with those described in scientific literature, which opens up the prospects of using ICR mice as animal models for monkeypox to develop preventive antismallpox drugs.
Collapse
Affiliation(s)
- Al A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Kabanov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L E Bulychev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - Ar A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - O V Pyankov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - S A Bodnev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - D O Galahova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Zamedyanskaya
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - K A Titova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A G Glotov
- State Scientific Establishment - Institute of Experimental Veterinary Science of Siberia and the Far East Russian Academy of Agricultural Sciences, Krasnoobsk, Russia
| | - O S Taranov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - V V Omigov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L N Shishkina
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A P Agafonov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A N Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| |
Collapse
|
43
|
Johnson RF, Hammoud DA, Perry DL, Solomon J, Moore IN, Lackemeyer MG, Bohannon JK, Sayre PJ, Minai M, Papaneri AB, Hagen KR, Janosko KB, Jett C, Cooper K, Blaney JE, Jahrling PB. Exposure of rhesus monkeys to cowpox virus Brighton Red by large-particle aerosol droplets results in an upper respiratory tract disease. J Gen Virol 2016; 97:1942-1954. [PMID: 27166137 PMCID: PMC5764124 DOI: 10.1099/jgv.0.000501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/07/2016] [Indexed: 01/13/2023] Open
Abstract
We previously demonstrated that small-particle (0.5-3.0 µm) aerosol infection of rhesus monkeys (Macaca mulatta) with cowpox virus (CPXV)-Brighton Red (BR) results in fulminant respiratory tract disease characterized by severe lung parenchymal pathology but only limited systemic virus dissemination and limited classic epidermal pox-like lesion development (Johnson et al., 2015). Based on these results, and to further develop CPXV as an improved model of human smallpox, we evaluated a novel large-particle aerosol (7.0-9.0 µm) exposure of rhesus monkeys to CPXV-BR and monitored for respiratory tract disease by serial computed tomography (CT). As expected, the upper respiratory tract and large airways were the major sites of virus-induced pathology following large-particle aerosol exposure. Large-particle aerosol CPXV exposure of rhesus macaques resulted in severe upper airway and large airway pathology with limited systemic dissemination.
Collapse
Affiliation(s)
- Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donna L. Perry
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Jeffrey Solomon
- Clinical Research Directorate/Clinical Monitoring Research Program Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Ian N. Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew G. Lackemeyer
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Jordan K. Bohannon
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Philip J. Sayre
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amy B. Papaneri
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Katie R. Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Krisztina B. Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Catherine Jett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Joseph E. Blaney
- Office of the Scientific Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter B. Jahrling
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
44
|
Leist SR, Pilzner C, van den Brand JMA, Dengler L, Geffers R, Kuiken T, Balling R, Kollmus H, Schughart K. Influenza H3N2 infection of the collaborative cross founder strains reveals highly divergent host responses and identifies a unique phenotype in CAST/EiJ mice. BMC Genomics 2016; 17:143. [PMID: 26921172 PMCID: PMC4769537 DOI: 10.1186/s12864-016-2483-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/17/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Influenza A virus is a zoonotic pathogen that poses a major threat to human and animal health. The severe course of influenza infection is not only influenced by viral virulence factors but also by individual differences in the host response. To determine the extent to which the genetic background can modulate severity of an infection, we studied the host responses to influenza infections in the eight genetically highly diverse Collaborative Cross (CC) founder mouse strains. RESULTS We observed highly divergent host responses between the CC founder strains with respect to survival, body weight loss, hematological parameters in the blood, relative lung weight and viral load. Mouse strain was the main factor with highest effect size on body weight loss after infection, demonstrating that this phenotype was highly heritable. Sex represented another significant main effect, although it was less strong. Analysis of survival rates and mean time to death suggested three groups of susceptibility phenotypes: highly susceptible (A/J, CAST/EiJ, WSB/EiJ), intermediate susceptible (C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ) and highly resistant strains (NZO/HlLtJ, PWK/PhJ). These three susceptibility groups were significantly different with respect to death/survival counts. Viral load was significantly different between susceptible and resistant strains but not between intermediate and highly susceptible strains. CAST/EiJ mice showed a unique phenotype. Despite high viral loads in their lungs, CAST/EiJ mice exhibited low counts of infiltrating granulocytes and showed increased numbers of macrophages in the lung. Histological studies of infected lungs and transcriptome analyses of peripheral blood cells and lungs confirmed an abnormal response in the leukocyte recruitment in CAST/EiJ mice. CONCLUSIONS The eight CC founder strains exhibited a large diversity in their response to influenza infections. Therefore, the CC will represent an ideal mouse genetic reference population to study the influence of genetic variation on the susceptibility and resistance to influenza infections which will be important to understand individual variations of disease severity in humans. The unique phenotype combination in the CAST/EiJ strain resembles human leukocyte adhesion deficiency and may thus represent a new mouse model to understand this and related abnormal immune responses to infections in humans.
Collapse
Affiliation(s)
- Sarah R Leist
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig and University of Veterinary Medicine Hannover, Inhoffenstr.7, D-38124, Braunschweig, Hannover, Germany
| | - Carolin Pilzner
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig and University of Veterinary Medicine Hannover, Inhoffenstr.7, D-38124, Braunschweig, Hannover, Germany
| | | | - Leonie Dengler
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig and University of Veterinary Medicine Hannover, Inhoffenstr.7, D-38124, Braunschweig, Hannover, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig and University of Veterinary Medicine Hannover, Inhoffenstr.7, D-38124, Braunschweig, Hannover, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig and University of Veterinary Medicine Hannover, Inhoffenstr.7, D-38124, Braunschweig, Hannover, Germany. .,University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
45
|
Evasion of the Innate Immune Type I Interferon System by Monkeypox Virus. J Virol 2015; 89:10489-99. [PMID: 26246580 DOI: 10.1128/jvi.00304-15] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/01/2015] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED The vaccinia virus (VACV) E3 protein has been shown to be important for blocking activation of the cellular innate immune system and allowing viral replication to occur unhindered. Mutation or deletion of E3L severely affects viral host range and pathogenesis. While the monkeypox virus (MPXV) genome encodes a homologue of the VACV E3 protein, encoded by the F3L gene, the MPXV gene is predicted to encode a protein with a truncation of 37 N-terminal amino acids. VACV with a genome encoding a similarly truncated E3L protein (VACV-E3LΔ37N) has been shown to be attenuated in mouse models, and infection with VACV-E3LΔ37N has been shown to lead to activation of the host antiviral protein kinase R pathway. In this report, we present data demonstrating that, despite containing a truncated E3 homologue, MPXV phenotypically resembles a wild-type (wt) VACV rather than VACV-E3LΔ37N. Thus, MPXV appears to contain a gene or genes that can suppress the phenotypes associated with an N-terminal truncation in E3. The suppression maps to sequences outside F3L, suggesting that the suppression is extragenic in nature. Thus, MPXV appears to have evolved mechanisms to minimize the effects of partial inactivation of its E3 homologue. IMPORTANCE Poxviruses have evolved to have many mechanisms to evade host antiviral innate immunity; these mechanisms may allow these viruses to cause disease. Within the family of poxviruses, variola virus (which causes smallpox) is the most pathogenic, while monkeypox virus is intermediate in pathogenicity between vaccinia virus and variola virus. Understanding the mechanisms of monkeypox virus innate immune evasion will help us to understand the evolution of poxvirus innate immune evasion capabilities, providing a better understanding of how poxviruses cause disease.
Collapse
|
46
|
Earl PL, Americo JL, Moss B. Genetic studies of the susceptibility of classical and wild-derived inbred mouse strains to monkeypox virus. Virology 2015; 481:161-5. [PMID: 25791934 PMCID: PMC4437815 DOI: 10.1016/j.virol.2015.02.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 02/18/2015] [Accepted: 02/21/2015] [Indexed: 11/24/2022]
Abstract
Previously, we screened 38 inbred mouse strains for susceptibility to monkeypox virus (MPXV) and focused on wild-derived CAST mice because of their extreme vulnerability. Here, we provide further analysis of inbred mouse strains. NZW/Lac and C58 mice exhibited more weight loss than other classical inbred strains but all survived intranasal challenges with 10(4) to 10(6)PFU of MPXV. Mice from three wild derived strains, in addition to CAST, exhibited severe weight loss and died or were euthanized. LD50 values for CASA, MOLF and PERA were 100, 6800 and >10(5)PFU, respectively. CASA was inbred independently from the same founders as CAST, whereas MOLF and PERA are genetically and geographically distinct. The MPXV susceptibility of the F1 progeny of CAST and either C57BL/6 or BALB/c indicated that resistance is dominant. Back-crossing the F1 progeny of C57BL/6 and CAST to CAST suggested more than one independent resistant locus.
Collapse
Affiliation(s)
- Patricia L Earl
- Laboratory of Viral Diseases, 33 North Drive, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, Unites States
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, 33 North Drive, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, Unites States
| | - Bernard Moss
- Laboratory of Viral Diseases, 33 North Drive, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, Unites States.
| |
Collapse
|
47
|
Franceschi V, Parker S, Jacca S, Crump RW, Doronin K, Hembrador E, Pompilio D, Tebaldi G, Estep RD, Wong SW, Buller MR, Donofrio G. BoHV-4-Based Vector Single Heterologous Antigen Delivery Protects STAT1(-/-) Mice from Monkeypoxvirus Lethal Challenge. PLoS Negl Trop Dis 2015; 9:e0003850. [PMID: 26086739 PMCID: PMC4473039 DOI: 10.1371/journal.pntd.0003850] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/27/2015] [Indexed: 01/13/2023] Open
Abstract
Monkeypox virus (MPXV) is the etiological agent of human (MPX). It is an emerging orthopoxvirus zoonosis in the tropical rain forest of Africa and is endemic in the Congo-basin and sporadic in West Africa; it remains a tropical neglected disease of persons in impoverished rural areas. Interaction of the human population with wildlife increases human infection with MPX virus (MPXV), and infection from human to human is possible. Smallpox vaccination provides good cross-protection against MPX; however, the vaccination campaign ended in Africa in 1980, meaning that a large proportion of the population is currently unprotected against MPXV infection. Disease control hinges on deterring zoonotic exposure to the virus and, barring that, interrupting person-to-person spread. However, there are no FDA-approved therapies against MPX, and current vaccines are limited due to safety concerns. For this reason, new studies on pathogenesis, prophylaxis and therapeutics are still of great interest, not only for the scientific community but also for the governments concerned that MPXV could be used as a bioterror agent. In the present study, a new vaccination strategy approach based on three recombinant bovine herpesvirus 4 (BoHV-4) vectors, each expressing different MPXV glycoproteins, A29L, M1R and B6R were investigated in terms of protection from a lethal MPXV challenge in STAT1 knockout mice. BoHV-4-A-CMV-A29LgD106ΔTK, BoHV-4-A-EF1α-M1RgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK were successfully constructed by recombineering, and their capacity to express their transgene was demonstrated. A small challenge study was performed, and all three recombinant BoHV-4 appeared safe (no weight-loss or obvious adverse events) following intraperitoneal administration. Further, BoHV-4-A-EF1α-M1RgD106ΔTK alone or in combination with BoHV-4-A-CMV-A29LgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK, was shown to be able to protect, 100% alone and 80% in combination, STAT1(-/-) mice against mortality and morbidity. This work demonstrated the efficacy of BoHV-4 based vectors and the use of BoHV-4 as a vaccine-vector platform.
Collapse
Affiliation(s)
| | - Scott Parker
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Sarah Jacca
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Ryan W. Crump
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Konstantin Doronin
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Edguardo Hembrador
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Daniela Pompilio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Giulia Tebaldi
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Ryan D. Estep
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Scott W. Wong
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Mark R. Buller
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
48
|
Lopera JG, Falendysz EA, Rocke TE, Osorio JE. Attenuation of monkeypox virus by deletion of genomic regions. Virology 2015; 475:129-38. [PMID: 25462353 PMCID: PMC4720520 DOI: 10.1016/j.virol.2014.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/20/2014] [Accepted: 11/03/2014] [Indexed: 01/04/2023]
Abstract
Monkeypox virus (MPXV) is an emerging pathogen from Africa that causes disease similar to smallpox. Two clades with different geographic distributions and virulence have been described. Here, we utilized bioinformatic tools to identify genomic regions in MPXV containing multiple virulence genes and explored their roles in pathogenicity; two selected regions were then deleted singularly or in combination. In vitro and in vivo studies indicated that these regions play a significant role in MPXV replication, tissue spread, and mortality in mice. Interestingly, while deletion of either region led to decreased virulence in mice, one region had no effect on in vitro replication. Deletion of both regions simultaneously also reduced cell culture replication and significantly increased the attenuation in vivo over either single deletion. Attenuated MPXV with genomic deletions present a safe and efficacious tool in the study of MPX pathogenesis and in the identification of genetic factors associated with virulence.
Collapse
Affiliation(s)
- Juan G Lopera
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA.
| | | | - Tonie E Rocke
- National Wildlife Health Center, U.S. Geological Survey, Madison, WI, USA
| | - Jorge E Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
49
|
Earl PL, Americo JL, Cotter CA, Moss B. Comparative live bioluminescence imaging of monkeypox virus dissemination in a wild-derived inbred mouse (Mus musculus castaneus) and outbred African dormouse (Graphiurus kelleni). Virology 2015; 475:150-8. [PMID: 25462355 PMCID: PMC4280325 DOI: 10.1016/j.virol.2014.11.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 11/28/2022]
Abstract
Monkeypox virus belongs to the orthopoxvirus genus, infects rodents and monkeys in Africa, produces a smallpox-like zoonotic disease in humans, and has the potential for global spread and exploitation for bioterrorism. Several small animal models for studying monkeypox virus pathogenesis have been investigated. The African dormouse is a candidate natural host but is outbred and no immunological reagents exist. Although not a natural host, the CAST/EiJ mouse is inbred and animals and reagents are commercially available. We compared the dissemination of monkeypox virus by bioluminescence imaging in CAST/EiJ mice and dormice. In CAST/EiJ mice, intense replication occurred at the intranasal site of inoculation and virus spread rapidly to lungs and abdominal organs, which had a lower virus burden. Compared to CAST/EiJ mice, dormice exhibited a greater variation of virus spread, a slower time course, less replication in the head and chest, and more replication in abdominal organs prior to death.
Collapse
Affiliation(s)
- Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
50
|
Israely T, Melamed S, Achdout H, Erez N, Politi B, Waner T, Lustig S, Paran N. TLR3 and TLR9 agonists improve postexposure vaccination efficacy of live smallpox vaccines. PLoS One 2014; 9:e110545. [PMID: 25350003 PMCID: PMC4211728 DOI: 10.1371/journal.pone.0110545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 09/14/2014] [Indexed: 12/14/2022] Open
Abstract
Eradication of smallpox and discontinuation of the vaccination campaign resulted in an increase in the percentage of unvaccinated individuals, highlighting the need for postexposure efficient countermeasures in case of accidental or deliberate viral release. Intranasal infection of mice with ectromelia virus (ECTV), a model for human smallpox, is curable by vaccination with a high vaccine dose given up to 3 days postexposure. To further extend this protective window and to reduce morbidity, mice were vaccinated postexposure with Vaccinia-Lister, the conventional smallpox vaccine or Modified Vaccinia Ankara, a highly attenuated vaccine in conjunction with TLR3 or TLR9 agonists. We show that co-administration of the TLR3 agonist poly(I:C) even 5 days postexposure conferred protection, avoiding the need to increase the vaccination dose. Efficacious treatments prevented death, ameliorated disease symptoms, reduced viral load and maintained tissue integrity of target organs. Protection was associated with significant elevation of serum IFNα and anti-vaccinia IgM antibodies, modulation of IFNγ response, and balanced activation of NK and T cells. TLR9 agonists (CpG ODNs) were less protective than the TLR3 agonist poly(I:C). We show that activation of type 1 IFN by poly(I:C) and protection is achievable even without co-vaccination, requiring sufficient amount of the viral antigens of the infective agent or the vaccine. This study demonstrated the therapeutic potential of postexposure immune modulation by TLR activation, allowing to alleviate the disease symptoms and to further extend the protective window of postexposure vaccination.
Collapse
Affiliation(s)
- Tomer Israely
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sharon Melamed
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hagit Achdout
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noam Erez
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Boaz Politi
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Trevor Waner
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shlomo Lustig
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Nir Paran
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
- * E-mail:
| |
Collapse
|