1
|
Ahamed MA, Politza AJ, Liu T, Khalid MAU, Zhang H, Guan W. CRISPR-based strategies for sample-to-answer monkeypox detection: current status and emerging opportunities. NANOTECHNOLOGY 2024; 36:042001. [PMID: 39433062 PMCID: PMC11533882 DOI: 10.1088/1361-6528/ad892b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/06/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
The global health threat posed by the Monkeypox virus (Mpox) requires swift, simple, and accurate detection methods for effective management, emphasizing the growing necessity for decentralized point-of-care (POC) diagnostic solutions. The clustered regularly interspaced short palindromic repeats (CRISPR), initially known for its effective nucleic acid detection abilities, presents itself as an attractive diagnostic strategy. CRISPR offers exceptional sensitivity, single-base specificity, and programmability. Here, we reviewed the latest developments in CRISPR-based POC devices and testing strategies for Mpox detection. We explored the crucial role of genetic sequencing in designing crRNA for CRISPR reaction and understanding Mpox transmission and mutations. Additionally, we showed the integration of CRISPR-Cas12 strategy with pre-amplification and amplification-free methods. Our study also focused on the significant role of Cas12 proteins and the effectiveness of Cas12 coupled with recombinase polymerase amplification (RPA) for Mpox detection. We envision the future prospects and challenges, positioning CRISPR-Cas12-based POC devices as a frontrunner in the next generation of molecular biosensing technologies.
Collapse
Affiliation(s)
- Md Ahasan Ahamed
- Department of Electrical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| | - Anthony J Politza
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| | - Tianyi Liu
- Department of Electrical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| | - Muhammad Asad Ullah Khalid
- Department of Electrical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| | - Huanshu Zhang
- Department of Electrical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| | - Weihua Guan
- Department of Electrical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, United States of America
| |
Collapse
|
2
|
Kabir F, Plaisance E, Portman A, Marfo A, Cirrincione K, Silva D, Amadi V, Stringer J, Short L. Mpox Viral Lineage Analysis and Technique Development Using Next-generation Sequencing Approach. J Infect Dis 2024; 229:S163-S171. [PMID: 37968965 DOI: 10.1093/infdis/jiad504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/07/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND In response to Mpox endemic and public health emergency, DCHHS aimed to develop NGS based techniques to streamline Mpox viral clade and lineage analysis. METHODS The Mpox sequencing workflow started with DNA extraction and adapted Illumina's COVIDSeq assay using hMpox primer pools from Yale School of Public Health. Sequencing steps included cDNA amplification, tagmentation, PCR indexing, pooling libraries, sequencing on MiSeq, data analysis, and report generation. The bioinformatic analysis comprised read assembly and consensus sequence mapping to reference genomes and variant identification, and utilized pipelines including Illumina BaseSpace, NextClade, CLC Workbench, Terra.bio for data quality control (QC) and validation. RESULTS In total, 171 mpox samples were sequenced using modified COVIDSeq workflow and QC metrics were assessed for read quality, depth, and coverage. Multiple analysis pipelines identified the West African clade IIb as the only clade during peak Mpox infection from July through October 2022. Analyses also indicated lineage B.1.2 as the dominant variant comprising the majority of Mpox viral genomes (77.7%), implying its geographical distribution in the United States. Viral sequences were uploaded to GISAID EpiPox. CONCLUSIONS We developed NGS workflows to precisely detect and analyze mpox viral clade and lineages aiding in public health genomic surveillance.
Collapse
Affiliation(s)
- Farruk Kabir
- Dallas County Health and Human Services, Dallas, Texas, USA
| | - Erin Plaisance
- Dallas County Health and Human Services, Dallas, Texas, USA
| | | | - Agnes Marfo
- Dallas County Health and Human Services, Dallas, Texas, USA
| | | | - David Silva
- Dallas County Health and Human Services, Dallas, Texas, USA
| | - Victor Amadi
- Dallas County Health and Human Services, Dallas, Texas, USA
| | - Joey Stringer
- Dallas County Health and Human Services, Dallas, Texas, USA
| | - Luke Short
- Dallas County Health and Human Services, Dallas, Texas, USA
| |
Collapse
|
3
|
Bell TM, Facemire P, Bearss JJ, Raymond JL, Chapman J, Zeng X, Shamblin JD, Williams JA, Grosenbach DW, Hruby DE, Damon IK, Goff AJ, Mucker EM. Smallpox lesion characterization in placebo-treated and tecovirimat-treated macaques using traditional and novel methods. PLoS Pathog 2024; 20:e1012007. [PMID: 38386661 PMCID: PMC10883539 DOI: 10.1371/journal.ppat.1012007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Smallpox was the most rampant infectious disease killer of the 20th century, yet much remains unknown about the pathogenesis of the variola virus. Using archived tissue from a study conducted at the Centers for Disease Control and Prevention we characterized pathology in 18 cynomolgus macaques intravenously infected with the Harper strain of variola virus. Six macaques were placebo-treated controls, six were tecovirimat-treated beginning at 2 days post-infection, and six were tecovirimat-treated beginning at 4 days post-infection. All macaques were treated daily until day 17. Archived tissues were interrogated using immunohistochemistry, in situ hybridization, immunofluorescence, and electron microscopy. Gross lesions in three placebo-treated animals that succumbed to infection primarily consisted of cutaneous vesicles, pustules, or crusts with lymphadenopathy. The only gross lesions noted at the conclusion of the study in the three surviving placebo-treated and the Day 4 treated animals consisted of resolving cutaneous pox lesions. No gross lesions attributable to poxviral infection were present in the Day 2 treated macaques. Histologic lesions in three placebo-treated macaques that succumbed to infection consisted of proliferative and necrotizing dermatitis with intracytoplasmic inclusion bodies and lymphoid depletion. The only notable histologic lesion in the Day 4 treated macaques was resolving dermatitis; no notable lesions were seen in the Day 2 treated macaques. Variola virus was detected in all three placebo-treated animals that succumbed to infection prior to the study's conclusion by all utilized methods (IHC, ISH, IFA, EM). None of the three placebo-treated animals that survived to the end of the study nor the animals in the two tecovirimat treatment groups showed evidence of variola virus by these methods. Our findings further characterize variola lesions in the macaque model and describe new molecular methods for variola detection.
Collapse
Affiliation(s)
- Todd M. Bell
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Paul Facemire
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jeremy J. Bearss
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jo Lynne Raymond
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jennifer Chapman
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Xiankun Zeng
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Joshua D. Shamblin
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Janice A. Williams
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | | | - Dennis E. Hruby
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | - Inger K. Damon
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention (CDC) Atlanta, Georgia, United States of America
| | - Arthur J. Goff
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Eric M. Mucker
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| |
Collapse
|
4
|
Ogunleye SC, Akinsulie OC, Aborode AT, Olorunshola MM, Gbore D, Oladoye M, Adesola RO, Gbadegoye JO, Olatoye BJ, Lawal MA, Bakare AB, Adekanye O, Chinyere EC. The re-emergence and transmission of Monkeypox virus in Nigeria: the role of one health. Front Public Health 2024; 11:1334238. [PMID: 38249416 PMCID: PMC10797020 DOI: 10.3389/fpubh.2023.1334238] [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: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024] Open
Abstract
The Monkeypox virus, commonly abbreviated as mpox, is a viral zoonosis that is experiencing a resurgence in prevalence. It is endemic to regions of West and Central Africa that are characterized by dense forested areas. Various measures pertaining to animals, humans, and the environment have been recognized as potential factors and catalysts for the spread of the disease throughout the impacted regions of Africa. This study examines the various factors contributing to the transmission of the virus in Nigeria, with a particular focus on the animal-human and inter-human modes of transmission in rural communities and healthcare facilities. The One Health approach was emphasized as crucial in the prevention and management of this issue. Literature suggests that preventing repeated zoonotic introductions could potentially halt the transmission of the mpox virus from animal to human hosts, leading to a potential decrease in human infections.
Collapse
Affiliation(s)
- Seto C. Ogunleye
- Faculty of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
| | - Olalekan C. Akinsulie
- Department of Veterinary Biochemistry, Nigeria College of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
| | | | - Mercy M. Olorunshola
- Department of Pharmaceutical Microbiology, Pharmacy, University of Ibadan, Ibada, Nigeria
| | - Damilola Gbore
- Faculty of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
| | | | - Ridwan O. Adesola
- Faculty of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
| | - Joy O. Gbadegoye
- Department of Veterinary Biochemistry, Nigeria College of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
- Healthy Africans Platform, Research and Development, Ibada, Nigeria
| | | | - Mariam A. Lawal
- Department of Biochemistry, Department of Biochemistry, Federal University of Agriculture, Abeokuta, Nigeria
| | - Akeem B. Bakare
- Faculty of Veterinary Medicine, University of Ibadan, Ibada, Nigeria
| | | | | |
Collapse
|
5
|
Kumar S. The Overview of Potential Antiviral Bioactive Compounds in Poxviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:331-336. [PMID: 38801588 DOI: 10.1007/978-3-031-57165-7_21] [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
Poxviruses belong to the family of double-stranded DNA viruses, and it is pathogenic for humans and spread worldwide. These viruses cause infections and various diseases in human. So, it is required to develop new drugs for the treatment of smallpox or other poxvirus infections. Very few potential compounds for the treatment of poxvirus such as smallpox, chickenpox, and monkeypox have been reported. Most of the compounds has used as vaccines. Cidofovir is most commonly used as a vaccine for the treatment of poxviruses. There are no phytochemicals reported for the treatment of poxviruses. Very few phytochemicals are under investigation for the treatment of poxviruses.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Chemistry, Sant Kavi Baba Baijnath Government P.G. College Harakh, Barabanki (UP), 225121, India.
- Dr. Rammanohar Lohia Avadh University, Ayodhya, Uttar Pradesh, 224001, India.
| |
Collapse
|
6
|
Chakraborty P, Kumar R, Karn S, Raviya DD, Mondal P. Poxviruses as Agents of Biological Warfare: The Importance of Ensuring Ethical Standards for Research with Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:399-412. [PMID: 38801593 DOI: 10.1007/978-3-031-57165-7_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Historically, biological agents have been used to target various populations. One of the earliest examples could be the catastrophic effect of smallpox in Australia in the eighteenth century (as alleged by some historians). Modern biological techniques can be used to both create or provide protection against various agents of biological warfare. Any microorganism (viruses, bacteria, and fungi) or its toxins can be used as biological agents. Minnesota Department of Health has listed Smallpox (variola major) as a category A bioterrorism agent, even though it has been eradicated in 1980 through an extensive vaccination campaign. Category A agents are considered the highest risk to public health. Laboratory-associated outbreaks of poxviruses could cause unprecedented occupational hazards. Only two WHO-approved BSL-4 facilities in the United States and Russia are allowed to perform research on the variola virus. So, poxviruses present themselves as a classical case of a dual-use dilemma, since research with them can be used for both beneficial and harmful purposes. Although the importance of ethics in scientific research requires no further elaboration, ethical norms assume greater significance during experimentation with poxviruses. In this chapter, we will update the readers on the sensitive nature of conducting research with poxviruses, and how these viruses can be a source of potential biological weapons. Finally, specified ethical guidelines are explored to ensure safe research practices in virology.
Collapse
Affiliation(s)
- Prasenjit Chakraborty
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India.
| | - Randhir Kumar
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Sanjay Karn
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Dharmiben D Raviya
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Priya Mondal
- Laboratory of Cell Biology, National Cancer Institute, National Institute of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
7
|
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
|
8
|
Human Monkeypox Experience in a Tertiary Level Hospital in Milan, Italy, between May and October 2022: Epidemiological Features and Clinical Characteristics. Viruses 2023; 15:v15030667. [PMID: 36992376 PMCID: PMC10051371 DOI: 10.3390/v15030667] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/17/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Background: Monkeypox virus (mpxv) started to spread to Europe and North America at the beginning of the current outbreak in May 2022, and the World Health Organization (WHO) declared Human Monkeypox (mpox) as a public health emergency of international concern (PHEIC) in July 2022. The aim of this observational analysis is to describe demographical data, symptoms presentation and clinical course till outcome of individuals diagnosed with mpox, between May and October 2022, at our open-access Sexual Health Clinic in IRCCS San Raffaele Hospital in Milan, Italy. Methods: Among people who accessed our Sexual Health Clinic, we considered, as suspected diagnosis of mpox, individuals with consistent symptoms and epidemiological criteria. Following the physical examination, oropharyngeal, anal, genital and cutaneous swabs, plus plasma, urine and seminal fluid were collected as biological materials to detect mpxv DNA. We also performed a screening for sexually transmitted infections (STIs). Results: Overall, 140 individuals with mpox were included in this study. Median age was 37 (interquartile, IQR 33, 43) years old. Males were 137 (98%) and men who have sex with men (MSM) were 134 (96%). As risk factors, we detected travels abroad in 35 (25%) individuals and close contact with mpox cases in 49 (35%). There were 66 (47%) people living with HIV (PLWH). Most frequent symptoms were fever (59%), lymphadenopathy (57%), cutaneous (77%), genital (42%), anal (34%) and oral (26%) lesions, proctitis (39%), sore throat (22%) and generalized rash (5%). At mpox diagnosis, we also observed N. gonorrhoeae in 18 (13%) cases, syphilis in 14 (10%) and C. trachomatis in 12 (9%). Two (1%) people received a concomitant diagnosis of HIV infection. We attended to 21 (15%) complications, with nine (6%) cases of hospitalization including six (IQR 3,7) median hospital days. Forty-five (32%) patients were treated with non-steroidal anti-inflammatory drugs (NSAIDs), 37 (26%) with antibiotics and eight (6%) with antiviral drugs. Conclusions: Similarly to other international cohorts, sexual transmission was most frequently present, and concomitant STIs were common. Symptoms were heterogenous, self-resolving and responsive to therapy. Hospitalization was necessary in few patients. There is uncertainty about the future development of mpox and further studies (e.g., potential disease reservoirs, other possible means of transmission, predictors of severe disease) are still needed.
Collapse
|
9
|
Current Insights into Diagnosis, Prevention Strategies, Treatment, Therapeutic Targets, and Challenges of Monkeypox (Mpox) Infections in Human Populations. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010249. [PMID: 36676198 PMCID: PMC9863601 DOI: 10.3390/life13010249] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
In the wake of the emergence and worldwide respread of a viral infection called Monkeypox (Mpox), there is a serious threat to the health and safety of the global population. This viral infection was endemic to the western and central parts of Africa, but has recently spread out of this endemic area to various countries, including the United Kingdom (UK), Portugal, Spain, the United States of America (USA), Canada, Sweden, Belgium, Italy, Australia, Germany, France, the Netherlands, Israel, and Mexico. This is a timely review focusing on recent findings and developments in the epidemiology, clinical features, therapeutic targets, diagnosis, prevention mechanisms, research challenges and possible treatment for Mpox. To date (29 November 2022), there have been around 81,225 reported cases of Mpox. In most cases, this illness is mild; however, there is a fatality rate ranging from 1 to 10%, which might be increased due to associated complications and/or secondary infections. There is a real challenge in the diagnosis of Mpox, since its symptoms are very similar to those of other infections, including smallpox and chickenpox. Generally, to prevent/limit the risk and transmission of Mpox, the detection and isolation of infected individuals, as well as hand hygiene and cleanliness, are essential and effective approaches to control/combat this viral infection. Nevertheless, updated information about Mpox from different angles is lacking. Thus, this review provides updated and comprehensive information about the Mpox illness, which should highlight the global burden, pathogenicity, symptoms, diagnosis, prevention measures and possible treatment of this emerging disease.
Collapse
|
10
|
Level of Knowledge Regarding Mpox among Peruvian Physicians during the 2022 Outbreak: A Cross-Sectional Study. Vaccines (Basel) 2023; 11:vaccines11010167. [PMID: 36680012 PMCID: PMC9860588 DOI: 10.3390/vaccines11010167] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Introduction: Due to the high incidence of mpox in Peru and the poor knowledge about this disease among healthcare workers in non-endemic countries, it is crucial to determine the knowledge status of Peruvian physicians. Methodology: We conducted an analytical cross-sectional study based on an online survey from August to September 2022. Physicians who had a medical license and lived and practiced medicine in Peru were included. To evaluate the factors associated with a higher level of knowledge, we used crude (cPR) and adjusted (aPR) prevalence ratios with 95% confidence intervals (95% CI) using Poisson regression. Results: We included 463 physicians. The mean age was 36.6 (SD: 10.3) years, and most were male (58.1%). Regarding knowledge, the median knowledge score was 14 [IQR: 13 to 15] out of 17 points. In terms of knowledge gaps, only 60.7% of the participants knew that there was an FDA-approved vaccine for mpox, 49.0% of participants knew about mpox proctitis and 33.3% acknowledged that it could be transmitted by the bite of an infected rodent. We found that taking care of patients with mpox (aPR: 1.39; 95% CI: 1.13 to 1.72) was associated with higher knowledge (>p50), while living in the eastern macro-region (aPR: 0.62; 95% CI: 0.42 to 0.93) was associated with lower knowledge (≤p50). Conclusions: Our study showed a high level of knowledge about mpox among Peruvian physicians. However, educational campaigns may be necessary, especially for physicians from the eastern region and those who do not have clinical experience with mpox.
Collapse
|
11
|
Mitjà O, Ogoina D, Titanji BK, Galvan C, Muyembe JJ, Marks M, Orkin CM. Monkeypox. Lancet 2023; 401:60-74. [PMID: 36403582 PMCID: PMC9671644 DOI: 10.1016/s0140-6736(22)02075-x] [Citation(s) in RCA: 191] [Impact Index Per Article: 191.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/19/2022]
Abstract
Monkeypox is a zoonotic illness caused by the monkeypox virus, an Orthopoxvirus in the same genus as the variola, vaccinia, and cowpox viruses. Since the detection of the first human case in the Democratic Republic of the Congo in 1970, the disease has caused sporadic infections and outbreaks, mainly restricted to some countries in west and central Africa. In July, 2022, WHO declared monkeypox a Public Health Emergency of International Concern, on account of the unprecedented global spread of the disease outside previously endemic countries in Africa and the need for global solidarity to address this previously neglected disease. The 2022 outbreak has been primarily associated with close intimate contact (including sexual activity) and most cases have been diagnosed among men who have sex with men, who often present with novel epidemiological and clinical characteristics. In the 2022 outbreak, the incubation period ranges from 7 days to 10 days and most patients present with a systemic illness that includes fever and myalgia and a characteristic rash, with papules that evolve to vesicles, pustules, and crusts in the genital, anal, or oral regions and often involve the mucosa. Complications that require medical treatment (eg, antiviral therapy, antibacterials, and pain control) occur in up to 40% of patients and include rectal pain, odynophagia, penile oedema, and skin and anorectal abscesses. Most patients have a self-limited illness; between 1% and 13% require hospital admission (for treatment or isolation), and the case-fatality rate is less than 0·1%. A diagnosis can be made through the presence of Orthopoxvirus DNA in PCRs from lesion swabs or body fluids. Patients with severe manifestations and people at risk of severe disease (eg, immunosuppressed people) could benefit from antiviral treatment (eg, tecovirimat). The current strategy for post-exposure prophylaxis or pre-exposure prophylaxis for people at high risk is vaccination with the non-replicating modified vaccinia Ankara. Antiviral treatment and vaccines are not yet available in endemic countries in Africa.
Collapse
Affiliation(s)
- Oriol Mitjà
- Skin Neglected Tropical Diseases and Sexually Transmitted Infections section, Hospital Universitari Germans Trías i Pujol, Badalona, Spain; Fight Infectious Diseases Foundation, Badalona, Spain; School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, Papua New Guinea.
| | - Dimie Ogoina
- Department of Internal Medicine, Infectious Diseases Unit, Niger Delta University and Niger Delta University Teaching Hospital, Bayelsa, Nigeria
| | - Boghuma K Titanji
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA; Medecins du Cameroun (Medcamer), Yaoundé, Cameroon
| | | | - Jean-Jacques Muyembe
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Michael Marks
- London School of Hygiene & Tropical Medicine, London, UK; Hospital for Tropical Diseases, University College London Hospital, London, UK; Division of Infection and Immunology, University College London, London, UK
| | - Chloe M Orkin
- Centre for Immunobiology, Blizard Institute, Queen Mary University, London, UK
| |
Collapse
|
12
|
Cheng K, Zhou Y, Wu H. Bibliometric analysis of global research trends on monkeypox: Are we ready to face this challenge? J Med Virol 2023; 95:e27892. [PMID: 35644836 DOI: 10.1002/jmv.27892] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 01/11/2023]
Affiliation(s)
- Kunming Cheng
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhou
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Haiyang Wu
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| |
Collapse
|
13
|
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
|
14
|
Ahsendorf HP, Diesterbeck US, Hotop SK, Winkler M, Brönstrup M, Czerny CP. Characterisation of an Anti-Vaccinia Virus F13 Single Chain Fragment Variable from a Human Anti-Vaccinia Virus-Specific Recombinant Immunoglobulin Library. Viruses 2022; 14:v14020197. [PMID: 35215792 PMCID: PMC8879190 DOI: 10.3390/v14020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/30/2022] Open
Abstract
Vaccinia virus (VACV) belongs to the genus Orthopoxvirus of the family Poxviridae. There are four different forms of infectious virus particles: intracellular mature virus (IMV), intracellular en-veloped virus (IEV), cell-associated enveloped virus (CEV) and extracellular enveloped virus (EEV). The F13 protein occupies the inner side of the CEV- and EEV-membranes and the outer side of the IEV-membranes. It plays an important role in wrapping progress and EEV production. We constructed a human single-chain fragment variable (scFv) library with a diversity of ≥4 × 108 independent colonies using peripheral blood from four vaccinated donors. One anti-F13 scFv was isolated and characterised after three rounds of panning. In Western blotting assays, the scFv 3E2 reacted with the recombinant F13VACV protein with a reduction of binding under denatured and reduced conditions. Two antigenic binding sites (139-GSIHTIKTLGVYSDY-153 and 169-AFNSAKNSWLNL-188) of scFv 3E2 were mapped using a cellulose membrane encompassing 372 15-mere peptides with 12 overlaps covering the whole F13 protein. No neutralisation capa-bilities were observed either in the presence or absence of complement. In conclusion, the con-struction of recombinant immunoglobulin libraries is a promising strategy to isolate specific scFvs to enable the study of the host-pathogen interaction.
Collapse
Affiliation(s)
- Henrike P. Ahsendorf
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
| | - Ulrike S. Diesterbeck
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
- Correspondence:
| | - Sven-Kevin Hotop
- Helmholtz Centre for Infection Research, Inhoffenstraβe 7, 38124 Braunschweig, Germany; (S.-K.H.); (M.B.)
| | - Michael Winkler
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Mark Brönstrup
- Helmholtz Centre for Infection Research, Inhoffenstraβe 7, 38124 Braunschweig, Germany; (S.-K.H.); (M.B.)
| | - Claus-Peter Czerny
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
| |
Collapse
|
15
|
Oliveira M, Mason-Buck G, Ballard D, Branicki W, Amorim A. Biowarfare, bioterrorism and biocrime: A historical overview on microbial harmful applications. Forensic Sci Int 2020; 314:110366. [PMID: 32683271 PMCID: PMC7305902 DOI: 10.1016/j.forsciint.2020.110366] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022]
Abstract
Microbial Forensics is a field that continues to grow in interest and application among the forensic community. This review, divided into two sections, covers several topics associated with this new field. The first section presents a historic overview concerning the use of microorganisms (or its product, i.e. toxins) as harmful biological agents in the context of biological warfare (biowarfare), bioterrorism, and biocrime. Each case is illustrated with the examination of case reports that span from prehistory to the present day. The second part of the manuscript is devoted to the role of MF and highlights the necessity to prepare for the pressing threat of the harmful use of biological agents as weapons. Preventative actions, developments within the field to ensure a timely and effective response and are discussed herein.
Collapse
MESH Headings
- Bacterial Infections
- Biological Warfare/history
- Bioterrorism/history
- Crime/history
- Forensic Sciences
- HIV Infections
- High-Throughput Screening Assays
- History, 15th Century
- History, 16th Century
- History, 17th Century
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- History, Ancient
- History, Medieval
- Humans
- Machine Learning
- Microbiological Techniques
- Toxins, Biological/adverse effects
Collapse
Affiliation(s)
- Manuela Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal.
| | | | - David Ballard
- King's Forensics, King's College London, London, United Kingdom
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology of the Jagiellonian University, Kraków, Poland
| | - António Amorim
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| |
Collapse
|
16
|
Abstract
Poxvirus (PXV) infections are a common cause of cutaneous signs. In France, certain forms of poxvirus are frequent and benign (molluscum contagiosum), while others are rare but potentially serious (cowpox virus [CPXV]). Whereas only smallpox and molluscum contagiosum viruses have a human reservoir and are transmitted between humans, most poxvirus infections are zoonoses having only animal reservoirs. Only a small number of poxviruses are responsible for infection in humans, but the increasing number of new pets, some of which are exotic, coupled with the rapid rise in international travel are creating a greater risk of transmission of zoonotic PXV to new vectors and of spread of these diseases to new regions throughout the world. In France, molluscum contagiosum, orf and milkers' nodule give rise to numerous consultations and are well known to dermatologists. However, dermatologists must also be able to identify other parapoxviruses of similar presentation to orf; thus, CPXV and monkeypox are considered potentially emergent viruses with a high risk of epidemic and spread due to increasing international transport and the loss of the maximum protection against smallpox. Finally, despite its declared eradication, smallpox is currently being monitored because of the potential risk of reintroduction, whether accidentally or deliberately through bioterrorism.
Collapse
Affiliation(s)
- G Bohelay
- Service de dermatologie, Hôpital Avicenne, AP-HP, 125, rue de Stalingrad, 93009 Bobigny, France
| | - T-A Duong
- Service de dermatologie, Hôpital Henri-Mondor, AP-HP, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France.
| |
Collapse
|
17
|
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
|
18
|
Sklenovská N, Van Ranst M. Emergence of Monkeypox as the Most Important Orthopoxvirus Infection in Humans. Front Public Health 2018; 6:241. [PMID: 30234087 PMCID: PMC6131633 DOI: 10.3389/fpubh.2018.00241] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/10/2018] [Indexed: 01/15/2023] Open
Abstract
Monkeypox is an emerging zoonotic disease recognized as the most important orthopoxvirus infection in humans in the smallpox post-eradication era. The clinical presentation of monkeypox is similar to the one of smallpox. The case fatality rate of monkeypox (10%) lies between the case fatality rate of variola major (30%) and variola minor (1%). The disease is endemic in the Democratic Republic of the Congo, but other countries of Central and West Africa either reported cases of monkeypox in humans or circulation in wildlife. The disease was also imported once into the USA. The disease has always been considered rare and self-limiting, however recent sporadic reports suggest otherwise. Unfortunately, the collected data is limited, dispersed and often incomplete. Therefore, the objective of this review is to trace all reported human monkeypox outbreaks and relevant epidemiological information. The frequency and geographical spread of human monkeypox cases have increased in recent years, and there are huge gaps in our understanding of the disease's emergence, epidemiology, and ecology. The monkeypox virus is considered a high threat pathogen causing a disease of public health importance. Therefore, there is an urgent need to focus on building surveillance capacities which will provide valuable information for designing appropriate prevention, preparedness and response activities.
Collapse
Affiliation(s)
- Nikola Sklenovská
- Laboratory of Clinical Virology, Department of Microbiology & Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Marc Van Ranst
- Laboratory of Clinical Virology, Department of Microbiology & Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Dynamics of Pathological and Virological Findings During Experimental Calpox Virus Infection of Common Marmosets (Callithrix jacchus). Viruses 2017; 9:v9120363. [PMID: 29182537 PMCID: PMC5744138 DOI: 10.3390/v9120363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022] Open
Abstract
Experimental intranasal infection of marmosets (Callithrix jacchus) with calpox virus results in fatal disease. Route and dose used for viral inoculation of the test animals mimics the natural transmission of smallpox, thus representing a suitable model to study pathogenesis and to evaluate new vaccines against orthopoxvirus infection. However, the pathogenic mechanisms leading to death are still unclear. Therefore, our study aimed at investigating the kinetics of pathological alterations to clarify the pathogenesis in calpox virus infection. Following intranasal inoculation with two different viral doses, common marmosets were sacrificed on days 3, 5, 7, 10 and 12 post inoculation. Collected tissue was screened using histopathology, immunohistochemistry, transmission electron microscopy, and virological assays. Our data suggest that primary replication took place in nasal and bronchial epithelia followed by secondary replication in submandibular lymph nodes and spleen. Parallel to viremia at day 7, virus was detectable in many organs, mainly located in epithelial cells and macrophages, as well as in endothelial cells. Based on the onset of clinical signs, the histological and ultrastructural lesions and the immunohistochemical distribution pattern of the virus, the incubation period was defined to last 11 days, which resembles human smallpox. In conclusion, the data indicate that the calpox model is highly suitable for studying orthopoxvirus-induced disease.
Collapse
|
20
|
Briguglio I, Loddo R, Laurini E, Fermeglia M, Piras S, Corona P, Giunchedi P, Gavini E, Sanna G, Giliberti G, Ibba C, Farci P, La Colla P, Pricl S, Carta A. Synthesis, cytotoxicity and antiviral evaluation of new series of imidazo[4,5-g]quinoline and pyrido[2,3-g]quinoxalinone derivatives. Eur J Med Chem 2015; 105:63-79. [DOI: 10.1016/j.ejmech.2015.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/01/2015] [Accepted: 10/03/2015] [Indexed: 10/23/2022]
|
21
|
Abstract
Although several biological agents have been recognized as presenting a significant threat to public health if used in a bioterrorist attack, those that are of greatest importance are known as the Category A agents: Bacillus anthracis (anthrax); variola major (smallpox); Yersinia pestis (plague); Francisella tularensis (tularemia); ribonucleic acid viruses (hemorrhagic fevers); and Clostridium botulinum (botulism toxin). In the previous issue, Part I of this review focused on the clinical presentation and treatment of anthrax, plague, and tularemia. In this second part of this 2-part review of these agents, the focus is on the clinical presentation and treatment of smallpox, viral hemorrhagic fevers, and botulism toxin. The utilization of mass prophylaxis to limit the morbidity and mortality associated with all these agents is also discussed along with the role emergency care personnel play in its implementation.
Collapse
|
22
|
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
|
23
|
Postchallenge administration of brincidofovir protects healthy and immune-deficient mice reconstituted with limited numbers of T cells from lethal challenge with IHD-J-Luc vaccinia virus. J Virol 2015; 89:3295-307. [PMID: 25589648 DOI: 10.1128/jvi.03340-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Protection from lethality by postchallenge administration of brincidofovir (BCV, CMX001) was studied in normal and immune-deficient (nude, nu/nu) BALB/c mice infected with vaccinia virus (VACV). Whole-body bioluminescence imaging was used to record total fluxes in the nasal cavity, lungs, spleen, and liver and to enumerate pox lesions on tails of mice infected via the intranasal route with 10(5) PFU of recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve (AUCs) were calculated for individual mice to assess viral loads. A three-dose regimen of 20 mg/kg BCV administered every 48 h starting either on day 1 or day 2 postchallenge protected 100% of mice. Initiating BCV treatment earlier was more efficient in reducing viral loads and in providing protection from pox lesion development. All BCV-treated mice that survived challenge were also protected from rechallenge with IHD-J-Luc or WRvFire VACV without additional treatment. In immune-deficient mice, BCV protected animals from lethality and reduced viral loads while animals were on the drug. Viral recrudescence occurred within 4 to 9 days, and mice succumbed ∼10 to 20 days after treatment termination. Nude mice reconstituted with 10(5) T cells prior to challenge with 10(4) PFU of IHD-J-Luc and treated with BCV postchallenge survived the infection, cleared the virus from all organs, and survived rechallenge with 10(5) PFU of IHD-J-Luc VACV without additional BCV treatment. Together, these data suggest that BCV protects immunocompetent and partially T cell-reconstituted immune-deficient mice from lethality, reduces viral dissemination in organs, prevents pox lesion development, and permits generation of VACV-specific memory. IMPORTANCE Mass vaccination is the primary element of the public health response to a smallpox outbreak. In addition to vaccination, however, antiviral drugs are required for individuals with uncertain exposure status to smallpox or for whom vaccination is contraindicated. Whole-body bioluminescence imaging was used to study the effect of brincidofovir (BCV) in normal and immune-deficient (nu/nu) mice infected with vaccinia virus, a model of smallpox. Postchallenge administration of 20 mg/kg BCV rescued normal and immune-deficient mice partially reconstituted with T cells from lethality and significantly reduced viral loads in organs. All BCV-treated mice that survived infection were protected from rechallenge without additional treatment. In immune-deficient mice, BCV extended survival. The data show that BCV controls viral replication at the site of challenge and reduces viral dissemination to internal organs, thus providing a shield for the developing adaptive immunity that clears the host of virus and builds virus-specific immunological memory.
Collapse
|
24
|
Synthesis and antiviral activity of new phenylimidazopyridines and N-benzylidenequinolinamines derived by molecular simplification of phenylimidazo[4,5-g]quinolines. Eur J Med Chem 2014; 84:8-16. [DOI: 10.1016/j.ejmech.2014.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 06/09/2014] [Accepted: 07/03/2014] [Indexed: 02/03/2023]
|
25
|
|
26
|
Rozelle DK, Filone CM, Dower K, Connor JH. Vaccinia reporter viruses for quantifying viral function at all stages of gene expression. J Vis Exp 2014:51522. [PMID: 24894622 PMCID: PMC4188345 DOI: 10.3791/51522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Poxviruses are a family of double stranded DNA viruses that include active human pathogens such as monkeypox, molluscum contagiousum, and Contagalo virus. The family also includes the smallpox virus, Variola. Due to the complexity of poxvirus replication, many questions still remain regarding their gene expression strategy. In this article we describe the conceptualization and usage of recombinant vaccinia viruses that enable real-time measurement of single and multiple stages of viral gene expression in a high-throughput format. This is enabled through the use of spectrally distinct fluorescent proteins as reporters for each of three stages of viral replication. These viruses provide a high signal-to-noise ratio while retaining stage specific expression patterns, enabling plate-based assays and microscopic observations of virus propagation and replication. These tools have uses for antiviral discovery, studies of the virus-host interaction, and evolutionary biology.
Collapse
Affiliation(s)
- Daniel K Rozelle
- Department of Microbiology, Boston University School of Medicine
| | | | - Ken Dower
- Department of Microbiology, Boston University School of Medicine
| | - John H Connor
- Department of Microbiology, Boston University School of Medicine;
| |
Collapse
|
27
|
|
28
|
Dai P, Wang W, Cao H, Avogadri F, Dai L, Drexler I, Joyce JA, Li XD, Chen Z, Merghoub T, Shuman S, Deng L. Modified vaccinia virus Ankara triggers type I IFN production in murine conventional dendritic cells via a cGAS/STING-mediated cytosolic DNA-sensing pathway. PLoS Pathog 2014; 10:e1003989. [PMID: 24743339 PMCID: PMC3990710 DOI: 10.1371/journal.ppat.1003989] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 01/26/2014] [Indexed: 11/23/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is an attenuated poxvirus that has been engineered as a vaccine against infectious agents and cancers. Our goal is to understand how MVA modulates innate immunity in dendritic cells (DCs), which can provide insights to vaccine design. In this study, using murine bone marrow-derived dendritic cells, we assessed type I interferon (IFN) gene induction and protein secretion in response to MVA infection. We report that MVA infection elicits the production of type I IFN in murine conventional dendritic cells (cDCs), but not in plasmacytoid dendritic cells (pDCs). Transcription factors IRF3 (IFN regulatory factor 3) and IRF7, and the positive feedback loop mediated by IFNAR1 (IFN alpha/beta receptor 1), are required for the induction. MVA induction of type I IFN is fully dependent on STING (stimulator of IFN genes) and the newly discovered cytosolic DNA sensor cGAS (cyclic guanosine monophosphate-adenosine monophosphate synthase). MVA infection of cDCs triggers phosphorylation of TBK1 (Tank-binding kinase 1) and IRF3, which is abolished in the absence of cGAS and STING. Furthermore, intravenous delivery of MVA induces type I IFN in wild-type mice, but not in mice lacking STING or IRF3. Treatment of cDCs with inhibitors of endosomal and lysosomal acidification or the lysosomal enzyme Cathepsin B attenuated MVA-induced type I IFN production, indicating that lysosomal enzymatic processing of virions is important for MVA sensing. Taken together, our results demonstrate a critical role of the cGAS/STING-mediated cytosolic DNA-sensing pathway for type I IFN induction in cDCs by MVA. We present evidence that vaccinia virulence factors E3 and N1 inhibit the activation of IRF3 and the induction of IFNB gene in MVA-infected cDCs.
Collapse
Affiliation(s)
- Peihong Dai
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Weiyi Wang
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Hua Cao
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Francesca Avogadri
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Lianpan Dai
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ingo Drexler
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Johanna A. Joyce
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Xiao-Dong Li
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhijian Chen
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Taha Merghoub
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Stewart Shuman
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Liang Deng
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
29
|
Umesha K, Sarojini BK, Darshan Raj CG, Bhanuprakash V, Yogisharadhya R, Raghavendra R, Khan MTH. In vitro and in silico biological studies of novel thiazolo[3,2-a]pyrimidine-6-carboxylate derivatives. Med Chem Res 2014. [DOI: 10.1007/s00044-013-0606-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Lloyd-Smith JO. Vacated niches, competitive release and the community ecology of pathogen eradication. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120150. [PMID: 23798698 PMCID: PMC3720048 DOI: 10.1098/rstb.2012.0150] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A recurring theme in the epidemiological literature on disease eradication is that each pathogen occupies an ecological niche, and eradication of one pathogen leaves a vacant niche that favours the emergence of new pathogens to replace it. However, eminent figures have rejected this view unequivocally, stating that there is no basis to fear pathogen replacement and even that pathogen niches do not exist. After exploring the roots of this controversy, I propose resolutions to disputed issues by drawing on broader ecological theory, and advance a new consensus based on robust mechanistic principles. I argue that pathogen eradication (and cessation of vaccination) leads to a 'vacated niche', which could be re-invaded by the original pathogen if introduced. Consequences for other pathogens will vary, with the crucial mechanisms being competitive release, whereby the decline of one species allows its competitors to perform better, and evolutionary adaptation. Hence, eradication can cause a quantitative rise in the incidence of another infection, but whether this leads to emergence as an endemic pathogen depends on additional factors. I focus on the case study of human monkeypox and its rise following smallpox eradication, but also survey how these ideas apply to other pathogens and discuss implications for eradication policy.
Collapse
Affiliation(s)
- James O Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, 610 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| |
Collapse
|
31
|
Raghavendra R, Mahadevan KM, Satyanarayan ND, Bhanuprakash V, Venkatesan G, Yogisharadhya R. Analgesic, Antibacterial and Antiviral Activities of 2-(5-Alkyl-1,3,4-oxadiazol-2-yl)-3H-benzo[f]chromen-3-ones. Indian J Pharm Sci 2013; 74:367-71. [PMID: 23626395 PMCID: PMC3630735 DOI: 10.4103/0250-474x.107079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/10/2012] [Accepted: 08/21/2012] [Indexed: 11/18/2022] Open
Abstract
A novel series of 2-(5-alkyl-1,3,4-oxadiazol-2-yl)-3H-benzo[f]chromen-3-ones (4a-e) have been evaluated for analgesic, antibacterial and antiviral activities. Analgesic activity was carried out using acetic acid-induced writhing method in Swiss albino male mice. The antibacterial activity was performed against Gram-positive and Gram-negative clinical strains by agar well diffusion method. The in vitro antiviral activity was carried out against camelpox and buffalopox viruses. The analgesic activity exhibited by the compounds 4a, 4c and 4d were found to be more significant compared to the standard. The bacterial activity was determined by the inhibition of growth of the organism by the drugs at different concentrations. All the compounds showed significant activity when compared with the drug ciprofloxacin. The in vitro antiviral activity of the compound 4b tested against camelpox and buffalopox viruses revealed no activity when tested at concentrations of 250 μg. The compound 4b did not alter the titres of both the viruses and the titres remain, respectively, 106.5 TCID50 and 106.74 TCID50 per ml for camelpox vaccine virus and buffalopox vaccine virus. However, the compounds 4a-e showed significant analgesic and antibacterial activities.
Collapse
Affiliation(s)
- R Raghavendra
- Department of Post Graduate Studies and Research in Biochemistry, Kuvempu University, Shankaraghatta, Shimoga-577 451, India
| | | | | | | | | | | |
Collapse
|
32
|
Kumar H, Rani R. Development of biosensors for the detection of biological warfare agents: its issues and challenges. Sci Prog 2013; 96:294-308. [PMID: 24244972 PMCID: PMC10365506 DOI: 10.3184/003685013x13777066241280] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This review discusses current development in biosensors for the detection of biological warfare agents (BWAs). BWAs include bacteria, virus and toxins that are added deliberately into air water and food to spread terrorism and cause disease or death. The rapid and unambiguous detection and identification of BWAs with early warning signals for detecting possible biological attack is a major challenge for government agencies particularly military and health. The detection devices--biosensors--can be classified (according to their physicochemical transducers) into four types: electrochemical, nucleic acid, optical and piezoelectric. Advantages and limitations of biosensors are discussed in this review followed by an assessment of the current state of development of different types of biosensors. The research and development in biosensors for biological warfare agent detection is of great interest for the public as well as for governments.
Collapse
Affiliation(s)
- Harish Kumar
- Department of Chemistry, Ch. Devi Lal University, Sirsa, Haryana 125 055, India.
| | | |
Collapse
|
33
|
He Y, Wang Y, Struble EB, Zhang P, Chowdhury S, Reed JL, Kennedy M, Scott DE, Fisher RW. Epitope mapping by random peptide phage display reveals essential residues for vaccinia extracellular enveloped virion spread. Virol J 2012; 9:217. [PMID: 23006741 PMCID: PMC3495767 DOI: 10.1186/1743-422x-9-217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/14/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A33 is a type II integral membrane protein expressed on the extracellular enveloped form of vaccinia virus (VACV). Passive transfer of A33-directed monoclonal antibodies or vaccination with an A33 subunit vaccine confers protection against lethal poxvirus challenge in animal models. Homologs of A33 are highly conserved among members of the Orthopoxvirus genus and are potential candidates for inclusion in vaccines or assays targeting extracellular enveloped virus activity. One monoclonal antibody directed against VACV A33, MAb-1G10, has been shown to target a conformation-dependent epitope. Interestingly, while it recognizes VACV A33 as well as the corresponding variola homolog, it does not bind to the monkeypox homolog. In this study, we utilized a random phage display library to investigate the epitope recognized by MAb-1G10 that is critical for facilitating cell-to-cell spread of the vaccinia virus. RESULTS By screening with linear or conformational random phage libraries, we found that phages binding to MAb-1G10 display the consensus motif CEPLC, with a disulfide bond formed between two cysteine residues required for MAb-1G10 binding. Although the phage motif contained no linear sequences homologous to VACV A33, structure modeling and analysis suggested that residue D115 is important to form the minimal epitope core. A panel of point mutants expressing the ectodomain of A33 protein was generated and analyzed by either binding assays such as ELISA and immunoprecipitation or a functional assessment by blocking MAb-1G10 mediated comet inhibition in cell culture. CONCLUSIONS These results confirm L118 as a component of the MAb-1G10 binding epitope, and further identify D115 as an essential residue. By defining the minimum conformational structure, as well as the conformational arrangement of a short peptide sequence recognized by MAb-1G10, these results introduce the possibility of designing small molecule mimetics that may interfere with the function of A33 in vivo. This information will also be useful for designing improved assays to evaluate the potency of monoclonal and polyclonal products that target A33 or A33-modulated EV dissemination.
Collapse
Affiliation(s)
- Yong He
- Laboratory of Plasma Derivatives, Division of Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, FDA/CBER/OBRR/DH/LPD, HFM-345, 1401 Rockville Pike, Rockville, MD 20852, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Prichard MN, Kern ER. Orthopoxvirus targets for the development of new antiviral agents. Antiviral Res 2012; 94:111-25. [PMID: 22406470 PMCID: PMC3773844 DOI: 10.1016/j.antiviral.2012.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/10/2012] [Accepted: 02/21/2012] [Indexed: 12/29/2022]
Abstract
Investments in the development of new drugs for orthopoxvirus infections have fostered new avenues of research, provided an improved understanding of orthopoxvirus biology and yielded new therapies that are currently progressing through clinical trials. These broad-based efforts have also resulted in the identification of new inhibitors of orthopoxvirus replication that target many different stages of viral replication cycle. This review will discuss progress in the development of new anti-poxvirus drugs and the identification of new molecular targets that can be exploited for the development of new inhibitors. The prototype of the orthopoxvirus group is vaccinia virus and its replication cycle will be discussed in detail noting specific viral functions and their associated gene products that have the potential to serve as new targets for drug development. Progress that has been achieved in recent years should yield new drugs for the treatment of these infections and might also reveal new approaches for antiviral drug development with other viruses.
Collapse
Affiliation(s)
- Mark N Prichard
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL 35233-1711, United States.
| | | |
Collapse
|
35
|
|
36
|
Effective antiviral treatment of systemic orthopoxvirus disease: ST-246 treatment of prairie dogs infected with monkeypox virus. J Virol 2011; 85:9176-87. [PMID: 21697474 DOI: 10.1128/jvi.02173-10] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smallpox preparedness research has led to development of antiviral therapies for treatment of serious orthopoxvirus infections. Monkeypox virus is an emerging, zoonotic orthopoxvirus which can cause severe and transmissible disease in humans, generating concerns for public health. Monkeypox virus infection results in a systemic, febrile-rash illness closely resembling smallpox. Currently, there are no small-molecule antiviral therapeutics approved to treat orthopoxvirus infections of humans. The prairie dog, using monkeypox virus as a challenge virus, has provided a valuable nonhuman animal model in which monkeypox virus infection closely resembles human systemic orthopoxvirus illness. Here, we assess the efficacy of the antiorthopoxvirus compound ST-246 in prairie dogs against a monkeypox virus challenge of 65 times the 50% lethal dose (LD(50)). Animals were infected intranasally and administered ST-246 for 14 days, beginning on days 0, 3, or after rash onset. Swab and blood samples were collected every 2 days and analyzed for presence of viral DNA by real-time PCR and for viable virus by tissue culture. Seventy-five percent of infected animals that received vehicle alone succumbed to infection. One hundred percent of animals that received ST-246 survived challenge, and animals that received treatment before symptom onset remained largely asymptomatic. Viable virus and viral DNA were undetected or at greatly reduced levels in animals that began treatment on 0 or 3 days postinfection, compared to control animals or animals treated post-rash onset. Animals treated after rash onset manifested illness, but all recovered. Our results indicate that ST-246 can be used therapeutically, following onset of rash illness, to treat systemic orthopoxvirus infections.
Collapse
|
37
|
Tarbet EB, Larson D, Anderson BJ, Bailey KW, Wong MH, Smee DF. Evaluation of imiquimod for topical treatment of vaccinia virus cutaneous infections in immunosuppressed hairless mice. Antiviral Res 2011; 90:126-33. [PMID: 21439326 DOI: 10.1016/j.antiviral.2011.03.181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 03/03/2011] [Accepted: 03/14/2011] [Indexed: 11/28/2022]
Abstract
Imiquimod is an immune response modifier prescribed as a topical medication for a number of viral and neoplastic conditions. We evaluated the antiviral activity of imiquimod against vaccinia virus (WR strain) cutaneous infections in immunosuppressed (with cyclophosphamide) hairless mice when administered after virus exposure. Primary lesions progressed in severity, satellite lesions developed, and infection eventually killed the mice. Once daily topical treatment with 1% imiquimod cream for 3, 4, or 5 days were compared to twice daily topical treatment with 1% cidofovir cream for 7 days. Survival time of mice in all treated groups was significantly prolonged compared to placebo controls. The mean day of death for the placebo group, 3-day imiquimod, 4-day imiquimod, 5-day imiquimod, and cidofovir groups were 15.5, 20.0, 20.5, 19.5, and 20.5 days post-infection, respectively. All treatment groups showed significant reductions in primary lesion size and in the number of satellite lesions. The cidofovir and 4-day imiquimod treatments delayed the appearance of lung virus titers by 3 and 6 days, respectively, although cutaneous lesion and snout virus titers were not as affected by treatment. Benefits in survival and lesion reduction were observed when imiquimod treatment was delayed from 24, 48, and 72 h post-infection. However, increasing the treatment dose of imiquimod from 1% to 5% led to a significant decrease in antiviral efficacy. These results demonstrate the protective effects of topically administered imiquimod against a disseminated vaccinia virus infection in this mouse model.
Collapse
Affiliation(s)
- E Bart Tarbet
- Institute for Antiviral Research and Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah 84322-5600, United States.
| | | | | | | | | | | |
Collapse
|
38
|
Moise L, Buller RM, Schriewer J, Lee J, Frey SE, Weiner DB, Martin W, De Groot AS. VennVax, a DNA-prime, peptide-boost multi-T-cell epitope poxvirus vaccine, induces protective immunity against vaccinia infection by T cell response alone. Vaccine 2010; 29:501-11. [PMID: 21055490 DOI: 10.1016/j.vaccine.2010.10.064] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/12/2010] [Accepted: 10/24/2010] [Indexed: 12/12/2022]
Abstract
The potential for smallpox to be disseminated in a bioterror attack has prompted development of new, safer smallpox vaccination strategies. We designed and evaluated immunogenicity and efficacy of a T-cell epitope vaccine based on conserved and antigenic vaccinia/variola sequences, identified using bioinformatics and immunological methods. Vaccination in HLA transgenic mice using a DNA-prime/peptide-boost strategy elicited significant T cell responses to multiple epitopes. No antibody response pre-challenge was observed, neither against whole vaccinia antigens nor vaccine epitope peptides. Remarkably, 100% of vaccinated mice survived lethal vaccinia challenge, demonstrating that protective immunity to vaccinia does not require B cell priming.
Collapse
|
39
|
Thavaselvam D, Vijayaraghavan R. Biological warfare agents. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2010; 2:179-88. [PMID: 21829313 PMCID: PMC3148622 DOI: 10.4103/0975-7406.68499] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 07/26/2010] [Accepted: 07/26/2010] [Indexed: 11/27/2022] Open
Abstract
The recent bioterrorist attacks using anthrax spores have emphasized the need to detect and decontaminate critical facilities in the shortest possible time. There has been a remarkable progress in the detection, protection and decontamination of biological warfare agents as many instrumentation platforms and detection methodologies are developed and commissioned. Even then the threat of biological warfare agents and their use in bioterrorist attacks still remain a leading cause of global concern. Furthermore in the past decade there have been threats due to the emerging new diseases and also the re-emergence of old diseases and development of antimicrobial resistance and spread to new geographical regions. The preparedness against these agents need complete knowledge about the disease, better research and training facilities, diagnostic facilities and improved public health system. This review on the biological warfare agents will provide information on the biological warfare agents, their mode of transmission and spread and also the detection systems available to detect them. In addition the current information on the availability of commercially available and developing technologies against biological warfare agents has also been discussed. The risk that arise due to the use of these agents in warfare or bioterrorism related scenario can be mitigated with the availability of improved detection technologies.
Collapse
|
40
|
De Clercq E. Historical perspectives in the development of antiviral agents against poxviruses. Viruses 2010; 2:1322-1339. [PMID: 21994682 PMCID: PMC3185982 DOI: 10.3390/v2061322] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 05/28/2010] [Accepted: 05/28/2010] [Indexed: 12/02/2022] Open
Abstract
The poxvirus vaccinia virus (VV) served as the model virus for which the first antivirals, the thiosemicarbazones, were identified. This dates back to 1950; and, although there is at present no single antiviral drug specifically licensed for the chemotherapy or -prophylaxis of poxvirus infections, numerous candidate compounds have been described over the past 50 years. These compounds include interferon and inducers thereof (i.e., polyacrylic acid), 5-substituted 2’-deoxyuridines (i.e., idoxuridine), IMP dehydrogenase inhibitors, S-adenosylhomocysteine hydrolase inhibitors, acyclic nucleoside phosphonates (such as cidofovir) and alkoxyalkyl prodrugs thereof (such as CMX001), viral egress inhibitors (such as tecovirimat), and cellular kinase inhibitors (such as imatinib).
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, Department of Microbiology and Immunology, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| |
Collapse
|
41
|
A mouse-based assay for the pre-clinical neurovirulence assessment of vaccinia virus-based smallpox vaccines. Biologicals 2009; 38:278-83. [PMID: 19896867 DOI: 10.1016/j.biologicals.2009.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/18/2009] [Accepted: 09/29/2009] [Indexed: 11/24/2022] Open
Abstract
Post-vaccinal encephalitis, although relatively uncommon, is a known adverse event associated with many live, attenuated smallpox vaccines. Although smallpox vaccination ceased globally in 1980, vaccine manufacture has resumed in response to concerns over the possible use of smallpox virus as an agent of bioterrorism. To better support the production of safer smallpox vaccines, we previously reported the development of a mouse model in which a relatively attenuated vaccine strain (Dryvax) could be discerned from a more virulent laboratory strain (WR). Here we have further tested the performance of this assay by evaluating the neurovirulence of several vaccinia virus-based smallpox vaccines spanning a known range in neurovirulence for humans. Our data indicate that testing of 10-100 pfu of virus in mice following intracranial inoculation reliably assesses the virus's neurovirulence potential for humans.
Collapse
|
42
|
Abstract
It has been demonstrated that HIV-1 gp120 resembles several important properties of immunoglobulins allowing it strong influence on the human immune system, especially through induction of the deceptive imprinting and deregulation of the immune network. On the other hand there are many unanswered questions concerning properties and control of the genetically modified viruses and bacteria used as vectors in AIDS vaccines. This situation opens a serious question about the safety of vectored AIDS vaccine and the ethics of their trials in humans.
Collapse
Affiliation(s)
- Veljko Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Yugoslavia.
| | | | | |
Collapse
|
43
|
Abstract
The use of non-conventional agents aimed at causing panic and terror among civilians has a long history. There have been uninterrupted threats and the use of biological and chemical weaponry from the time of early tribal conflicts to the Iran-Iraq war. The sole practical experience has come from the release of the nerve gas Sarin in a Tokyo subway (1994) and the inhalational anthrax discovered in Florida (2001). Drills that simulate scenarios of biological/chemical mass infestation have yielded valuable theoretical experience. This chapter reviews the main chemical and biological agents possibly obtainable by individuals and groups, and the anaesthesiologist's tasks during the resultant non-conventional multi-casualty scenarios. It briefly illustrates the chemical and biological pathological effects of the various intoxicants on the human body, and describes modes of protection and the currently available treatment, based on both military and civilian materials and on the authors' own experience derived from specially designed drills.
Collapse
|
44
|
Lucey DR, Breman JG, Henderson DA. Smallpox and Bioterrorism. BEYOND ANTHRAX 2009. [PMCID: PMC7120204 DOI: 10.1007/978-1-59745-326-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Abstract
In an earlier report, we provided evidence that expression of CCR5 by primary human T cells renders them permissive for vaccinia virus (VACV) replication. This may represent a mechanism for dissemination throughout the lymphatic system. To test this hypothesis, wild-type CCR5(+/+) and CCR5 null mice were challenged with VACV by intranasal inoculation. In time course studies using different infective doses of VACV, we identified viral replication in the lungs of both CCR5(+/+) and CCR5(-/-) mice, yet there were diminished viral loads in the spleens and brains of CCR5(-/-) mice compared with CCR5(+/+) mice. Moreover, in association with VACV infection, we provide evidence for CD4+ and CD8+ T-cell as well as CD11c+ and F4/80+ cell infiltration into the lungs of CCR5(+/+) but not CCR5(-/-) mice, and we show that the CCR5-expressing T cells harbor virus. We demonstrate that this CCR5 dependence is VACV specific, since CCR5(-/-) mice are as susceptible to intranasal influenza virus (A/WSN/33) infection as CCR5(+/+) mice. In a final series of experiments, we provide evidence that adoptive transfer of CCR5(+/+) bone marrow leukocytes into CCR5(-/-) mice restores VACV permissiveness, with evidence of lung and spleen infection. Taken together, our data suggest a novel role for CCR5 in VACV dissemination in vivo.
Collapse
|
46
|
Artenstein AW, Grabenstein JD. Smallpox vaccines for biodefense: need and feasibility. Expert Rev Vaccines 2008; 7:1225-37. [PMID: 18844596 DOI: 10.1586/14760584.7.8.1225] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Smallpox, eradicated as a cause of natural disease through an intensive global effort in the later part of the 20th Century, has resurfaced as a possible agent of bioterrorism. For this reason, there is renewed interest in smallpox vaccines. Live vaccinia virus, an orthopoxvirus related to smallpox, has a long and successful clinical track record as an effective smallpox vaccine; however, its use is associated with uncommon yet serious adverse events. This has led to a surge of recent research into newer-generation smallpox vaccines with improved safety profiles and retained efficacy. This article will review the history of smallpox vaccines, assess the status of newer-generation vaccines and examine the overall risk-versus-benefit profile of smallpox vaccination.
Collapse
Affiliation(s)
- Andrew W Artenstein
- Department of Medicine, Brown University, Memorial Hospital of RI, 111 Brewster Street, Pawtucket, RI 02860, USA.
| | | |
Collapse
|
47
|
Abstract
SUMMARY Monkeypox is a disease that is endemic in Central and Western Africa. However, in 2003, there was an outbreak in the United States, representing the first documented monkeypox cases in the Western hemisphere. Although monkeypox virus is less fatal and not as transmissible as variola virus, the causative agent of smallpox, there is concern that monkeypox virus could become a more efficient human pathogen. The reason for this may lie in the virus' genetic makeup, ecological changes, changes in host behavior, and the fact that with the eradication of variola virus, routine smallpox vaccination is no longer carried out. In this review, we focus on the viral proteins that are predicted to modulate the host immune response and compare the genome of monkeypox virus with the genomes of variola virus and the vaccinia virus, the orthopoxvirus that represented the smallpox vaccine. There are differences found in several of these immune-modulating genes including genes that express proteins that affect cytokines such as interleukin-1, tumor necrosis factor, and interferon. There are also differences in genes that code for virulence factors and host range proteins. Genetic differences likely also explain the differences in virulence between two strains of monkeypox virus found in two different regions of Africa. In the current setting of limited smallpox vaccination and little orthopoxvirus immunity in parts of the world, monkeypox could become a more efficient human pathogen under the right circumstances.
Collapse
Affiliation(s)
- Jessica R Weaver
- Department of Medicine, Division of Infectious Diseases, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6073, USA
| | | |
Collapse
|
48
|
Smee DF. Progress in the Discovery of Compounds Inhibiting Orthopoxviruses in Animal Models. ACTA ACUST UNITED AC 2008; 19:115-24. [DOI: 10.1177/095632020801900302] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Surrogate animal models must be used for testing antiviral agents against variola (smallpox) virus infections. Once developed, these compounds can be stockpiled for use in the event of a bioterrorist incident involving either variola or monkeypox virus, or used to treat an occasional serious orthopoxvirus infection, such as disseminated vaccinia complication following expo-sure to the live virus vaccine. Recently, considerable progress has been made in the discovery of novel anti-viral agents found active against orthopoxviruses in vivo. This includes the development of new animal models or refinement of existing ones for compound efficacy testing. Current mouse models employ ectromelia, cowpox and vaccinia (WR and IHD strains) viruses with respiratory (lung) or tail lesion infections commonly studied. Rabbitpox and vaccinia (WR strain) viruses are available for rabbit infections. Monkeypox and variola viruses are used for infecting monkeys. This review describes these and other animal models, and covers compounds found active in vivo from 2003 to date. Cidofovir, known to be active against orthopox virus infections prior to 2003, has been studied extensively over recent years. New compounds showing promise are orally active inhibitors of orthopoxvirus infections that include ether lipid prodrugs of cidofovir and ( S)-HPMPA, ST-246, N-meth-anocarbathymidine ( N-MCT) and SRI 21950 (a 4'-thio derivative of iododeoxyuridine). Another compound with high activity but requiring parenteral administration is HPMPO-DAPy. Further development of these compounds is warranted.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
49
|
Goff A, Twenhafel N, Garrison A, Mucker E, Lawler J, Paragas J. In vivo imaging of cidofovir treatment of cowpox virus infection. Virus Res 2007; 128:88-98. [PMID: 17524511 DOI: 10.1016/j.virusres.2007.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 04/07/2007] [Accepted: 04/12/2007] [Indexed: 10/23/2022]
Abstract
Variola virus and other members of the genus Orthopoxviruses constitute a prominent bioterrorism and public health threat. Treatment with the anti-viral drug cidofovir inhibits replication of orthopoxviruses in vitro and in vivo. In this study, we visualized the effect of cidofovir on viral kinetics in orthopoxvirus infected mice by using whole-body fluorescence imaging (FI). We engineered a cowpox virus (CPV) expressing the enhanced green fluorescent protein (GFP). Single-step growth curves and calculated 50% lethal doses (LD(50)) of wild-type CPX (Wt-CPV) and GFP-expressing CPX (GFP-CPV) were comparable. Whole-body FI first detected GFP fluorescence in the mesenteric tissue of untreated animals on post-infection day (PID) 1. On PID 3 GFP signal was detected throughout the mesentery, in all abdominal organs by PID 5 and in most major organs, except for the heart and brain by PID 6. Infected animals treated with 25mg/kg of cidofovir also began showing signs of viral replication on PID 1, however, the fluorescent signal was limited only to discrete foci throughout the course of the infection. This work describes the first use of an established Orthopox model of infection to evaluate drug efficacy and track virus progression on a macroscopic level.
Collapse
Affiliation(s)
- Arthur Goff
- Virology Division, United States Army Medical Research Institute for Infectious Diseases, MD 21702-5011, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Pickup DJ. Understanding orthopoxvirus interference with host immune responses to inform novel vaccine design. Expert Rev Vaccines 2007; 6:87-95. [PMID: 17280481 DOI: 10.1586/14760584.6.1.87] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Jenner's original vaccine used cowpox virus. Cowpox virus and, subsequently, vaccinia virus, a closely related Orthopoxvirus, provided the means to eradicate smallpox. This history and the unique properties of the virus suggest that vaccinia virus will continue to provide a useful vaccine platform. Yet, surprisingly, it has become apparent that much of the virus genome encodes accessory proteins that interfere with host immune responses to infection. Manipulation of these genes offers the potential for new generations of orthopoxvirus vaccines in which we will have far greater control over key features of the vaccination, including the sites of virus infection, the degree of virus replication, the pathogenicity of the virus and, most importantly, the suppression or induction of immune responses of specific types.
Collapse
Affiliation(s)
- David J Pickup
- Department of Molecular Genetics and Microbiology, and Duke Human Vaccine Institute, Box 3020, Duke University Medical Center, Durham, NC 27710 USA.
| |
Collapse
|