Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Pathogens ; 12(3)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36986317

ABSTRACT

Monkeypox virus (MPXV), a member of the Orthopoxvirus (OPXV) genus, is a zoonotic virus, endemic to central and western Africa that can cause smallpox-like symptoms in humans with fatal outcomes in up to 15% of patients. The incidence of MPXV infections in the Democratic Republic of the Congo, where the majority of cases have occurred historically, has been estimated to have increased as much as 20-fold since the end of smallpox vaccination in 1980. Considering the risk global travel carries for future disease outbreaks, accurate epidemiological surveillance of MPXV is warranted as demonstrated by the recent Mpox outbreak, where the majority of cases were occurring in non-endemic areas. Serological differentiation between childhood vaccination and recent infection with MPXV or other OPXVs is difficult due to the high level of conservation within OPXV proteins. Here, a peptide-based serological assay was developed to specifically detect exposure to MPXV. A comparative analysis of immunogenic proteins across human OPXVs identified a large subset of proteins that could potentially be specifically recognized in response to a MPXV infection. Peptides were chosen based upon MPXV sequence specificity and predicted immunogenicity. Peptides individually and combined were screened in an ELISA against serum from well-characterized Mpox outbreaks, vaccinee sera, and smallpox sera collected prior to eradication. One peptide combination was successful with ~86% sensitivity and ~90% specificity. The performance of the assay was assessed against the OPXV IgG ELISA in the context of a serosurvey by retrospectively screening a set of serum specimens from the region in Ghana believed to have harbored the MPXV-infected rodents involved in the 2003 United States outbreak.

2.
Vaccines (Basel) ; 8(3)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698399

ABSTRACT

The protection provided by smallpox vaccines when used after exposure to Orthopoxviruses is poorly understood. Postexposu re administration of 1st generation smallpox vaccines was effective during eradication. However, historical epidemiological reports and animal studies on postexposure vaccination are difficult to extrapolate to today's populations, and 2nd and 3rd generation vaccines, developed after eradication, have not been widely tested in postexposure vaccination scenarios. In addition to concerns about preparedness for a potential malevolent reintroduction of variola virus, humans are becoming increasingly exposed to naturally occurring zoonotic orthopoxviruses and, following these exposures, disease severity is worse in individuals who never received smallpox vaccination. This study investigated whether postexposure vaccination of prairie dogs with 2nd and 3rd generation smallpox vaccines was protective against monkeypox disease in four exposure scenarios. We infected animals with monkeypox virus at doses of 104 pfu (2× LD50) or 106 pfu (170× LD50) and vaccinated the animals with IMVAMUNE® or ACAM2000® either 1 or 3 days after challenge. Our results indicated that postexposure vaccination protected the animals to some degree from the 2× LD50, but not the 170× LD5 challenge. In the 2× LD50 challenge, we also observed that administration of vaccine at 1 day was more effective than administration at 3 days postexposure for IMVAMUNE®, but ACAM2000® was similarly effective at either postexposure vaccination time-point. The effects of postexposure vaccination and correlations with survival of total and neutralizing antibody responses, protein targets, take formation, weight loss, rash burden, and viral DNA are also presented.

3.
Virology ; 544: 55-63, 2020 05.
Article in English | MEDLINE | ID: mdl-32174514

ABSTRACT

Historic observations suggest that survivors of smallpox maintained lifelong immunity and protection to subsequent infection compared to vaccinated individuals. Although protective immunity by vaccination using a related virus (vaccinia virus (VACV) strains) was the key for smallpox eradication, it does not uniformly provide long term, or lifelong protective immunity (Heiner et al., 1971). To determine differences in humoral immune responses, mice were inoculated with VACV either systemically, using intranasal inoculation (IN), or locally by an intradermal (ID) route. We hypothesized that sub-lethal IN infections may mimic systemic or naturally occurring infection and lead to an immunodominance reaction, in contrast to localized ID immunization. The results demonstrated systemic immunization through an IN route led to enhanced adaptive immunity to VACV-expressed protein targets both in magnitude and in diversity when compared to an ID route using a VACV protein microarray. In addition, cytokine responses, assessed using a Luminex® mouse cytokine multiplex kit, following IN infection was greater than that stemming from ID infection. Overall, the results suggest that the route of immunization (or infection) influences antibody responses. The greater magnitude and diversity of response in systemic infection provides indirect evidence for anecdotal observations made during the smallpox era that survivors maintain lifelong protection. These findings also suggest that systemic or disseminated host immune induction may result in a superior response, that may influence the magnitude of, as well as duration of protective responses.


Subject(s)
Immunity, Humoral , Vaccinia virus/immunology , Vaccinia/immunology , Adaptive Immunity , Administration, Intranasal , Animals , Antibodies, Neutralizing , Antibodies, Viral , Injections, Intradermal , Mice , Mice, Inbred BALB C , Neutralization Tests , Vaccinia/virology
4.
Viruses ; 9(10)2017 10 03.
Article in English | MEDLINE | ID: mdl-28972544

ABSTRACT

During 2012, 2013 and 2015, we collected small mammals within 25 km of the town of Boende in Tshuapa Province, the Democratic Republic of the Congo. The prevalence of monkeypox virus (MPXV) in this area is unknown; however, cases of human infection were previously confirmed near these collection sites. Samples were collected from 353 mammals (rodents, shrews, pangolins, elephant shrews, a potamogale, and a hyrax). Some rodents and shrews were captured from houses where human monkeypox cases have recently been identified, but most were trapped in forests and agricultural areas near villages. Real-time PCR and ELISA were used to assess evidence of MPXV infection and other Orthopoxvirus (OPXV) infections in these small mammals. Seven (2.0%) of these animal samples were found to be anti-orthopoxvirus immunoglobulin G (IgG) antibody positive (six rodents: two Funisciurus spp.; one Graphiurus lorraineus; one Cricetomys emini; one Heliosciurus sp.; one Oenomys hypoxanthus, and one elephant shrew Petrodromus tetradactylus); no individuals were found positive in PCR-based assays. These results suggest that a variety of animals can be infected with OPXVs, and that epidemiology studies and educational campaigns should focus on animals that people are regularly contacting, including larger rodents used as protein sources.


Subject(s)
Animals, Wild/virology , Monkeypox virus/isolation & purification , Mpox (monkeypox)/veterinary , Animals , Antibodies, Viral/blood , Democratic Republic of the Congo/epidemiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Mammals/virology , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/transmission , Mpox (monkeypox)/virology , Monkeypox virus/genetics , Monkeypox virus/immunology , Monkeypox virus/pathogenicity , Poxviridae Infections/epidemiology , Poxviridae Infections/immunology , Poxviridae Infections/veterinary , Poxviridae Infections/virology , Prevalence , Real-Time Polymerase Chain Reaction , Risk Factors , Sciuridae/virology , Shrews/virology
5.
Crit Rev Immunol ; 37(2-6): 483-498, 2017.
Article in English | MEDLINE | ID: mdl-29773031

ABSTRACT

The reports in 1993 that naked DNA encoding viral genes conferred protective immunity came as a surprise to most vaccinologists. This review analyses the expanding number of examples where plasmid DNA induces immune responses. Issues such as the type of immunity induced, mechanisms of immune protection, and how DNA vaccines compare with other approaches are emphasized. Additional issues discussed include the likely means by which DNA vaccines induce CTL, how the potency and type of immunity induced can be modified, and whether DNA vaccines represent a practical means of manipulating unwanted immune response occurring during immunoinflammatory diseases. It seems doubtful if DNA vaccines will replace currently effective vaccines, but they may prove useful for prophylactic use against some agents that at present lack an effective vaccine. DNA vaccines promise to be valuable to manipulate the immune response in situations where responses to agents are inappropriate or ineffective.


Subject(s)
Immunogenicity, Vaccine/genetics , Vaccination/methods , Vaccines, DNA/immunology , Vaccinology/trends , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , History, 20th Century , History, 21st Century , Humans , Plasmids/genetics , Plasmids/immunology , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Vaccination/trends , Vaccines, DNA/genetics , Vaccines, DNA/history , Vaccinology/history , Viral Vaccines/genetics , Viral Vaccines/history , Viral Vaccines/immunology
6.
PLoS Negl Trop Dis ; 9(10): e0004013, 2015.
Article in English | MEDLINE | ID: mdl-26517724

ABSTRACT

Monkeypox is a zoonotic disease endemic to central and western Africa, where it is a major public health concern. Although Monkeypox virus (MPXV) and monkeypox disease in humans have been well characterized, little is known about its natural history, or its maintenance in animal populations of sylvatic reservoir(s). In 2003, several species of rodents imported from Ghana were involved in a monkeypox outbreak in the United States with individuals of three African rodent genera (Cricetomys, Graphiurus, Funisciurus) shown to be infected with MPXV. Here, we examine the course of MPXV infection in Cricetomys gambianus (pouched Gambian rats) and this rodent species' competence as a host for the virus. We obtained ten Gambian rats from an introduced colony in Grassy Key, Florida and infected eight of these via scarification with a challenge dose of 4X104 plaque forming units (pfu) from either of the two primary clades of MPXV: Congo Basin (C-MPXV: n = 4) or West African (W-MPXV: n = 4); an additional 2 animals served as PBS controls. Viral shedding and the effect of infection on activity and physiological aspects of the animals were measured. MPXV challenged animals had significantly higher core body temperatures, reduced activity and increased weight loss than PBS controls. Viable virus was found in samples taken from animals in both experimental groups (C-MPXV and W-MPXV) between 3 and 27 days post infection (p.i.) (up to 1X108 pfu/ml), with viral DNA found until day 56 p.i. The results from this work show that Cricetomys gambianus (and by inference, probably the closely related species, Cricetomys emini) can be infected with MPXV and shed viable virus particles; thus suggesting that these animals may be involved in the maintenance of MPXV in wildlife mammalian populations. More research is needed to elucidate the epidemiology of MPXV and the role of Gambian rats and other species.


Subject(s)
Disease Reservoirs , Monkeypox virus/isolation & purification , Mpox (monkeypox)/veterinary , Rodent Diseases/pathology , Rodent Diseases/virology , Rodentia/virology , Animals , Body Temperature , Body Weight , Locomotion , Models, Theoretical , Mpox (monkeypox)/pathology , Mpox (monkeypox)/virology , Virus Shedding
7.
Viruses ; 7(4): 2168-84, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25912718

ABSTRACT

Monkeypox is a zoonotic disease caused by a virus member of the genus Orthopoxvirus and is endemic to Central and Western African countries. Previous work has identified two geographically disjuct clades of monkeypox virus based on the analysis of a few genomes coupled with epidemiological and clinical analyses; however, environmental and geographic causes of this differentiation have not been explored. Here, we expand previous phylogenetic studies by analyzing a larger set of monkeypox virus genomes originating throughout Sub-Saharan Africa to identify possible biogeographic barriers associated with genetic differentiation; and projected ecological niche models onto environmental conditions at three periods in the past to explore the potential role of climate oscillations in the evolution of the two primary clades. Analyses supported the separation of the Congo Basin and West Africa clades; the Congo Basin clade shows much shorter branches, which likely indicate a more recent diversification of isolates within this clade. The area between the Sanaga and Cross Rivers divides the two clades and the Dahomey Gap seems to have also served as a barrier within the West African clade. Contraction of areas with suitable environments for monkeypox virus during the Last Glacial Maximum, suggests that the Congo Basin clade of monkeypox virus experienced a severe bottleneck and has since expanded its geographic range.


Subject(s)
Monkeypox virus/classification , Monkeypox virus/genetics , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/virology , Phylogeography , Africa South of the Sahara/epidemiology , Animals , Cluster Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Ecosystem , Humans , Molecular Sequence Data , Monkeypox virus/isolation & purification , Sequence Analysis, DNA , Sequence Homology
8.
Vet Parasitol ; 210(1-2): 91-7, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25881801

ABSTRACT

Since 2005, black-tailed prairie dogs (Cynomys ludovicianus) have been collected for use as research animals from field sites in Kansas, Colorado, and Texas. In January of 2012, Giardia trophozoites were identified by histology, thin-section electron microscopy, and immunofluorescent staining in the lumen of the small intestine and colon of a prairie dog euthanized because of extreme weight loss. With giardiasis suspected as the cause of weight loss, a survey of Giardia duodenalis in the laboratory colony of prairie dogs was initiated. Direct immunofluorescent testing of feces revealed active shedding of Giardia cysts in 40% (n=60) of animals held in the vivarium. All tested fecal samples (n=29) from animals in another holding facility where the index case originated were PCR positive for G. duodenalis with assemblages A and B identified from sequencing triosephosphate isomerase (tpi), glutamate dehydrogenase (gdh), and ß-giardin (bg) genes. Both assemblages are considered zoonotic, thus the parasites in prairie dogs are potential human pathogens and indicate prairie dogs as a possible wildlife reservoir or the victims of pathogen spill-over. Molecular testing for other protozoan gastrointestinal parasites revealed no Cryptosporidium infections but identified a host-adapted Enterocytozoon bieneusi genotype group.


Subject(s)
Enterocytozoon/isolation & purification , Giardia lamblia/isolation & purification , Giardiasis/veterinary , Microsporidiosis/veterinary , Sciuridae/parasitology , Animals , DNA, Protozoan/genetics , Enterocytozoon/genetics , Feces/parasitology , Fenbendazole/therapeutic use , Giardia lamblia/genetics , Giardiasis/drug therapy , Giardiasis/parasitology , Laboratory Animal Science , Microsporidiosis/parasitology , Phylogeny , Polymerase Chain Reaction , Zoonoses
9.
Virology ; 464-465: 264-273, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108113

ABSTRACT

The eradication of smallpox and the cessation of global vaccination led to the increased prevalence of human infections in Central Africa. Serologic and protein-based diagnostic assay for MPXV detection is difficult due to cross-reactive antibodies that do not differentiate between diverse orthopoxvirus (OPXV) species. A previously characterized monoclonal antibody (mAb 69-126-3-7) against MPXV [1] was retested for cross-reactivity with various OPXVs. The 14.5 kDa band protein that reacted with mAb 69-126-3 was identified to be MPXV A29 protein (homolog of vaccinia virus Copenhagen A27). Amino acid sequence analysis of the MPXV A29 with other OPXV homologs identified four amino acid changes. Peptides corresponding to these regions were designed and evaluated for binding to mAb 69-126-3 by ELISA and BioLayer Interferometry (BLI). Further refinement and truncations mapped the specificity of this antibody to a single amino acid difference in a 30-mer peptide compared to other OPXV homologs. This particular residue is proposed to be essential for heparin binding by VACV A27 protein. Despite this substitution, MPXV A29 bound to heparin with similar affinity to that of VACV A27 protein, suggesting flexibility of this motif for heparin binding. Although binding of mAb 69-126-3-7 to MPXV A29 prevented interaction with heparin, it did not have any effect on the infectivity of MPXV. Characterization of 69-126-3-7 mAb antibody allows for the possibility of the generation of a serological based species-specific detection of OPXVs despite high proteomic homology.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Heparin/immunology , Monkeypox virus/immunology , Mpox (monkeypox)/virology , Viral Proteins/immunology , Amino Acid Sequence , Cross Reactions , Humans , Molecular Sequence Data , Mpox (monkeypox)/immunology , Monkeypox virus/chemistry , Protein Structure, Tertiary , Sequence Alignment , Viral Proteins/chemistry
10.
Emerg Infect Dis ; 20(2): 177-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24447382

ABSTRACT

Although it has been >30 years since the eradication of smallpox, the unearthing of well-preserved tissue material in which the virus may reside has called into question the viability of variola virus decades or centuries after its original occurrence. Experimental data to address the long-term stability and viability of the virus are limited. There are several instances of well-preserved corpses and tissues that have been examined for poxvirus viability and viral DNA. These historical specimens cause concern for potential exposures, and each situation should be approached cautiously and independently with the available information. Nevertheless, these specimens provide information on the history of a major disease and vaccination against it.


Subject(s)
DNA, Viral/isolation & purification , Microbial Viability , Smallpox/prevention & control , Smallpox/virology , Variola virus/physiology , Autopsy , Cadaver , History, 18th Century , History, 19th Century , History, 20th Century , History, Ancient , Humans , Smallpox/history , Smallpox/transmission , Smallpox Vaccine/immunology , Specimen Handling/methods , Vaccination , Vaccinia virus/immunology , Variola virus/isolation & purification , Variola virus/pathogenicity
11.
Vaccine ; 31(45): 5192-201, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-23994378

ABSTRACT

Decades after public health interventions - including pre- and post-exposure vaccination - were used to eradicate smallpox, zoonotic orthopoxvirus outbreaks and the potential threat of a release of variola virus remain public health concerns. Routine prophylactic smallpox vaccination of the public ceased worldwide in 1980, and the adverse event rate associated with the currently licensed live vaccinia virus vaccine makes reinstatement of policies recommending routine pre-exposure vaccination unlikely in the absence of an orthopoxvirus outbreak. Consequently, licensing of safer vaccines and therapeutics that can be used post-orthopoxvirus exposure is necessary to protect the global population from these threats. Variola virus is a solely human pathogen that does not naturally infect any other known animal species. Therefore, the use of surrogate viruses in animal models of orthopoxvirus infection is important for the development of novel vaccines and therapeutics. Major complications involved with the use of surrogate models include both the absence of a model that accurately mimics all aspects of human smallpox disease and a lack of reproducibility across model species. These complications limit our ability to model post-exposure vaccination with newer vaccines for application to human orthopoxvirus outbreaks. This review seeks to (1) summarize conclusions about the efficacy of post-exposure smallpox vaccination from historic epidemiological reports and modern animal studies; (2) identify data gaps in these studies; and (3) summarize the clinical features of orthopoxvirus-associated infections in various animal models to identify those models that are most useful for post-exposure vaccination studies. The ultimate purpose of this review is to provide observations and comments regarding available model systems and data gaps for use in improving post-exposure medical countermeasures against orthopoxviruses.


Subject(s)
Post-Exposure Prophylaxis/methods , Smallpox Vaccine/administration & dosage , Smallpox/pathology , Smallpox/prevention & control , Vaccination/methods , Animals , Disease Models, Animal , Humans
12.
PLoS One ; 8(9): e74816, 2013.
Article in English | MEDLINE | ID: mdl-24040344

ABSTRACT

Monkeypox is a major public health concern in the Congo Basin area, with changing patterns of human case occurrences reported in recent years. Whether this trend results from better surveillance and detection methods, reduced proportions of vaccinated vs. non-vaccinated human populations, or changing environmental conditions remains unclear. Our objective is to examine potential correlations between environment and transmission of monkeypox events in the Congo Basin. We created ecological niche models based on human cases reported in the Congo Basin by the World Health Organization at the end of the smallpox eradication campaign, in relation to remotely-sensed Normalized Difference Vegetation Index datasets from the same time period. These models predicted independent spatial subsets of monkeypox occurrences with high confidence; models were then projected onto parallel environmental datasets for the 2000s to create present-day monkeypox suitability maps. Recent trends in human monkeypox infection are associated with broad environmental changes across the Congo Basin. Our results demonstrate that ecological niche models provide useful tools for identification of areas suitable for transmission, even for poorly-known diseases like monkeypox.


Subject(s)
Geographic Mapping , Models, Biological , Mpox (monkeypox)/transmission , Algorithms , Animals , Area Under Curve , Cameroon/epidemiology , Central African Republic/epidemiology , Congo/epidemiology , Democratic Republic of the Congo/epidemiology , Environment , Epidemiological Monitoring , Gabon/epidemiology , Geographic Information Systems , Geography , Humans , Macaca fascicularis , ROC Curve , Risk , Spatio-Temporal Analysis
13.
Virology ; 443(2): 358-62, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23809939

ABSTRACT

Since the eradication of Smallpox, researchers have attempted to study Orthopoxvirus pathogenesis and immunity in animal models in order to correlate results human smallpox. A solely human pathogen, Orthopoxvirus variola fails to produce authentic smallpox illness in any other animal species tested to date. In 2003, an outbreak in the USA of Orthopoxvirus monkeypox, revealed the susceptibility of the North American black-tailed prairie dog (Cynomys ludovicianus) to infection and fulminate disease. Prairie dogs infected with Orthopoxvirus monkeypox present with a clinical scenario similar to ordinary smallpox, including prodrome, rash, and high mortality. This study examines if Black-tailed prairie dogs can become infected with O. variola and serve as a surrogate model for the study of human smallpox disease. Substantive evidence of infection is found in immunological seroconversion of animals to either intranasal or intradermal challenges with O. variola, but in the absence of overt illness.


Subject(s)
Disease Models, Animal , Orthopoxvirus/pathogenicity , Sciuridae/virology , Smallpox/pathology , Animals , Antibodies, Viral/blood , Female , Humans , Immunity , Male , Orthopoxvirus/genetics , Orthopoxvirus/immunology , Poxviridae Infections/immunology , Poxviridae Infections/pathology , Smallpox/immunology , Smallpox/virology
14.
Emerg Infect Dis ; 19(6): 1002-4, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23735421

ABSTRACT

A wildlife hospital and rehabilitation center in northwestern United States received several big brown bats with necrosuppurative osteomyelitis in multiple joints. Wing and joint tissues were positive by PCR for poxvirus. Thin-section electron microscopy showed poxvirus particles within A-type inclusions. Phylogenetic comparison supports establishment of a new genus of Poxviridae.


Subject(s)
Chiroptera/virology , Poxviridae/classification , Animals , Genome, Viral , Male , Molecular Sequence Data , Phylogeny , Poxviridae/genetics , Poxviridae/isolation & purification , Poxviridae/ultrastructure
15.
Hum Vaccin Immunother ; 9(7): 1489-96, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23571177

ABSTRACT

BACKGROUND: Routine vaccination against smallpox (variola) ceased in the US in 1976. However, in 2002 limited coverage for military personnel and some healthcare workers was reinstituted. In March 2008, ACAM2000® replaced Dryvax® as the vaccine used in the United States against smallpox. Unintentional transfer of vaccinia virus from a vaccination site by autoinoculation or contact transmission, can have significant public health implications. We summarize unintentional virus transfer AEs associated with ACAM2000® since March 2008 and compare with Dryvax®. RESULTS: We identified 309 reports for ACAM2000® with skin or ocular involvement, of which 93 were autoinoculation cases and 20 were contact transmission cases. The rate for reported cases of autoinoculation was 20.6 per 100,000 vaccinations and for contact transmission was 4.4 per 100,000 vaccinations. Eighteen contact transmission cases could be attributed to contact during a sporting activity (45%) or intimate contact (45%). Of the 113 unintentional transfer cases, 6 met the case definition for ocular vaccinia. The most common locations for all autoinoculation and contact cases were arm/elbow/shoulder (35/113; 31%) and face (24/113; 21%). Methods We reviewed 753 reports associated with smallpox in the Vaccine Adverse Event Reporting System and CDC Poxvirus consultation log, reported from March 2008 to August 2010. Reports were classified into categories based upon standard case definitions. CONCLUSION: Overall, unintentional transfer events for ACAM2000® and Dryvax® are similar. We recommend continued efforts to prevent transfer events and continuing education for healthcare providers focused on recognition of vaccinia lesions, proper sample collection, and laboratory testing to confirm diagnosis.


Subject(s)
Product Surveillance, Postmarketing , Smallpox Vaccine/adverse effects , Smallpox/prevention & control , Vaccination/adverse effects , Vaccinia virus/isolation & purification , Vaccinia/transmission , Adolescent , Adult , Child , Child, Preschool , Female , Health Personnel , Humans , Male , Middle Aged , Military Personnel , Smallpox Vaccine/administration & dosage , United States , Vaccinia/epidemiology , Vaccinia/prevention & control , Young Adult
16.
Am J Trop Med Hyg ; 88(5): 982-985, 2013 May.
Article in English | MEDLINE | ID: mdl-23400570

ABSTRACT

Monkeypox is an acute viral infection with a clinical course resembling smallpox. It is endemic in northern and central Democratic Republic of the Congo (DRC), but it is reported only sporadically in neighboring Republic of the Congo (ROC). In October 2009, interethnic violence in northwestern DRC precipitated the movement of refugees across the Ubangi River into ROC. The influx of refugees into ROC heightened concerns about monkeypox in the area, because of the possibility that the virus could be imported, or that incidence could increase caused by food insecurity and over reliance on bush meat. As part of a broad-based campaign to improve health standards in refugee settlement areas, the United Nations International Children's Emergency Fund (UNICEF) sponsored a program of intensive community education that included modules on monkeypox recognition and prevention. In the 6 months immediately following the outreach, 10 suspected cases of monkeypox were reported to health authorities. Laboratory testing confirmed monkeypox virus infection in two individuals, one of whom was part of a cluster of four suspected cases identified retrospectively. Anecdotes collected at the time of case reporting suggest that the outreach campaign contributed to detection of suspected cases of monkeypox.


Subject(s)
Health Education , Monkeypox virus/isolation & purification , Mpox (monkeypox)/diagnosis , Adolescent , Animals , Child , Congo/epidemiology , Female , Humans , Male , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/pathology , Mpox (monkeypox)/virology , Monkeypox virus/classification , Monkeypox virus/genetics , Phylogeny , Sequence Analysis, DNA
17.
PLoS One ; 8(2): e55488, 2013.
Article in English | MEDLINE | ID: mdl-23408990

ABSTRACT

Monkeypox virus (MPXV) is endemic within Africa where it sporadically is reported to cause outbreaks of human disease. In 2003, an outbreak of human MPXV occurred in the US after the importation of infected African rodents. Since the eradication of smallpox (caused by an orthopoxvirus (OPXV) related to MPXV) and cessation of routine smallpox vaccination (with the live OPXV vaccinia), there is an increasing population of people susceptible to OPXV diseases. Previous studies have shown that the prairie dog MPXV model is a functional animal model for the study of systemic human OPXV illness. Studies with this model have demonstrated that infected animals are able to transmit the virus to naive animals through multiple routes of exposure causing subsequent infection, but were not able to prove that infected animals could transmit the virus exclusively via the respiratory route. Herein we used the model system to evaluate the hypothesis that the Congo Basin clade of MPXV is more easily transmitted, via respiratory route, than the West African clade. Using a small number of test animals, we show that transmission of viruses from each of the MPXV clade was minimal via respiratory transmission. However, transmissibility of the Congo Basin clade was slightly greater than West African MXPV clade (16.7% and 0% respectively). Based on these findings, respiratory transmission appears to be less efficient than those of previous studies assessing contact as a mechanism of transmission within the prairie dog MPXV animal model.


Subject(s)
Monkeypox virus/physiology , Mpox (monkeypox)/transmission , Respiratory Tract Infections/virology , Sciuridae/virology , Animals
18.
J Wildl Dis ; 49(1): 125-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23307378

ABSTRACT

The prevalence of orthopoxviruses (OPXV) among wildlife, including monkeypox virus (MPXV), remains largely unknown. Outbreaks of human monkeypox in central Africa have been associated with hunting, butchering, and consuming infected forest animals, primarily rodents and primates. Monkeypox cases have not been reported in east Africa, where human contact with wildlife is more limited. Whether this lack of human disease is due to the absence of MPXV in rodents is unknown. However, testing of wildlife beyond the known geographic distribution of human cases of monkeypox has rarely been conducted, limiting our knowledge of the natural distribution of MPXV and other OPXV. To improve our understanding of the natural distribution of OPXV in Africa and related risks to public health, we conducted a serosurvey of peridomestic rodents (Rattus rattus) in and around traditional dwellings in Kabarole District, Uganda, from May 2008 to July 2008. We tested for OPXV antibody in areas free of human monkeypox. Sera from 41% of the R. rattus individuals sampled reacted to OPXV-specific proteins from multiple, purified OPXV samples, but did not react by enzyme-linked immunosorbent assay. The specific OPXV could not be identified because poxvirus DNA was undetectable in corresponding tissues. We conclude that an OPXV or a similar poxvirus is circulating among wild rodents in Uganda. With the known geographic range of OPXV in rodents now increased, factors that dictate OPXV prevalence and disease will be identified.


Subject(s)
Antibodies, Viral/blood , Monkeypox virus/immunology , Orthopoxvirus/immunology , Poxviridae Infections/veterinary , Rodent Diseases/epidemiology , Animals , Animals, Wild , Female , Humans , Male , Poxviridae Infections/epidemiology , Poxviridae Infections/transmission , Poxviridae Infections/virology , Public Health , Risk Factors , Rodent Diseases/virology , Rodentia , Seroepidemiologic Studies , Uganda/epidemiology , Zoonoses
19.
Emerg Infect Dis ; 19(2): 237-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23347770

ABSTRACT

Identification of human monkeypox cases during 2005 in southern Sudan (now South Sudan) raised several questions about the natural history of monkeypox virus (MPXV) in Africa. The outbreak area, characterized by seasonally dry riverine grasslands, is not identified as environmentally suitable for MPXV transmission. We examined possible origins of this outbreak by performing phylogenetic analysis of genome sequences of MPXV isolates from the outbreak in Sudan and from differing localities. We also compared the environmental suitability of study localities for monkeypox transmission. Phylogenetically, the viruses isolated from Sudan outbreak specimens belong to a clade identified in the Congo Basin. This finding, added to the political instability of the area during the time of the outbreak, supports the hypothesis of importation by infected animals or humans entering Sudan from the Congo Basin, and person-to-person transmission of virus, rather than transmission of indigenous virus from infected animals to humans.


Subject(s)
Disease Outbreaks , Mpox (monkeypox)/virology , Animals , Genes, Viral , Humans , Molecular Typing , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/transmission , Monkeypox virus/classification , Monkeypox virus/genetics , Monkeypox virus/isolation & purification , Phylogeny , Phylogeography , Sequence Analysis, DNA , Sudan/epidemiology
20.
J Virol Methods ; 187(1): 37-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22981983

ABSTRACT

The commercially available Orthopox BioThreat® Alert assay for orthopoxvirus (OPV) detection is piloted. This antibody-based lateral-flow assay labels and captures OPV viral agents to detect their presence. Serial dilutions of cultured Vaccinia virus (VACV) and Monkeypox virus (MPXV) were used to evaluate the sensitivity of the Tetracore assay by visual and quantitative determinations; specificity was assessed using a small but diverse set of diagnostically relevant blinded samples from viral lesions submitted for routine OPV diagnostic testing. The BioThreat® Alert assay reproducibly detected samples at concentrations of 10(7)pfu/ml for VACV and MPXV and positively identified samples containing 10(6)pfu/ml in 4 of 7 independent experiments. The assay correctly identified 9 of 11 OPV clinical samples and had only one false positive when testing 11 non-OPV samples. Results suggest applicability for use of the BioThreat® Alert assay as a rapid screening assay and point of care diagnosis for suspect human monkeypox cases.


Subject(s)
DNA, Viral/analysis , Orthopoxvirus/genetics , Polymerase Chain Reaction , Poxviridae Infections/diagnosis , Biological Assay , Bioterrorism , Humans , Mpox (monkeypox)/diagnosis , Mpox (monkeypox)/virology , Monkeypox virus/genetics , Poxviridae Infections/virology , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...