Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Volume 29, Number 1—January 2023
CME ACTIVITY - Synopsis

Comprehensive Review of Emergence and Virology of Tickborne Bourbon Virus in the United States

Author affiliation: Butler University, Indianapolis, Indiana, USA (

Cite This Article

Introduction

CME Logo

Medscape CME ACTIVITY

In support of improving patient care, this activity has been planned and implemented by Medscape, LLC and Emerging Infectious Diseases. Medscape, LLC is jointly accredited with commendation by the Accreditation Council for Continuing Medical Education (ACCME), the Accreditation Council for Pharmacy Education (ACPE), and the American Nurses Credentialing Center (ANCC), to provide continuing education for the healthcare team.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1.00 AMA PRA Category 1 Credit(s). Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Successful completion of this CME activity, which includes participation in the evaluation component, enables the participant to earn up to 1.0 MOC points in the American Board of Internal Medicine's (ABIM) Maintenance of Certification (MOC) program. Participants will earn MOC points equivalent to the amount of CME credits claimed for the activity. It is the CME activity provider's responsibility to submit participant completion information to ACCME for the purpose of granting ABIM MOC credit.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 75% minimum passing score and complete the evaluation at https://www.medscape.org/journal/eid; and (4) view/print certificate.

Release date: December 21, 2022; Expiration date: December 21, 2023
Learning Objectives

Upon completion of this activity, participants will be able to:

  • Assess the virology of the Bourbon virus

  • Distinguish the geographic distribution of the Bourbon virus infection in the US

  • Evaluate the transmission of the Bourbon virus

  • Assess the clinical picture of the Bourbon virus infection

CME Editor

Susan Zunino, PhD, Technical Writer/Editor, Emerging Infectious Diseases.

CME Author

Charles P. Vega, MD, Health Sciences Clinical Professor of Family Medicine, University of California, Irvine School of Medicine, Irvine, California. Disclosure: Charles P. Vega, MD, has the following relevant financial relationships: consultant or advisor for GlaxoSmithKline; Johnson & Johnson Pharmaceutical Research & Development, L.L.C.

Authors

Molly K. Roe, BS; Elise R. Huffman, BS; Yara S. Batista, BS; George G. Papadeas, BS; Sydney R. Kastelitz, BS; Anna M. Restivo, BS; and Christopher C. Stobart, PhD.

Top

Abstract

The emergence of SARS-CoV-2 and the worldwide COVID-19 pandemic triggered considerable attention to the emergence and evolution of novel human pathogens. Bourbon virus (BRBV) was first discovered in 2014 in Bourbon County, Kansas, USA. Since its initial discovery, several cases of BRBV infection in humans have been identified in Kansas, Oklahoma, and Missouri. BRBV is classified within the Thogotovirus genus; these negative-strand RNA viruses appear to be transmitted by ticks, and much of their biology remains unknown. In this review, we describe the emergence, virology, geographic range and ecology, and human disease caused by BRBV and discuss potential treatments for active BRBV infections. This virus and other emerging viral pathogens remain key public health concerns and require continued surveillance and study to mitigate human exposure and disease.

The first case of Bourbon virus (BRBV) was identified in June 2014 in Bourbon County, Kansas, USA, after severe febrile illness developed in a previously healthy middle-aged (>50 years of age) man (1). Several days after he removed an engorged tick from his shoulder, nonspecific symptoms of disease appeared. After 3 days of worsening fever, myalgia, arthralgia, and diarrhea, the patient visited his primary care physician and was prescribed doxycycline. The next day, the patient was admitted to the hospital because of dehydration, syncope, and a possible tickborne illness. Doxycycline treatment was continued; however, the patient did not respond, and symptoms continued to progress toward multiorgan failure. Laboratory results revealed progressive leukopenia and thrombocytopenia (which are now considered identifiers of potential BRBV infection). Patient blood samples tested negative for all known regional tickborne diseases. Therefore, a whole blood sample was sent to the US Centers for Disease Control and Prevention (CDC) to test for Heartland virus (HRTV), a similar emerging tickborne virus in the region. The index patient died 11 days after symptom onset.

Initial efforts at CDC to identify the causative agent of disease in this first case revealed heterologous (non-HRTV) viral plaques in plaque reduction neutralization tests performed by using serum from the deceased patient and including a control HRTV strain. Subsequent electron microscopy revealed pleiomorphic viral particles consistent with the family Orthomyxoviridae (1). Phylogenetic analyses revealed a close relationship between the patient’s novel virus and Thogoto and Dhori viruses, placing it within the genus Thogotovirus. Subsequent genetic analyses supported this initial genus classification (2,3). Recently, another novel thogotovirus (Oz virus) was discovered in ticks in Japan; this virus was capable of replicating in mammalian cell lines and is the closest known relative of BRBV (3). BRBV was the first human pathogen in the genus Thogotovirus identified in the Western Hemisphere; Aransas Bay virus, another pathogenic member of this genus, was reported in ticks found in seabird nests in the United States (2). Since its initial identification, >5 human cases of BRBV-associated disease have been reported in the Midwest region of the United States (1,48). Because little is known about BRBV biology and no specific treatments or vaccines are available, further studies of BRBV are needed.

Bourbon Virus Genetics and Replication

Genetics and Classification

BRBV consists of a segmented, ≈10–11-kb, single-stranded negative-sense RNA genome (Figure 1) (2,3). Phylogenetic analysis indicates that BRBV has the greatest similarity to Oz, Dhori, and Batken viruses within the genus Thogotovirus and family Orthomyxoviridae (13). The 6 negative-strand RNA segments of the BRBV genome encode the putative glycoprotein (GP), nucleoprotein (NP), matrix protein (M), and the 3 polymerase subunits PA, PB1, and PB2 (2). Gene expression and genetic organization are consistent with other orthomyxoviruses.

The genus Thogotovirus contains several other emerging viruses: Araguari, Aransas Bay, Dhori (including the subtype Batken), Jos, Oz, Thogoto, and Upolu viruses (3). Most of these species have yet to be accepted by the International Committee on Taxonomy of Viruses. Thogoto, Dhori, and Bourbon viruses are known to cause infectious disease in humans.

Despite BRBV sharing several properties with Thogoto and Dhori viruses, such as dependence on hard ticks for transmission and similar virion structures, BRBV disease pathology remains distinct from those viruses and more closely resembles that of HRTV and severe fever with thrombocytopenia syndrome virus. However, genomic analysis of virus open reading frames (ORFs) revealed that BRBV has genetic identity with Dhori and Oz viruses ranging from ≈59% in the most divergent GP gene to ≈82% in the most conserved PB1 gene (2,3). Consequently, BRBV is classified in the Thogotovirus genus and recognized as a relative of both Oz and Dhori viruses.

BRBV Virion Structure and Replication

BRBV forms a pleomorphic (filamentous or round), ≈100–130-nm enveloped virion that is consistent with virions of other orthomyxoviruses (Figure 2) (1,3). Electron microscopy of BRBV virions shows multiple genomic segments that are likely coated internally with NPs and numerous GP molecules studding the virion surface (1,5). Although replication of BRBV has not yet been directly investigated, BRBV genetic analysis, recent crystallization of the BRBV postfusion GP, and studies of replication of related thogotoviruses provide several clues regarding the replication cycle of this virus.

All thogotoviruses use a single attachment GP to mediate virus attachment to and fusion with the host cell (5,9). The postfusion conformation of BRBV GP was recently crystallized; several structural similarities and distinct differences to previously crystallized Dhori and Thogoto virus GPs were observed (2,5). The BRBV GP is a type III fusion protein related to baculovirus Gp64 and consists of 5 distinct domains that assemble into homotrimers on the virus surface (5,10). The cell receptor for BRBV remains unknown; however, ecologic surveillance and in vitro cell culture studies collectively suggest that BRBV exhibits wide vertebrate and invertebrate species tropism (2,11,12). Electron microscopy and postfusion GP crystal structure suggest that BRBV attaches to host cells through a glycan-like cellular receptor and initiates entry by endocytosis (1,5).

After an endosome has formed, acidification of the endocytic compartment triggers a conformational change in the GP that causes fusion of the viral envelope with the endosomal membrane and release of the genome into the cytoplasm (5,13). All 6 genomic RNA segments are encapsidated by viral NPs, forming viral ribonucleoprotein complexes (14). Intracellular trafficking of viral ribonucleoprotein complexes into the nucleus, the site of replication for thogotoviruses, is driven by viral NPs, which contain a nuclear translocation signal and are known to accumulate in the nucleus during active infection (14,15). Virus replication is induced by heterotrimeric polymerase complexes formed from PB1, PB2, and PA protein subunits and is believed to be consistent with mechanisms described for other orthomyxoviruses (16). Similar to influenza virus and other orthomyxoviruses, thogotoviruses depend on host RNA polymerase II activity and a unique cap-snatching mechanism, whereby the viral polymerase complex cleaves the 5′-methylated cap of cellular mRNA and uses this capped leader sequence to prime viral mRNA transcription (1618). Little is known about the nuclear export pathways of newly formed viral ribonucleoprotein complexes, but both M and NP have been implicated in aiding this process (15,19). Thogotovirus assembly and release appears to occur at the plasma membrane, activated by pH-dependent oligomerization of M particles in the cytoplasm (1,19).

Vectors, Hosts, and Geographic Range of BRBV

Detection and Distribution of BRBV in Invertebrates

To date, all known human cases of BRBV infection have been found in 3 US states, Kansas, Oklahoma, and Missouri (Figure 3). In each case, recent tick bites were associated with the onset of disease (1,8). Since the initial identification of BRBV in a tick-infected person, several studies have used PCR-based surveillance testing to show that BRBV can be detected in all life stages (larvae, nymph, and adult) of lone star ticks (Amblyomma americanum) (4,20). Lone star ticks (also known as northeastern water ticks or turkey ticks) are a species of hard tick with a wide range throughout the eastern and central United States (Figures 3, 4); they are commonly found in both wooded and grassy areas and known to harbor several human pathogens, including HRTV and Tacaribe virus and the bacteria Ehrlichia spp., Francisella tularensis, Coxiella burnetti, and Rickettsia amblyommii (21). Although other common species of ticks have been tested at surveillance sites, such as Amblyomma maculatum, Dermacentor variabilis, Haemaphysalis leporispalustris, Ixodes scapularis, and Ixodes dentatus, to date, BRBV has only been found in A. americanum ticks (4,20). Recently, A. americanum ticks were shown to be capable of sustaining and transmitting BRBV through cofeeding on animal hosts (22). Although the A. americanum tick remains the only vector to harbor BRBV thus far, in vitro studies using cell culture have shown wide species tropism of BRBV. Multi-logarithmic BRBV replication was shown in the vertebrate cell lines Vero, Vero E6, LLC-MK2, BHK21Cl-15, HeLa, and HUH-7 and tick cell lines RAE/CTVM1, HAE/CTVM9, and AVL/CTVM17, indicating further surveillance will be necessary to detect additional invertebrate hosts (2).

Detection of BRBV in Nonhuman Vertebrates

Similar to many other tickborne viruses, BRBV is believed to be transmitted to and amplified in nonhuman vertebrate hosts. Serologic testing was performed on a wide array of common mammal and avian fauna found near the sites of confirmed human cases in Missouri and at a distant site in North Carolina (still within the range of A. americanum ticks); numerous mammals were seropositive for BRBV, including domestic dogs, eastern cottontail rabbits, horses, raccoons, and white-tailed deer (11,12). The 2 most common seropositive animals identified were raccoons and white-tailed deer. No evidence of prior infections was observed in any tested bird species, suggesting that common nonhuman mammals likely serve as potential BRBV amplifier hosts (12).

Bourbon Virus Disease in Humans and Potential Treatments

Five confirmed cases of BRBV infection have been reported in humans (5,7), and all 5 cases are believed to have been caused by tick bites. Although A. americanum ticks remain the only confirmed competent vector for BRBV, no tick species identification was made in those human cases (22). Limited data are available on BRBV disease in humans; however, initial symptoms of infection appear ≈2–7 days postexposure (tick bite) and include weakness, nausea, myalgia, arthralgia, fatigue, and diarrhea (1,7,8). Concurrently or shortly after the onset of initial symptoms, a fever and papular rash developed in all described cases. Laboratory testing of blood samples from infected persons showed consistent evidence of thrombocytopenia, leukopenia, lymphopenia, and elevated levels of aspartate transferase and alanine transferase (1,8). Late-stage BRBV disease is associated with shock, cardiac dysregulation, and pleural effusions (1,7,8). In confirmed fatal cases, time from initial symptoms to death was ≈11–24 days. In postmortem analysis of the index case, acute bone marrow suppression was noted (1).

The pathogenesis of BRBV in humans remains largely unknown. However, studies using footpad or intraperitoneal BRBV inoculations in type I interferon receptor deficient Ifnar1−/− mice showed the virus caused active viremia and lethal systemic infection; the highest viral loads were detected in the liver and spleen, and lower viral loads were detected in the blood, kidneys, and heart (8,23). Pathogenesis observed in those mice was consistent with progressive infection from the initial entry site to multiple organs, including the liver (supported by altered aspartate transferase and alanine transferase levels), lungs (pleural effusions), and heart (cardiac dysregulation). Efforts to establish infection or lethal disease in wild-type mice were largely unsuccessful (8,23). BRBV is highly sensitive to type I and II interferons, suggesting that advanced human disease and death might be caused, in part, by existing weaknesses in antiviral innate host immunity (23).

Because of the low incidence rate of BBRV infection and similarities to other tickborne diseases, additional cases of BRBV disease have likely been either misidentified or unreported. All cases to date have been confirmed at CDC by using PCR (4,24). No established treatment for BRBV disease has been reported other than supportive care. Studies in Ifnar1−/− mice indicate that early introduction of several known antiviral treatments might be effective, including interferon-α or the viral replication inhibitors ribavirin, favipiravir, or myricetin (8,23,25). Recently, a reporter system for BRBV was developed, which will enable more efficient screening of putative inhibitors of this virus (25).

Future Outlook

During the past 3 years, the emergence and pandemic spread of SARS-CoV-2 has highlighted the potential for evolution and proliferation of new pathogens. Thus far, BRBV has remained limited to a small number of confirmed human cases. However, many unanswered questions persist that are related to both virology and ecology of BRBV. Most of what is currently known about BRBV originates from 2 published confirmed cases of disease or ecologic surveillance studies in a small number of states in the US Midwest or North Carolina. Therefore, a substantial need remains to determine mechanisms of viral replication, detect other potential vector hosts, and conduct additional surveillance in unexplored regions within the geographic range of A. americanum ticks.

The A. americanum tick is the only known vector responsible for BRBV spread to humans. The expanding range of A. americanum ticks, partly driven by climate change, might lead to more exposure events (26). Monitoring trends in tickborne and mosquitoborne diseases has become more prominent in recent literature (27). Proactive safety and awareness of tickborne diseases has been encouraged, especially because ticks such as A. americanum continue to be dominant health threats in much of the forested regions of the United States (21). Because of the lack of knowledge and established treatments or vaccines for BRBV, CDC recommends using insect repellent, wearing long sleeves and pants, and conducting a thorough tick check after spending time in known tick-infested regions (28).

Recent evidence suggests increasing potential for BRBV genetic evolution through recombination with related thogotoviruses. The recent discovery in Japan of Oz virus, which exhibits high sequence identity to BRBV, in Amblyomma sp. ticks, which share the same genus as the lone star tick, illustrates the necessity for further examination of thogotoviruses and their geographic distribution (3). To survive in its human host, BRBV must first overcome the interferon-induced myxovirus resistance protein A (MxA) to avoid the host’s innate antiviral defense system. Thogotoviruses are normally susceptible to inactivation by MxA, but a recent study of Jos virus showed that mutations in the viral NP can lead to resistance (14). Although this position is conserved among viruses in the Thogotovirus genus, only 1 amino acid change in the viral NP was required to fully escape MxA without replicative fitness loss. As described previously, BRBV is particularly sensitive to interferon signaling, which suggests a possible therapeutic agent for active BRBV infections (23). However, those studies indicate that thogotoviruses, including BRBV, might find novel mechanisms to evade host interferon-stimulated gene expression.

A substantial need exists for further research on other possible tick vectors of BRBV and the role of amplifier hosts. Seropositivity of several common animals and the ability of BRBV to replicate in multiple animal and tick cells in vitro collectively highlight the need for more BRBV surveillance to mitigate human exposure and disease. Increases in population dynamics, climate change, and vectors mean that vectorborne pathogens such as BRBV remain a major public health concern. More surveillance of both viruses and vectors will elucidate the potential for increased transmission to and pathogenicity in humans.

Addendum

As of 2022, Bourbon virus has now been detected in the Asian longhorned tick (Haemaphysalis longicornis) through a surveillance study performed in Virginia, USA (29). This recent finding provides evidence of possible transmission of Bourbon virus in 2 separate species of ticks, A. americanum and H. longicornis. In addition, that study expands the known region of Bourbon virus to include Virginia.

Top

Acknowledgment

This work was supported by an Indiana Academy of Sciences Senior Research Grant (C.C.S.), a Holcomb Awards Committee Grant of Butler University (C.C.S.), and the Butler University Department of Biological Sciences (C.C.S.).

Top

References

  1. Kosoy  OI, Lambert  AJ, Hawkinson  DJ, Pastula  DM, Goldsmith  CS, Hunt  DC, et al. Novel thogotovirus associated with febrile illness and death, United States, 2014. Emerg Infect Dis. 2015;21:7604. DOIPubMedGoogle Scholar
  2. Lambert  AJ, Velez  JO, Brault  AC, Calvert  AE, Bell-Sakyi  L, Bosco-Lauth  AM, et al. Molecular, serological and in vitro culture-based characterization of Bourbon virus, a newly described human pathogen of the genus Thogotovirus. J Clin Virol. 2015;73:12732. DOIPubMedGoogle Scholar
  3. Ejiri  H, Lim  C-K, Isawa  H, Fujita  R, Murota  K, Sato  T, et al. Characterization of a novel thogotovirus isolated from Amblyomma testudinarium ticks in Ehime, Japan: A significant phylogenetic relationship to Bourbon virus. Virus Res. 2018;249:5765. DOIPubMedGoogle Scholar
  4. Savage  HM, Burkhalter  KL, Godsey  MS Jr, Panella  NA, Ashley  DC, Nicholson  WL, et al. Bourbon virus in field-collected ticks, Missouri, USA. Emerg Infect Dis. 2017;23:201722. DOIPubMedGoogle Scholar
  5. Bai  C, Qi  J, Wu  Y, Wang  X, Gao  GF, Peng  R, et al. Postfusion structure of human-infecting Bourbon virus envelope glycoprotein. J Struct Biol. 2019;208:99106. DOIPubMedGoogle Scholar
  6. Schweon  SJ. Bourbon virus: A novel pathogen. Nursing. 2016;46:65. DOIPubMedGoogle Scholar
  7. Miller  M. Bourbon virus linked to death of Park official—CDC testing tick samples. March 8, 2018 [cited 2021 Oct 31]. The Missourian. https://www.emissourian.com/local_news/county/bourbon-virus-linked-to-death-of-park-official—cdc-testing-tick-samples/article_057f0f0a-bddd-5f7a-aa0d-735187e4c379.html
  8. Bricker  TL, Shafiuddin  M, Gounder  AP, Janowski  AB, Zhao  G, Williams  GD, et al. Therapeutic efficacy of favipiravir against Bourbon virus in mice. PLoS Pathog. 2019;15:e1007790. DOIPubMedGoogle Scholar
  9. Portela  A, Jones  LD, Nuttall  P. Identification of viral structural polypeptides of Thogoto virus (a tick-borne orthomyxo-like virus) and functions associated with the glycoprotein. J Gen Virol. 1992;73:282330. DOIPubMedGoogle Scholar
  10. Peng  R, Zhang  S, Cui  Y, Shi  Y, Gao  GF, Qi  J. Structures of human-infecting Thogotovirus fusogens support a common ancestor with insect baculovirus. Proc Natl Acad Sci U S A. 2017;114:E890512. DOIPubMedGoogle Scholar
  11. Komar  N, Hamby  N, Palamar  MB, Staples  JE, Williams  C. Indirect evidence of Bourbon virus (Thogotovirus, Orthomyxoviridae) infection in North Carolina. N C Med J. 2020;81:2145. DOIPubMedGoogle Scholar
  12. Jackson  KC, Gidlewski  T, Root  JJ, Bosco-Lauth  AM, Lash  RR, Harmon  JR, et al. Bourbon virus in wild and domestic animals, Missouri, USA, 2012–2013. Emerg Infect Dis. 2019;25:17523. DOIPubMedGoogle Scholar
  13. Relich  RF, Grabowski  JM. Tick-borne viruses of North America. Clin Microbiol Newsl. 2020;42:7986. DOIGoogle Scholar
  14. Fuchs  J, Oschwald  A, Graf  L, Kochs  G. Tick-transmitted thogotovirus gains high virulence by a single MxA escape mutation in the viral nucleoprotein. PLoS Pathog. 2020;16:e1009038. DOIPubMedGoogle Scholar
  15. Cros  JF, Palese  P. Trafficking of viral genomic RNA into and out of the nucleus: influenza, Thogoto and Borna disease viruses. Virus Res. 2003;95:312. DOIPubMedGoogle Scholar
  16. Guilligay  D, Kadlec  J, Crépin  T, Lunardi  T, Bouvier  D, Kochs  G, et al. Comparative structural and functional analysis of orthomyxovirus polymerase cap-snatching domains. PLoS One. 2014;9:e84973. DOIPubMedGoogle Scholar
  17. Leahy  MB, Dessens  JT, Nuttall  PA. In vitro polymerase activity of Thogoto virus: evidence for a unique cap-snatching mechanism in a tick-borne orthomyxovirus. J Virol. 1997;71:834751. DOIPubMedGoogle Scholar
  18. Siebler  J, Haller  O, Kochs  G. Thogoto and Dhori virus replication is blocked by inhibitors of cellular polymerase II activity but does not cause shutoff of host cell protein synthesis. Arch Virol. 1996;141:158794. DOIPubMedGoogle Scholar
  19. Yang  M, Feng  F, Liu  Y, Wang  H, Yang  Z, Hou  W, et al. pH-dependent conformational changes of a Thogoto virus matrix protein reveal mechanisms of viral assembly and uncoating. J Gen Virol. 2016;97:214956. DOIPubMedGoogle Scholar
  20. Savage  HM, Godsey  MS Jr, Panella  NA, Burkhalter  KL, Manford  J, Trevino-Garrison  IC, et al. Surveillance for tick-borne viruses near the location of a fatal human case of Bourbon virus (family Orthomyxoviridae: genus Thogotovirus) in eastern Kansas, 2015. J Med Entomol. 2018;55:7015. DOIPubMedGoogle Scholar
  21. Kennedy  AC, Marshall  E; BCE1. BCE1. Lone star ticks (Amblyomma americanum): an emerging threat in Delaware. Del J Public Health. 2021;7:6671. DOIPubMedGoogle Scholar
  22. Godsey  MS Jr, Rose  D, Burkhalter  KL, Breuner  N, Bosco-Lauth  AM, Kosoy  OI, et al. Experimental infection of Amblyomma americanum (Acari: Ixodidae) with Bourbon virus (Orthomyxoviridae: Thogotovirus). J Med Entomol. 2021;58:8739. DOIPubMedGoogle Scholar
  23. Fuchs  J, Straub  T, Seidl  M, Kochs  G. Essential role of interferon response in containing human pathogenic Bourbon virus. Emerg Infect Dis. 2019;25:130413. DOIPubMedGoogle Scholar
  24. Warang  A, Zhang  M, Zhang  S, Shen  Z. A panel of real-time PCR assays for the detection of Bourbon virus, Heartland virus, West Nile virus, and Trypanosoma cruzi in major disease-transmitting vectors. J Vet Diagn Invest. 2021;33:111522. DOIPubMedGoogle Scholar
  25. Hao  S, Ning  K, Wang  X, Wang  J, Cheng  F, Ganaie  SS, et al. Establishment of a replicon reporter of the emerging tick-borne Bourbon virus and use it for evaluation of antivirals. Front Microbiol. 2020;11:572631. DOIPubMedGoogle Scholar
  26. Ma  D, Lun  X, Li  C, Zhou  R, Zhao  Z, Wang  J, et al. Predicting the potential global distribution of Amblyomma americanum (Acari: Ixodidae) under near current and future climatic conditions, using the maximum entropy model. Biology (Basel). 2021;10:1057. DOIPubMedGoogle Scholar
  27. Rosenberg  R, Lindsey  NP, Fischer  M, Gregory  CJ, Hinckley  AF, Mead  PS, et al. Vital signs: trends in reported vectorborne disease cases—United States and Territories, 2004–2016. MMWR Morb Mortal Wkly Rep. 2018;67:496501. DOIPubMedGoogle Scholar
  28. Centers for Disease Control and Prevention. Bourbon virus [cited 2021 Nov 8]. https://www.cdc.gov/bourbon-virus/index.html
  29. Cumbie  AN, Trimble  RN, Eastwood  G. Pathogen spillover to an invasive tick species: first detection of Bourbon virus in Haemaphysalis longicornis in the United States. Pathogens. 2022;11:454.

Top

Figures

Top

Follow Up

Earning CME Credit

To obtain credit, you should first read the journal article. After reading the article, you should be able to answer the following, related, multiple-choice questions. To complete the questions (with a minimum 75% passing score) and earn continuing medical education (CME) credit, please go to https://www.medscape.org/journal/eid. Credit cannot be obtained for tests completed on paper, although you may use the worksheet below to keep a record of your answers.

You must be a registered user on http://www.medscape.org. If you are not registered on http://www.medscape.org, please click on the “Register” link on the right hand side of the website.

Only one answer is correct for each question. Once you successfully answer all post-test questions, you will be able to view and/or print your certificate. For questions regarding this activity, contact the accredited provider, CME@medscape.net. For technical assistance, contact CME@medscape.net. American Medical Association’s Physician’s Recognition Award (AMA PRA) credits are accepted in the US as evidence of participation in CME activities. For further information on this award, please go to https://www.ama-assn.org. The AMA has determined that physicians not licensed in the US who participate in this CME activity are eligible for AMA PRA Category 1 Credits™. Through agreements that the AMA has made with agencies in some countries, AMA PRA credit may be acceptable as evidence of participation in CME activities. If you are not licensed in the US, please complete the questions online, print the AMA PRA CME credit certificate, and present it to your national medical association for review.

Article Title: 
Comprehensive Review of Emergence and Virology of Tickborne Bourbon Virus in the United States
CME Questions
  • Which one of the following statements about the Bourbon virus is most accurate?

    • It is a DNA virus

    • It is a member of the Thogotovirus genus

    • It is a member of the orthopoxvirus family

    • It uses multiple glycoproteins for attachment and fusion

  • All US cases of infection with the Bourbon virus have occurred in which states?

    • Kansas, Oklahoma, and Missouri

    • Maine, New Hampshire, and Vermont

    • Virginia, West Virginia, and North Carolina

    • Colorado, Wyoming, and New Mexico

  • Which one of the following statements regarding the transmission of the Bourbon virus is most accurate?

    • The virus is only prevalent during the adult stage of Lone Star ticks

    • Lone Star Ticks harbor the Bourbon virus exclusively

    • The most common animals that are seropositive for Bourbon virus are raccoons and white-tailed deer

    • Birds are an important amplifier of the Bourbon virus infection for humans

  • Which one of the following statements regarding the clinical presentation of the Bourbon virus infection is most accurate?

    • The latency period between tick bite and the onset of symptoms is about 2 weeks

    • Rash is uncommon

    • Leukocytosis is the most common finding on laboratory evaluation

    • Treatment for the Bourbon virus infection currently consists of supportive care

Top

Cite This Article

DOI: 10.3201/eid2901.212295

Original Publication Date: December 21, 2022

1These first authors contributed equally to this article.

Related Links

Table of Contents – Volume 29, Number 1—January 2023

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Christopher C. Stobart, Butler University, 4600 Sunset Ave, Indianapolis, IN 46208, USA

Send To

10000 character(s) remaining.

Top

Page created: November 02, 2022
Page updated: December 21, 2022
Page reviewed: December 21, 2022
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external