Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Ethics Resources and Policies
  • About the Journal
    • About CMR
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Clinical Microbiology Reviews
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Ethics Resources and Policies
  • About the Journal
    • About CMR
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Review

Tick-Borne Flaviviruses, with a Focus on Powassan Virus

Gábor Kemenesi, Krisztián Bányai
Gábor Kemenesi
aVirological Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
bInstitute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Krisztián Bányai
cInstitute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/CMR.00106-17
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

SUMMARY

The tick-borne pathogen Powassan virus is a rare cause of encephalitis in North America and the Russian Far East. The number of documented cases described since the discovery of Powassan virus in 1958 may be <150, although detection of cases has increased over the past decade. In the United States, the incidence of Powassan virus infections expanded from the estimated 1 case per year prior to 2005 to 10 cases per year during the subsequent decade. The increased detection rate may be associated with several factors, including enhanced surveillance, the availability of improved laboratory diagnostic methods, the expansion of the vector population, and, perhaps, altered human activities that lead to more exposure. Nonetheless, it remains unclear whether Powassan virus is indeed an emerging threat or if enzootic cycles in nature remain more-or-less stable with periodic fluctuations of host and vector population sizes. Despite the low disease incidence, the approximately 10% to 15% case fatality rate of neuroinvasive Powassan virus infection and the temporary or prolonged sequelae in >50% of survivors make Powassan virus a medical concern requiring the attention of public health authorities and clinicians. The medical importance of Powassan virus justifies more research on developing specific and effective treatments and prevention and control measures.

INTRODUCTION

Ticks are known to host and transmit a variety of viruses, bacteria, and protozoa pathogenic to animals and human beings (1). The medical and veterinary importance of numerous tick-borne viral infections has been recognized for decades. Tick-borne viruses belong to different virus families, including Asfarviridae, Bunyaviridae, Flaviviridae, Orthomyxoviridae, Reoviridae, and Rhabdoviridae (2). The most severe disease manifestations associated with tick-borne viruses include central nervous system (CNS) infections and hemorrhagic symptoms, which may be lethal in a high proportion (up to 20% to 30%) of patients. Tick-borne flaviviruses (TBFVs) comprise a variety of viruses worldwide that are thought to occur predominantly in the temperate zone and subtropical areas of the Northern Hemisphere and less prominently in (sub)tropical areas of Africa and Australia (3).

Powassan virus (POWV) is a neglected tick-borne virus, the only known member of the TBFVs that occurs naturally in North America. Although the number of associated human diseases can be considered low, the reporting system for notifiable diseases in the United States has shown a tendency toward an increase in the annual number of cases over the past decade. The case fatality rate of POWV encephalitis exceeds 30% in some reports, and long-term sequelae are very common (4, 5). The aim of this paper is to give an overview of POWV. The introductory sections focus on the general features of TBFVs, whereas the second part gives a comprehensive view on Powassan virus, with particular attention being given to epidemiology, genetic diversity, ecology, clinical disease, and diagnosis.

TICK-BORNE FLAVIVIRUSES

Taxonomy and ClassificationAt present, the International Committee on Taxonomy of Viruses lists four genera within the family Flaviviridae: Hepacivirus, Pegivirus, Pestivirus, and Flavivirus. The genus Flavivirus includes over 50 virus species (6); mosquitoes and ticks serve as primary vectors for numerous members of the genus, while mammals and birds serve as common primary hosts. Viruses without vertebrate hosts (e.g., Culex flavivirus and Aedes flavivirus, etc.) and viruses without arthropod vectors (e.g., Tamana bat virus) are also known (6–8).

Dual-host flaviviruses can be divided into mosquito/vertebrate and tick/vertebrate viruses (9). Tick-transmitted flaviviruses include the following species and viruses (listed in alphabetical order): Gadgets Gully virus, Kadam virus, Karshi virus, Kyasanur Forest disease virus (KFDV), Langat virus, Louping ill virus (LIV), Meaban virus, Omsk hemorrhagic fever virus (OHFV), Powassan virus (POWV), Royal Farm virus, Saumarez Reef virus, Tick-borne encephalitis virus (TBEV), and Tyuleniy virus.

Antigenic classification of flaviviruses is based on serological cross-reactivity. In this system, TBEV (including Russian spring-summer encephalitis and Central European encephalitis), Langat virus, KFDV, Royal Farm virus, Karshi virus, Negishi virus (a representative strain of LIV), and POWV were found to share considerable antigenic cross-reactivity, a finding based upon which these viruses were classified into the tick-borne encephalitis (TBE) serocomplex (10). Genetic classification, based on genome sequence data and phylogenetic relationships, provided further insights into the relationship among flaviviruses, including TBFVs. TBFVs were classified into two genetic clades, named mammalian and seabird TBFVs, to indicate a difference in the primary vertebrate host (11). Currently, mammalian TBFVs include seven species of known or putative medical importance (KFDV, Karshi virus, Langat virus, LIV, OHFV, POWV, and TBEV) (Table 1), which are distributed over the Northern Hemisphere: TBEV and LIV occur in Europe and Asia, and OHFV, Langat virus, KFDV, and Karshi virus are endemic in Asia, while POWV is endemic across North America and occurs in the Russian Far East (12). In humans, OHFV and KFDV cause hemorrhagic fever, whereas TBEV, LIV, Langat virus, Karshi virus, and POWV cause meningitis and encephalitis. Lesser-known members are those not associated with causing diseases in humans or animals; these include Gadgets Gully virus, Kadam virus, and Royal Farm virus (11).

View this table:
  • View inline
  • View popup
TABLE 1

General features of human-pathogenic TBFVs

Structure and BiologyInfectious flavivirus particles are enveloped, smooth, spherical structures about 50 nm in diameter. The virion contains an electron-dense, 30-nm nucleocapsid which encapsulates the genomic RNA. The nucleocapsid protein (C protein) interacts with viral RNA and forms a less-ordered structure beneath the lipid membrane. This structure is surrounded by a cell-derived lipid membrane and viral E and M proteins, which are embedded via their transmembrane domains into the external lipid layer (13, 14). In the mature virion, the E proteins form homodimers in a head-to-tail orientation, displaying a herringbone pattern typical for all flaviviruses (13). The M protein protrudes at 5-fold symmetry axes between the dimerization domains of E proteins (9).

Members of the Flavivirus genus possess a single-stranded plus-sense RNA genome with an average length of 11 kb (Fig. 1). The GC content is around 52% to 55% (15). The 5ʹ end of genomic RNA has a type I cap structure (m7GpppAmpN2), whereas the 3ʹ end lacks a poly(A) tail and terminates in a stable stem-loop structure (16). The cap directs the process of translation and also protects the viral RNA from degradation by cellular exonucleases (16). The genomic RNA is infectious upon transfection. The coding region of flavivirus genomic RNA encodes a single large polyprotein, composed of roughly 3,300 to 3,400 amino acids (aa) (17, 18). The coding region is flanked by 5ʹ and 3ʹ noncoding regions (NCRs). The lengths of the 5ʹ NCR and 3ʹ NCR are ∼100 nucleotides (nt) and ∼400 to 700 nt, respectively; the 3ʹ NCR may show significant length variation even within closely related TBFV strains (19). The NCR structures contain both sequential and structural motifs and are essential for flavivirus life cycle processes such as translation, replication, and virion assembly (20).

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

(A) Schematic representation of the flavivirus genome and translation of the viral polyprotein. (B) Proteolytic cleavage sites, individual proteins, and localization of nascent proteins in the endoplasmic reticulum. Scissors indicate viral (red) and cellular (green) proteases that are responsible for cleavage.

The structural proteins are encoded at the 5ʹ end of the genomic RNA (Fig. 1). The C protein forms the nucleocapsid; its C-terminal helices have a net positively charged interface and interact with the negatively charged genomic RNA (9). The M protein has two functional forms. In the immature virion, the longer, glycosylated precursor M (prM) protein stabilizes the E protein, preventing the virion from undergoing maturation into the structure that displays fusion activity (21, 22). During virion maturation, furin removes the N-terminal region of prM, leaving the C-terminal region to be anchored into the virion membrane (23). The E protein has multiple roles; it mediates virus attachment to cellular receptors and, consequently, is a major determinant of host and tissue tropism, and it also mediates membrane fusion and specifies immunological properties responsible for eliciting neutralizing antibodies in vivo. It contains three structural domains. Domain I and domain II are responsible for dimerization and contain a highly conserved hydrophobic peptide region that mediates membrane fusion following internalization of the virion. Domain III contains receptor binding sites and the major antigenic sites with predominantly linear epitopes that are the main targets of neutralizing antibodies (24).

The nonstructural proteins are encoded by the central region and at the 3ʹ end of the genomic RNA (Fig. 1). Flavivirus NS proteins are components of the viral replication complex embedded in the endoplasmic reticulum (ER), play a role in flaviviral polyprotein processing, and/or serve as effectors against cellular antiviral defense mechanisms. The glycosylated NS1 protein has multiple forms. Of interest, antibodies raised against NS1 protect mice from virus challenge (25, 26). The multidomain NS3 protein with its N-terminal part functions as a serine protease, playing a role in flaviviral polyprotein processing, whereas the C-terminal part of the protein contains RNA triphosphatase and RNA helicase activities involved in capping and synthesis of viral RNA (27). NS5 consists of an N-terminal methyltransferase domain and the C-terminal RNA-dependent RNA polymerase domain containing both viral RNA capping and RNA synthesis activities. NS5 also plays a role in the evasion of the innate immune response in both mosquito- and tick-borne flaviviruses (28, 29).

The flavivirus replication cycle begins with the attachment of mature virus particles to the surface of the host cell. For TBEV, heparan sulfate has been shown to be involved in cellular binding (30). Additional cellular factors for attachment may be laminin binding protein and dendritic cell-specific intracellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) (31, 32). After budding into the cytoplasm, vesicles containing the virus particles fuse with endosomes, where the acidic pH triggers irreversible structural rearrangement in the E protein layer of the virion and E protein binding to the endosomal membrane. Following the fusion of the virion lipid layer with the endosome membrane, the nucleocapsid is released into the cytoplasm. Subsequently, the nucleocapsid disassembles, releasing the genomic RNA. Virus proteins are translated from the viral RNA on the cytoplasmic surface of the ER membrane as a single large polyprotein (33). Specific signals mediate the translocation of the nascent protein chain between the cytosolic and the ER interior spaces. Co- and posttranslational processing of the flaviviral polyprotein involves proteolysis by viral and cellular proteases (Fig. 1). The membrane-bound nonstructural and structural proteins accumulate on the cytoplasmic surface and the ER lumen (33). The replication complex on the ER forms specific membrane-bound compartments and provides a protective milieu where replication intermediates are hidden from cellular factors responsible for activating the double-stranded RNA (dsRNA)-dependent pathways of the innate immune system (34). Assembly of virions of the nascent genomic RNA and the three structural protein components (C, prM, and E) occurs in close proximity to the viral replication complex on the endoplasmic reticulum. The virions mature during the journey through the secretory pathway involving the Golgi network (27). The mature virions are released from the cell through exocytosis (33).

Epidemiology and EcologyAdaptation of tick-borne flaviviruses (TBFVs) to specific invertebrate and vertebrate hosts directly influences the dispersal of the pathogens and shapes the epizootiology of these viruses. Therefore, the emergence and reemergence of TBFVs are strongly correlated with population dynamics of the vector and the amplifying host species. It should be noted that similarly to other vector-borne infections, a broad range of factors affect the dynamics of TBFVs, such as climatic, ecological, or human-related changes (i.e., mobility and agricultural patterns) (11, 35). Since the occurrence and distribution of vector tick species are highly influenced by climate, global warming and its side effects may contribute to the future expansion of the geographic distribution of some tick species (36, 37). Such scenarios are well discussed in the cases of Dermacentor reticulatus and Ixodes ricinus, two tick species that seem to be establishing new foci in Europe (38, 39). Migratory birds are also thought to play a role in the natural dispersal of some TBFVs at long distances (40). Human activities were also reported as factors in the long-distance spread of TBEV, and at least two major events were described: the building of a Siberian highway and the construction of the Trans-Siberian Railway (41). Human activities not only affect virus dispersal events but also have been demonstrated to influence the incidence of TBFV diseases. For instance, DDT (dichloro-diphenyl-trichloroethane) usage in the former Soviet Union greatly reduced the tick vector populations, which in turn resulted in a significant decrease in TBE incidence (42). Discontinuing the routine use of DDT worldwide may have led to an increase in the tick population, which played a role in the increase in disease incidence over the past 20 years (43). Based on these findings, it is anticipated that utilization of pesticides that are environmentally safe may successfully reduce the risk of TBFV infections by controlling the tick vector populations.

Ticks can serve as both reservoirs and vectors, while the vertebrate host provides a transport route between cofeeding ticks with so-called nonviremic transmission (Milan Labuda’s enduring paradigm) (44–49). This unique transmission route seems to be facilitated mainly by leukocytes migrating between collaterally present tick feeding sites and, interestingly, does not depend on the viremic stage of the vertebrate host (49, 50). The major role in the maintenance of these viruses can therefore be attributed to horizontal means of transmission with leukocyte-mediated cofeeding transmission, which might be more important than transovarial transmission from the adult female tick to the eggs (51, 52). Although this transmission route has been fully characterized for TBEV, it was also stated that transovarial transmission is not the main route of transmission between tick vectors (53, 54). While the viremic stage seems to be less important for the natural transmission of TBFVs, viral RNA can be detected in various rodent organ samples (55–57). Replication in the vertebrate host might play an important role in virus overwintering (58). The significance of different tick species in overwintering of TBEV has also been reported (59, 60).

Numerous rodent, ungulate, lagomorph, and bird species have been recognized as intermediate hosts for TBFVs (Table 1). Despite the fact that TBFV infection induces viremia in these hosts, the question of whether these species are able to participate in virus maintenance in nature remains open. These vertebrates are thought to serve as intermediates for horizontal transmission events, which directly influence the transmission cycle by providing hosts for reproducing ticks (61, 62). Probably due to the very short and low viral titer during the viremic period, humans are considered dead-end hosts for TBFVs (36).

TBEV is the most significant TBFV, with an estimated 10,000 to 15,000 annual cases in Europe and Asia. The incidence greatly depends on vaccination coverage among residents at the greatest risk of infection in affected areas (63). The case fatality rate largely depends on the infectious subtype of TBEV and is known to range between 1% (for the Western subtype) and 20% (for the Far Eastern subtype) (36, 64–66). Although TBEV is the most abundant of the TBFVs, other viruses should be noted as characteristic tick-borne pathogens worldwide. OHFV is represented in Siberia (67), while KFDV is a commonly identified pathogen in some regions of India (68, 69). LIV causes infections in the United Kingdom, Ireland, and southwestern Norway, with an introduction to Spain (70), and POWV is the causative agent of severe illnesses in North America (71). Alkhurma virus (AHFV), a subtype of KFDV, is present in Saudi Arabia and causes infections associated with hemorrhagic fever manifestations (69). Langat virus and Karshi virus are not considered medically important TBFVs (Table 1).

PathogenesisTBFVs can be divided into two groups based on their different tissue tropisms. Due to the high affinity of the virus for neural tissue, neurological disease manifestation is a characteristic pathogenetic scheme in TBEV, LIV, and POWV infections (70, 72–76). In contrast, KFDV, AHFV, and OHFV infections are typically associated with hemorrhagic manifestations, although neurotropism has been described for KFDV (68, 77–80). A strict hematopoietic and vascular tissue tropism has been linked to OHFV infections (81). Details about the pathomechanism of TBFVs are only partially understood, and most data are available from TBEV infections. Similarly to the majority of arthropod-borne viruses, the initial replication site of TBFVs is usually proximal to the tick bite. Macrophages and dendritic cells (DCs) are believed to transport the replicating virus to draining lymph nodes, where replication continues. This event results in a transient viremic stage of the host, which may lead to multiorgan dissemination of the virus. In POWV-based infection experiments, macrophages and fibroblasts have been identified as early cellular targets of infection close to the tick feeding site (82).

During the viremic stage, neurotropic flaviviruses can invade the central nervous system (CNS) by crossing the blood-brain barrier (BBB). Studies suggest that tumor necrosis factor alpha (TNF-α) may serve as a key factor in modulating the permeability of the BBB for some neurotropic flaviviruses (83). Although the exact molecular background of flaviviral BBB crossing is largely unknown, the brain capillary endothelium may play a key role in this mechanism (84). The main target cells for TBEV in the CNS are neurons, and virus-induced neuronal injury has been characterized in detail (74, 85–87). In addition, glial cells may also be involved in viral pathogenesis, particularly in the neurological stage of infection. Interestingly, rat astrocytes resist TBEV-mediated cell death, which might explain dormant TBEV infection in rodents (88). This capacity of astrocytes might contribute to maintaining the enzootic cycle in nature via long-term infections (58). In this process, a rapid type I interferon (IFN) response is thought to protect astrocytes from flavivirus-induced cell death (89). For TBFVs causing hemorrhagic disease, a characteristic liver and spleen tropism is seen, resulting in viscerotropic disease manifestations (11, 67, 90).

Genetic factors of the host and virus may be important factors in clinical disease severity. Recent data indicate that a lower expression level of the chemokine receptor CCR5 in peripheral blood lymphocytes is associated with greater susceptibility to TBEV infection (91). Toll-like receptor 3 gene mutations were also suggested to be an important genetic marker for the severity of TBEV infection; however, this observation needs further confirmatory studies (92). Mutations in the promoter region of the CD209 gene (encoding DC-SIGN, a C-type lectin pathogen recognition receptor [PRR]) may be associated with a predisposition for severe forms of tick-borne encephalitis (31). Evidence that viral genetic factors may also contribute to the severity of infection was supported by the genetic characterization of different TBEV strains from symptomatic and asymptomatic infections. Mutations affecting the virion proteins, NS3, NS5 proteins, and even the 3ʹ NCR were linked to altered pathogenic properties (93, 94). Despite the progress in investigating viral pathogenesis, the background of the TBFV pathomechanism remains poorly understood, especially for less-common TBFVs.

ImmunityTick-borne flaviviral infections affect the immune system of vertebrate hosts in a unique manner, in which the proximity of the tick bite site is the first step. This special environment, along with its distinctive molecular mechanisms, can highly affect the outcome of infection. Saliva of the tick Ixodes ricinus is able to alter dendritic cell-induced responses by decreasing TNF-α and interleukin-6 (IL-6) production and reducing TBEV-induced apoptosis (95). This proinflammatory environment is the first barrier against infection, and it has been reported that the bite of a POWV-infected tick induces more-rapid immune cell recruitment at this site than a bite from an uninfected tick (82, 96).

The mammalian immune response against flaviviral infection includes both innate and adaptive immune processes. As an early response, innate immunity activates multifaceted pathways rapidly after virus introduction with the primary aim of reducing the replication efficiency at the site of infection. The virus is primarily detected by PRRs of the host cell. PRRs detect pathogen-related single- and double-stranded RNA molecules and initiate multiple signaling pathways that lead to the production of IFNs (97, 98). Type I IFNs play a central role in the immune response. Processes involved in this pathway have been reviewed in detail by Kawai and Akira (98). IFNs stimulate the JAK-STAT signal transduction pathway and amplify further IFN responses, resulting in the expression of numerous interferon-stimulated genes.

DCs are the early targets of invading flaviviruses (47, 99), along with macrophages and fibroblasts, as was suggested in a recent study (82). Additional important functions of DCs are PRR-based virus detection, which leads to the production of type I IFNs, the transportation of the virus into draining lymph nodes, and antigen presentation that induces the adaptive immune response (100). The latter mechanism is what makes DCs the bridge between innate and adaptive immunity. TBFVs developed multiple countermeasures against the DC-dependent immune response, as exemplified by the antagonism of IFN regulatory factor 1 (101) or the suppression of JAK-STAT signaling via virus-encoded IFN antagonists (28, 102, 103).

DCs undergo a maturation program which leads to the activation of naive T cells, resulting in the production of T helper type 1/type 2 and cytotoxic T lymphocytes. DC-related PRR-based virus recognition controls the activation of multiple signaling pathways, such as higher expression levels of major histocompatibility complex (MHC) class II molecules, T-cell-costimulatory molecules, and proinflammatory cytokines (104). DCs are also responsible for generating protective CD8+ T-cell immunity using molecular signaling (104). The CD8+ T-cell response was recently examined in depth in acute human TBEV infections, including temporal dynamics, specificity, and functional and phenotypical characteristics of the response (105). Another recent study suggested some major differences in the natural killer (NK) T-cell response against human TBEV infection compared to other human viral infections (106).

In primary TBFV infections, the antibody response appears mostly at the end of the viremic phase, when IgM antibody titers increase rapidly. Normally, within 1 to 2 days following infection, specific IgG levels also start to increase. In instances of secondary infections, IgM levels show only a slight increase, whereas the IgG response is rapid and robust and efficiently terminates viremia (107).

Prevention, Control, and TreatmentWith the exception of TBEV, specific prophylactic treatment against TBFVs is not available. For prevention of TBEV infection, four commercially available inactivated vaccines have been marketed to date. Two of them are licensed in Russia and produced from the Far Eastern subtype of the virus (TBE-Moscow; EnceVir), while the other two vaccines available in Europe contain the inactivated European subtype (FSME-Immun; Encepur) (108). Vaccination in areas of endemicity significantly reduced the incidence of TBE, but it remained high in nonvaccinated populations (109). The annual incidence of TBEV infections has slightly increased during the past decades in several European countries (110, 111). Therefore, some authors argue for the need for mass immunization programs in affected areas and recommend the extension of vaccination to possible new regions of endemicity (112).

Immune therapy using hyperimmune IgG is not recommended for TBEV infections because of concerns of possible antibody-enhanced infection (113, 114).

Along with vaccination programs, tick bite prevention actions are the most efficient way to avoid infections. The risk of viral infection must be highlighted for those visiting areas of endemicity for professional or sporting activities (e.g., forestry, farming, hunting, and military activities) and even for leisure (e.g., hiking and orienteering). Also, control of small mammals (mainly rodents) should be implemented around home environments to minimize contact with putative primary vertebrate hosts, and our pets (dogs and cats) should be protected against tick bites and examined regularly to prevent tick introduction into the living space. Preventive arrangements and basic risk factors were recently reviewed (115).

Novel approaches based on targeted microRNA (miRNA) control of TBFV infections have recently been successfully tested using cell culture and mouse models. This novel therapeutic method could theoretically inhibit virus replication in both arthropod and vertebrate hosts. The available data on abundant host microRNAs (116, 117) enable the design of target sites along with various virus genomes specific for host microRNAs (118, 119). One major disadvantage of virus attenuation by this strategy is the instability of the target miRNA sequence in the modified genome, which can result in a reversion of the virus to the virulent phenotype, although several results on counterstrategies were reported (120–122). Other approaches offer replication-defective, chimeric vaccines, in which parts of the TBFV genome are replaced by homologous genomic regions derived from a heterologous (e.g., mosquito-borne) flavivirus. In animal models, these vaccine candidates have been shown to be highly immunogenic even after a single dose (123).

Dual-action vaccines have been extensively examined during the past years in order to control tick-borne pathogens. The principle of dual-action vaccination lies in the inhibition of blood-feeding mechanisms and rupture of the tick midgut, causing death of engorged ticks. Promising results have been obtained in controlling parasitic and bacterial tick-borne pathogens with such experiments. In laboratory mice, TBEV infection was successfully prevented with recombinant tick cement protein (a protein derived from the cement cone that secures the tick’s mouthparts during feeding) (124, 125). Results of subsequent studies were recently reviewed (126).

Concerning virus-specific therapy, although etiological treatment has not been approved to date, several studies dealing with possible antiviral agents for treatment of TBFV infections were reported. For example, 6-azauridine, 2ʹ-C-methylcytidine, and IFN-α2a successfully inhibited AHFV, KFDV, OHFV, and POWV replication in cell culture (127). Further studies were performed with the adenosine analogue NITD008, which was considered a potentially efficient panflaviviral inhibitor (128). Additionally, high antiviral activity along with low cytotoxicity were observed ex vivo with a 7-deaza-2ʹ-C-methyladenosine nucleoside analogue, which might be another antiviral candidate in the future (129).

POWASSAN VIRUS

HistoryPowassan virus (POWV) was named after the town Powassan (Ontario, Canada), where it caused the death of a 5-year-old child diagnosed with encephalitis (130). In the United States, the first human POWV case was described in 1970 in New Jersey (131). Russia was the third country that identified human POWV infection in 1978 (132). Interestingly, the first POWV isolates originated from tick vectors and had been collected before human infections were documented; this is exemplified by the earliest U.S. isolate of POWV from a Dermacentor andersoni tick collected during 1952 in Colorado (133) and the first POWV isolates from Russia, including those detected in ixodid ticks collected in 1970 (GenBank accession number KT224351).

EpidemiologyAs a result of detections across North America and the Russian Far East, POWV is now considered the only known member of the TBFVs that naturally occurs in both the Old and New World (Fig. 2). Human illnesses associated with POWV infection have been reported in Canada, the United States, and Russia (130–132, 134). Humans most likely contract POWV from biting ticks carrying the virus. However, most patients with POWV disease (or their caregivers) cannot recall a preceding tick encounter, very likely because tick bites are easily overlooked (135–139). Alternative infection routes cannot be excluded either; however, the hypothesis that individuals who consume unpasteurized goat milk in areas where POWV is endemic are, theoretically, at risk of POWV infection awaits formal demonstration (140, 141). From the perspective of disease prevention, understanding the time required for the transmission of the pathogen from tick to vertebrate is a key factor. In infection experiments that aimed at determining the dynamics of transmission of POWV from ticks to laboratory mice, the majority of experimental animals exposed to feeding by Ixodes scapularis ticks carrying POWV became infected 15 min following attachment, and all animals became infected after 30 min (142). Although the length of attachment time required for the development of human disease is unknown, relatively short tick attachment times (less than 3 h) associated with subsequent clinical illness progression have been reported (143). The short time for effective tick-to-vertebrate host transmission of POWV in mice, and likely in humans, sharply contrasts with the longer (up to 72 h) skin contact time needed for effective transmission of nonviral tick-borne pathogens, including Borrelia burgdorferi (142, 144). Thus, while the removal of ticks several hours after attachment to skin may prevent infection with Borrelia burgdorferi, it may not prevent infection with POWV.

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Geographic distribution of POWV. Dots indicate the locations where POWV was identified and/or serological assays provided indirect evidence for local circulation.

From the late 1950s to the mid-2000s, around 40 to 45 human POWV illnesses were documented in North America, with the majority of cases being reported in the United States, and at least 16 cases were reported in the Russian literature, encompassing a 10-year study period in the 1980s (132, 137, 145–147). The low POWV detection rate in this period (∼1 to 1.5 cases per year) was followed by a marked increase in reporting of POWV illnesses during the subsequent decade in the United States (4). Between 2007 and 2016, a total of 98 cases were documented by the U.S. Centers for Disease Control and Prevention (CDC), with an average of ∼10 cases (range, 2 to 22 cases) per year (Fig. 3) (see https://diseasemaps.usgs.gov/mapviewer/ and https://www.cdc.gov/powassan/). POWV illness occurs in all age groups; however, while early reports showed a greater incidence in children under 15 years of age (∼70% [reviewed by Gholam and coworkers for 1958 and 1998 {139}]), more-recent data reported by the U.S. CDC showed that only 8% of patients with POWV illness were younger than 18 years of age, whereas ∼50% of patients were older than 60 years of age (139, 148–153) (Fig. 3). Similarly to other tick-borne infections in North America, POWV infections tend to show a peak incidence from May to November (Fig. 3). In the United States, the most heavily affected states are Massachusetts, Minnesota, New York, and Wisconsin, but human illnesses were also reported in New Hampshire, New Jersey, Maine, North Dakota, Pennsylvania, Tennessee, Vermont, Virginia, and Connecticut (135–137, 143, 154–158). The highest incidence of POWV neuroinvasive disease (≥0.5 cases per 100,000 residents) was documented in some counties in the states of Minnesota and Wisconsin (see https://diseasemaps.usgs.gov/mapviewer/ and https://www.cdc.gov/powassan/). POWVs were identified in a few tick species (including Ixodes and Dermacentor spp.), whereas POWV or POWV-specific antibodies were detected in at least 38 wild and domestic mammals and a few bird species from well-known areas of endemicity and from other regions across the United States, including Alaska (147, 159). Virus serology data suggest that POWV may occur in northwest Mexico (Sonora state) (160). In Canada, human infections have been identified primarily in New Brunswick, Quebec, and Ontario; however, epidemiological and ecological surveys also detected POWV in Nova Scotia, Prince Edward Island, Alberta, and British Columbia (139, 161–163). In Russia, POWV disease has been reported in the Russian province of Primorsky Krai (Maritime Territory), whereas seropositive rodents whose sera reacted with POWV antigen were also detected in other parts of Siberia (134, 146, 159). It is important to note that while data based on serology assays indicate a wide distribution of POWV across North America, cautious interpretation is needed for results obtained by some of the available laboratory tests that tend to exhibit cross-reactivity due to shared epitopes on POWV-derived antigens and those expressed by other flaviviruses (147, 159).

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Epidemiological features of POWV infections between 2004 and 2016 in the United States. The panels show the annual number (A), the seasonal distribution (B), and the age distribution (C) of cases reported to the Centers for Disease Control and Prevention.

Because of the low incidence of human illness in areas of endemicity, seroprevalence of POWV in the general population is thought be a good proxy to better describe the overall exposure to and geographical distribution of human infections with POWV. However, data reported so far are scant and not spatiotemporally representative. Seroprevalence rates ranged between 0 and 5.8% among residents of Ontario in the late 1950s and 1960s, showing variation among communities, a finding that suggested a focal occurrence of both tick vectors and POWVs (141, 164, 165). In seroepidemiological studies conducted later among residents in British Columbia, New York, Minnesota, and Wisconsin, 6.1%, 0.7%, 4%, and 4% of individuals, respectively, had POWV antibodies in their sera (147, 163, 166, 167). These data from areas of endemicity are consistent with the hypothesis that many infections remain undetected due to absent or mild symptoms. In addition, a portion of symptomatic POWV infections could be masked by the clinical manifestations associated with other human-pathogenic tick-borne microorganisms cotransmitted with POWV by tick bite. So far, no longitudinal serological studies have been conducted to determine the dynamics of POWV seroprevalence in residents of areas of endemicity, and only limited data are available for animals. Notably, a recent seroepidemiological study that used sera collected in Connecticut from white-tailed deer (Odocoileus virginianus) demonstrated a marked temporal variation in seroprevalence but overall an increasing trend of seropositivity for POWV, rising from <25% before 1996 to 80% to 91% between 2005 and 2009 (168).

The increased reporting of POWV encephalitis in humans may be, in part, explained as a result of enhanced surveillance activity for arboviruses in North America established following the incursion of West Nile virus (WNV) in the United States in the late 1990s (4, 169, 170). However, the increasing seroprevalence in white-tailed deer cannot be explained by the enhanced monitoring of arbovirus-related human diseases. Thus, other contributing factors must also be taken into account. The dramatic territory expansion of an important competent tick species, Ixodes scapularis, seen over the past decade is probably the most important contributing factor (171).

Risk factors for human POWV infection include outdoor activities in areas of endemicity, contact with wild animals thought to serve as natural hosts of POWV, as well as family pets infested by ticks. To reduce the risk of being infected with POWV, it is recommended that individuals use tick repellents, wear adequate clothes (long sleeves and pants), avoid dense underbrush and wooded habitats, minimize the presence of wild mammals around homes, and, very importantly, perform thorough tick checks after spending time outdoors (136, 147) (https://www.cdc.gov/powassan/index.html).

Diversity and EvolutionPOWV is not genetically uniform. Evidence of genetic diversity was reported in 1997 when a novel POWV, called deer tick virus (DTV), was detected in Ixodes scapularis (172). Comparison of the genome sequences of representative POWV-like (genome size, 10,839 bases; encoded polyprotein, 3,415 aa) and DTV-like (genome size, 10,834 to 10,837 bases; encoded polyprotein, 3,415 aa) isolates showed 84% to 85% nucleotide and 92% to 95% amino acid similarities between the two related groups of isolates. Minor length differences are localized to the 3ʹ untranslated region (UTR) (134, 173–176). Analysis of the coding region revealed that POWV-like isolates (unlike DTV-like isolates) have two consecutive initiation codons for the C protein. One or two amino acid differences were described at the cleavage sites of the polyproteins of POWV and DTV at junctions of C/PrM, PrM/M, NS1/NS2A, NS2B/NS3, NS3/NS4A, and NS4A/NS4B (174). The functional consequences of these changes, if any, remain to be elucidated. Despite the genetic differences, the phenotypic implications are currently unclear, although Ebel (71) anticipated that phenotypic differences may exist in the respective primary tick vectors. Experimental data indicate that DTV and POWV are antigenically closely related, as demonstrated by hemagglutination inhibition assays and cross-neutralization tests, and also display similarities in neurovirulence in laboratory mice (174, 175). Both major variants are pathogenic for humans (130, 131, 177, 178).

Currently, the publicly available sequences are derived from around 60 separate virus isolates or clinical specimens. Several sequencing studies were initiated to better understand associations between the major variants of POWV and their tick vectors and to characterize strains identified from human disease (134, 177, 179–184). Thus, the majority (>90%) of POWV sequences originate from ticks and humans, and only a few sequences are available from wild animals (e.g., black-faced bunting, fox, woodchuck, and red squirrel) or other possible vectors (Aedes sp.). Although POWV-specific sequence data are accumulating slowly, researchers have used the available genetic information to clarify the evolutionary history and genetic relationships among North American isolates and between North American and Russian POWVs.

Molecular epidemiology and phylogenetics studies showed that POWV and DTV form two major genetic clusters (Fig. 4), designated lineage I for POWV-like strains and lineage II for DTV-like strains (71, 185). POWV isolate LB that originated from the first recorded human case in 1958 (Powassan, Ontario, Canada) is the prototype isolate of lineage I, while the first isolate from Dermacentor andersoni collected in 1952 (Colorado) is the prototype DTV isolate representing lineage II (174). Epidemiological surveillance conducted by combining various laboratory methods (including virus neutralization tests, reverse transcription-PCR [RT-PCR], and sequencing) indicated that lineage I POWVs occur in the United States, Canada, and Russia, while lineage II POWVs are known to occur in the United States and Canada (134, 176). Although the geographic dispersals of lineage I and II POWVs seem to overlap in at least Ontario, Canada, and in the northeastern regions of the United States, collecting additional sequence data from different geographic locations is needed to construct a more detailed distribution map for the two lineages.

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

Phylogenetic tree showing the geographic distribution and species association of lineage I and II POWVs.

POWV is a distant relative of other, Eurasian TBFVs, which is believed to have diverged ca. 12,000 to 13,000 years ago as a likely consequence of animal migration across the Bering land bridge during the latest ice age (186). Further sequence analyses offered intriguing explanations of the genetic diversity and the geographic distribution of POWV. The level of genetic divergence between and the molecular evolutionary analysis of lineage I and II POWVs suggested that they diverged around 500 to 2,000 years ago (this uncertainty comes principally from differences in data sets and analytical methods used to estimate the time when the most recent common ancestors existed) (176, 187, 188). Additional analyses identified molecular traces on the E protein being under selective pressure, providing evidence for the adaptation of POWV lineages to different enzootic cycles (176). Phylogenetic analysis also demonstrated further divergence to sublineages within both major POWV lineages; however, the evolutionary driving forces behind this diversification have not yet been clarified (176). Of interest, all currently known Russian isolates of POWV cluster with the prototype LB strain identified in Ontario, Canada, and share very high genetic similarity (up to 99.8% nucleotide identity) with all North American lineage I POWV strains, suggesting a single introduction of POWV into Russia during recent human history. Russian authors hypothesize that although natural expansion of POWV to Russia, for example, by long-distance-migrating birds, cannot be excluded, a more likely scenario links this introduction to human activity, such as importation of muskrat or mink to supply fur farms in Russia or shipment of commercial or military cargo (134).

Apparently, further studies are needed to clarify the evolutionary history of POWVs, and these estimates will require larger data sets that include sequences from neglected tick and vertebrate species. Nonetheless, POWVs continue to evolve in their vectors and vertebrate hosts. Overall, the rate of evolution was estimated to be higher in North American POWVs than in Russian POWVs (5.7 × 10−4 versus 2.4 × 10−5 substitutions per site per year for the E protein, respectively) (134, 176). This phenomenon is not clearly understood; however, differences in the vector-host relationships that may act as a limiting factor for POWV evolution leading to lower virus diversity in the Old World territory of POWV have been implicated (134). It was observed that the population complexity of lineage II POWV in ticks is very low and that POWV resists the pressure to diversify in response to RNA interference (RNAi) within ticks; these features contrast with the RNAi-mediated diversification of mosquito-borne flaviviruses in mosquitoes (189, 190). Viral population genetic analyses demonstrated that the majority of genetic changes occur during the few days of horizontal transmission from tick to small mammal. However, seeing the low rate of evolution in nature, which corresponds to ≤2 mutations per genome per year, it is conceivable that horizontal transmission is infrequent and may occur only once a year (190).

Ecology, Vectors, and Vertebrate HostsEfforts to describe the natural history of POWV began after the first human case in Powassan (Ontario, Canada) was identified. Initial virus isolation attempts and serological surveys performed in the vicinity of Powassan revealed that the natural cycle of POWV includes some small mammals and ticks and that peak transmission from ticks to mammals occurs during spring and summer (191–194). Virus activity was found to vary year by year, which may be affected by the tick population density, the rate of tick infestation of mammals, and the availability of various vertebrate host species. Antibody prevalence was detected in a number of small- to medium-sized mammalian species and also some large animals but not universally in all study sites, showing geographic differences in the possible role for these host species in the natural cycle of POWV infection (71). The study design, with a small sample size tested, or the antibody detection methods chosen may have an impact when determining the reservoir role for a particular host species in a geographic region. Unlike serological evidence that showed numerous possible reservoirs and tangential host species, POWV itself was relatively infrequently isolated in vertebrate hosts.

Regardless of the serological methods used to measure circulating serum antibody, the mammalian hosts incriminated in the ecology of POWV infections included a variety of rodents and lagomorphs, such as red squirrel (Tamiasciurus hudsonicus), gray squirrel (Sciurus carolinensis), chipmunk (Tamias striatus), yellow-pine chipmunk (Tamias amoenus), Columbian ground squirrel (Citellus columbianus), golden-mantled ground squirrel (Callospermophilus lateralis), groundhog (Marmota monax), yellow-bellied marmot (Marmota flaviventris), deer mouse (Peromyscus maniculatus), white-footed mouse (Peromyscus leucopus), Pinon mouse (Peromyscus truei), northern red-backed vole (Myodes rutilus), southern red-backed vole (Myodes gapperi), porcupine (Erethizon dorsatum), and snowshoe hare (Lepus americanus). Virus isolation or serological studies also found evidence of natural infection in mustelids, such as spotted skunk (Spilogale putorius), striped skunk (Mephitis mephitis), long-tailed weasel (Mustela frenata), and short-tailed weasel (Mustela erminea), and some other mammals, including Virginia opossum (Didelphis virginiana), coyote (Canis latrans), fox (Vulpes vulpes), gray fox (Urocyon cinereoargenteus), raccoon (Procyon lotor), and white-tailed deer (Odocoileus virginianus) (184, 191–209). Birds are not thought to play a role in POWV maintenance in nature; however, a recent survey analyzing virus prevalence and serological reactivity to POWV in New York state birds detected serum antibody in veery (Catharus fuscescens), gray catbird (Dumetella carolinensis), northern cardinal (Cardinalis cardinalis), and Eastern towhee (Pipilo erythrophthalmus) (184). In addition, POWV was isolated from common teal (Anas crecca), mallard (Anas platyrhynchos), and black-faced bunting (Emberiza spodocephala) in Russia (209) (GenBank accession number KU297222). Collectively, while the host species list of wild animals that are susceptible to POWV infection is becoming more complete, the role of most species in POWV perpetuation remains unknown (71).

Seroepidemiological studies conducted in some regions showed that domestic animals display significant variation in susceptibility to POWV infection and, in general, are not thought to be involved in the persistence of POWV in nature. However, some species might have a role in the transmission of POWV to humans, primarily by increasing the likelihood of human exposure to infected ticks. Experimental infection resulted in severe neuroinvasive disease in horses inoculated with POWV, and seroepidemiological studies conducted in Ontario found that 13% of examined horses had circulating serum antibodies against POWV (210). Cows, sheep, and goats occasionally sampled in serosurveys were not found to be naturally infected with POWV; however, goats infected experimentally developed transient viremia without clinical signs of infection and also released infectious virus in milk (140). In infection experiments, rabbits have developed viremia (210). Experimentally infected dogs and cats did not show symptoms of neuroinvasive disease; however, antibody was detected in their sera. Family cats and dogs were also shown to possess POWV-specific antibodies in a case report of a patient with POWV encephalitis, and around 1% of dogs surveyed in Ontario and British Columbia had antibodies to POWV (138, 211–213). Collectively, some lactating ruminant species may pose a theoretical risk for the alimentary route of infection with POWV in cases when unpasteurized milk is consumed, although this route of infection for POWV has never been documented. In addition, pets may serve as incidental hosts for ticks involved in the natural life cycle of POWV; thus, given that they may bring ticks into the home, pets may pose a risk for family members to become bitten by ticks carrying POWV.

The host preference of ticks has been studied extensively (214). These studies provide information about different vectors that may pose a major public health risk for disease transmission due to their host selection. For POWV, tick species that show high host specificity may be effective in POWV maintenance in the enzootic cycle but are thought to be inefficient in POWV transmission to humans. On the contrary, ticks that are more opportunistic in host feeding may pose a greater public health risk because they may more frequently transmit POWV from the common enzootic cycle to incidental hosts, including humans (71). Although the first isolation of POWV was made from a Dermacentor andersoni tick, the most important vectors in North America are thought to be Ixodes species, including Ixodes cookei, Ixodes marxi, Ixodes spinipalpis, and Ixodes scapularis. Detections of POWVs have been made from Ixodes cookei, Ixodes marxi, and Ixodes scapularis at a fairly constant rate over study sites and time periods that rarely exceeds 5% (184, 192, 193, 198, 199, 203). Other North American tick species are also thought to play a role in POWV maintenance; however, these other tick species are rarely studied for POWV carriage (71). Thus, for example, Dermacentor spp. have been only occasionally assayed for POWV, and no evidence so far indicates that Dermacentor variabilis or Dermacentor andersoni would play a major role in the infection cycle of POWV. In addition, laboratory investigations demonstrated that Dermacentor andersoni does not support the vertical transmission of POWV, thus confirming previous ecological observations (215). Nonetheless, data reported from Russia are consistent with the possibility that a number of other tick species may serve as vectors for POWV transmission, given that prototype POWVs in the Russian Far East were isolated from Ixodes persulcatus, Dermacentor silvarum, Haemaphysalis japonica, and Haemaphysalis longicornis ticks (216) (GenBank accession numbers KU297218, KU297219, KU297221, KU160627, and MG652438).

When trying to put epidemiological surveillance data in context with results from ecological studies, it is important to recall that POWVs exist in at least two genetic lineages that are associated with different vectors (71). Considering the feeding behavior and host preference of the implicated tick species, the natural cycles of lineage I POWV maintenance include Ixodes cookei and groundhog/skunk, and Ixodes marxi and red squirrel, whereas the hitherto-recognized natural cycle for lineage II POWV consists of the more opportunistic tick Ixodes scapularis and a variety of vertebrate hosts, including white-footed mouse. All three tick species may transmit POWV to humans, although Ixodes cookei and Ixodes marxi are generally considered more host specific, whereas Ixodes scapularis is more opportunistic and aggressively attacks humans, thus representing a major role in disease transmission to humans (71). Of interest, some host-specific tick vectors may display an extended host preference, as exemplified by Ixodes cookei, which shows a greater-than-usual preference for feeding on humans in Maine and thus may be a more common source of POWV infection in humans in this area than in other geographic regions (71) (Fig. 5). Although up-to-date information on POWV ecology in far-east Asia is not available, early reports from Russia suggest that ixodid ticks and murine rodents play a role in the perpetuation of lineage I POWV in enzootic foci (217, 218). Interestingly, virus monitoring conducted in the Russian Far East documented the isolation of POWV from mosquitoes (Aedes togoi); however, due to a lack of convincing evidence, the role for mosquitoes in POWV maintenance in enzootic areas was questioned (71, 219, 220).

FIG 5
  • Open in new tab
  • Download powerpoint
FIG 5

Enzootic cycles of lineage I and II POWVs linked to the main tick vector and some important vertebrate hosts. Humans are considered tangential hosts; arrows pointing to the silhouette on the right are indicative of the relative public health risk posed by different tick species (i.e., low risk for Ixodes marxi and Ixodes cookei and greater risk for Ixodes scapularis).

Clinical Disease and DiagnosisThe majority of POWV infections are thought to remain unseen by health professionals due to the presumably high rate of subclinical infections (164). When POWV infection is symptomatic, patients may develop flu-like illness or encephalitis/meningoencephalitis; these are the most commonly seen manifestations and are the major indications for the need for laboratory testing. Specimens collected from patients with CNS involvement are more likely to be tested for POWV. For example, the U.S. CDC reported that between 2010 and 2015, 85% (annual range, 75% to 100%) of tested patients had encephalitis or meningitis, while 15% (annual range, 0% to 25%) were reported to present with nonneuroinvasive disease (148–153). These findings suggest that the majority of patients without CNS manifestations are not routinely tested for the virus, and thus, published reports might underestimate the incidence of nonneuroinvasive POWV disease.

Clinical characteristics (Table 2) of POWV infection have been comprehensively overviewed in previous publications (147, 221, 222). Typical POWV infections have a 7- to 34-day incubation period followed by a prodrome phase with nonspecific flu-like symptoms, such as fever, chills, malaise, generalized weakness, sore throat, headache, myalgia, lethargy, somnolence, dizziness, and gastrointestinal symptoms (nausea and vomiting), in the majority of patients. Headache and fever up to a temperature of 38.5°C to 41°C are reported by all patients (4, 145, 157). A fine erythematous morbilliform rash was observed in some cases (138, 223–225). The prodrome phase lasts approximately 1 to 3 days and precedes CNS manifestations. The proportion of patients who develop CNS infection and manifest CNS symptoms is unknown.

View this table:
  • View inline
  • View popup
TABLE 2

Clinical presentation of POWV infectionsa

Altered sensorium ranging from confusion and stupor to coma is a universal finding in patients. Many patients (mainly children) exhibit evidence of meningismus or meningeal irritation upon physical examination (130, 138, 161, 162, 226, 227). Although children are commonly observed to have seizures, which are rarely seen in adults, febrile seizures are more common in children and thus may not be due simply to POWV infection (130, 138, 143, 161, 162, 226–229). Severe neuroinvasive symptoms, such as hemiplegia or even quadriplegia, are commonly observed in patients of all ages. Also, paresis, tremors, twitching, focal palsies, and pyramidal tract signs may occur. Moreover, but less frequently, nystagmus, ophthalmoplegia, facial palsy, myelitis, hallucinations, and respiratory failure were reported. A case of acute flaccid paralysis with residual consequence was also recorded (130, 139, 161, 162, 227, 228, 230–232).

POWV infection is associated with a high case fatality rate in patients with CNS disease and a high incidence of severe long-term sequelae (4, 130, 139, 178, 233). In one report, 12 of 14 patients (85%) with neuroinvasive POWV infection were admitted to the intensive care unit, and 5 patients (35%; all >60 years of age) died (178). Combining data from multiple case reports, for patients with POWV neuroinvasive illness, an ∼10 to 15% case fatality rate is estimated; however, this might be a conservative estimate, as extended follow-up is performed for only a portion of patients (136, 139, 178). Where data on outcomes have been described, either short-term or long-term significant neurological deficits were observed in about 75% of cases. Apnea, dysarthria, psychosis, and spasticity were recorded as short-term sequelae. Long-term sequelae include prolonged headache, cognitive deficit, significant limitation in activities of daily living, bed-bound state, spastic hemiplegia and quadriplegia, hemiparesis, mental retardation, aphasia, hearing impairment, muscle weakness, imbalance, ventilator dependence, ophthalmoplegia, and memory dysfunction (Table 3) (4, 145, 147, 178).

View this table:
  • View inline
  • View popup
TABLE 3

Prevalence of clinical symptoms, radiography findings, and neurological deficits among patients hospitalized with POWV neuroinvasive illness between 2004 and 2012 in New Yorka

General laboratory findings showed mild thrombocytopenia, lymphocytic pleocytosis (with fewer than 500 white blood cells per µl cerebrospinal fluid [CSF]), and elevated protein levels in the CSF in a majority of patients with CNS involvement. Neuroimaging findings and histopathological examinations helped to understand the background of clinical symptoms. In patients with POWV meningoencephalitis and encephalitis, magnetic resonance imaging (MRI) demonstrated deep foci of increased T2/FLAIR (T2-weighted fluid-attenuated inversion recovery) signal intensity (157). T2-hyperintense foci predominantly affected the gray matter, and microvascular ischemic changes were predominantly found throughout the brain. In 13 hospitalized patients with POWV encephalitis, cerebral cortex, basal ganglia, brain stem, cerebellum, thalamus, and meninges, all dominantly on the left side, were the affected brain areas (Table 3) (178, 221). Histological examination of brain samples obtained at autopsy revealed changes characteristic of acute meningoencephalitis. Histopathological analysis showed reactive gliosis, increased numbers of microglial cells, and necrotizing inflammation with a lymphocytic infiltrate, predominantly affecting the gray matter (139, 178).

Because the geographic distribution of POWV overlaps those of a variety of encephalitogenic viruses and bacteria in the Americas and Russia, and due to the wide range of clinical manifestations, a broad differential diagnosis is warranted. Depending on the geographic location of exposure, differential diagnosis should take into account TBEV, WNV, Saint Louis encephalitis virus (SLEV), or herpes simplex virus (HSV) encephalitis; bacterial meningitis; Lyme disease; anaplasmosis; ehrlichiosis; and tick-borne relapsing fever. Coinfections further complicate the situation and may contribute significantly to patient morbidity and mortality from tick-borne infections due to diagnostic difficulties and inadequate treatment (167). Despite the dramatic consequences for disease management, little is known about the prevalence of tick-borne pathogen coinfections in patients with POWV disease. One study reported the occurrence of Anaplasma phagocytophilum coinfection in one out of three patients with POWV encephalitis (137). In another study, among 41 patients with evidence of Borrelia burgdorferi infection, 7 (17.1%) showed serological evidence of acute POWV infection, and 3 (7.3%) had laboratory-confirmed POWV infection (167). In a case series analysis, which processed data from 14 patients hospitalized with POWV encephalitis, 2 patients (14.2%) with erythema migrans had detectable antibodies to Borrelia burgdorferi (178). These findings are in agreement with vector surveillance data that showed that ticks infected with POWV may carry other tick-borne pathogens simultaneously. Rates of detection of such coinfections in Ixodes scapularis ticks ranged from 1.1% to 3.4% in Connecticut and New York, respectively; in these studies, the ticks, which carried lineage II POWV, carried either Borrelia burgdorferi, Ehrlichia chaffeensis, Anaplasma phagocytophilum, or a combination thereof (234, 235).

The case definition for POWV-associated neuroinvasive infection includes clinical criteria (fever of >38°C with any peripheral or central nervous system dysfunction) and at least one of the following laboratory findings: (i) direct detection of POWV in tissue, blood, or CSF by virus culture or a molecular detection method; (ii) a 4-fold rise of POWV-specific antibody titers in paired serum samples; (iii) the presence of POWV-specific IgM in serum and the presence of POWV-neutralizing antibodies in the same specimen or a specimen collected during convalescence; and (iv) the presence of POWV-specific IgM in CSF and the simultaneous absence of IgM for other arboviruses endemic to the region where exposure occurred (https://wwwn.cdc.gov/nndss/conditions/powassan-encephalitis-meningitis/case-definition/2001/) (Fig. 6). Commercial diagnostic tests for POWV serology are not available for use in hospital laboratories, and although POWV-specific molecular detection methods have been marketed, these tests are recommended currently for research use only. In the United States, until recently, laboratory diagnosis of POWV has been carried out exclusively in specialized diagnostic laboratories, including state health laboratories and the CDC, where multiple diagnostic assays (including serology, virus isolation, and RT-PCR) were developed and validated (236–238). More recently, a POWV serology-based diagnostic panel has been developed at a private commercial laboratory (239).

FIG 6
  • Open in new tab
  • Download powerpoint
FIG 6

Clinical course and laboratory diagnosis of POWV infections. The timeline aligned with the phase of infection and typical clinical manifestations is shown in the top panel, whereas laboratory diagnostic opportunities relative to the timeline of clinical disease are shown in the middle and bottom panels.

The gold standard for laboratory diagnosis of POWV infection remains serology despite the fact that it may take several weeks to obtain adequate results by this approach due to the need for paired clinical specimens. A positive IgM test via an immunofluorescence assay (IFA), IgM antibody capture enzyme-linked immunosorbent assay (MAC-ELISA), or a microsphere-based immunoassay (MIA), with confirmation using plaque reduction neutralization testing (PRNT), is the primary choice for laboratory diagnosis. However, PRNT requires infectious viruses and an adequate biosafety level for handling infectious POWV, and the sensitivity of PRNT may be inferior to the sensitivity of other serological assays. Commercial serology tests specific for another flavivirus (e.g., ELISAs that are available for WNV and SLEV diagnoses) may give a positive test result with sera collected from POWV-infected patients due to cross-reactivity among flavivirus antigen epitopes (240, 241); importantly, IgM cross-reactivity can occur for other non-flavivirus-associated conditions, such as lupus or Epstein-Barr virus infection. In recent years, ELISAs and IFAs have been replaced by more-convenient medium- to high-throughput, multiplexed microsphere-based immunoassays (MIAs) that may target numerous North American arboviruses (including POWV) in a single test run to detect either IgM or IgG antibodies in CSF or sera (238).

Immunohistochemistry and virus culture from autopsy tissues can be useful and are indicative of active virus replication in the examined tissue sample. Keeping in mind that short-duration viremia may limit the sensitivity of direct virus detection, POWV can be isolated from the blood during the prodrome phase or later from the CSF when CNS manifestations develop. For the purpose of virus isolation, several cell lines are available, but suckling laboratory mice can also be used (130). Permissive cell lines include, but are not limited to, pig (PKE)-, monkey (Vero; LLC-MK2)-, and baby hamster (BHK21)-origin kidney cells and the human lung epithelial cell line A549 (127, 134, 180, 242). Although a cytopathogenic effect upon tissue culture may not be evident in some cultured cells, ex vivo culturing may be chosen to increase the virus titer for a subsequent in vitro detection method (134, 180, 242).

Nucleic acid amplification methods are increasingly becoming routine in virological diagnosis. Similarly to the virus isolation method, POWV RNA can be detected in brain tissue samples upon postmortem examination and, at various success rates, in CSF and blood samples collected during the prodrome phase and in CSF during the acute phase of illness (177, 224). The most widely used assay format is RT-PCR or RT combined with heminested PCR that preferentially targets either the E or the NS5 protein-coding genomic region; assays targeting the genomic NCRs have also been developed. Panflavivirus PCR-based detection has been reported to detect a variety of viruses in the Flavivirus genus, and some of these broad-range RT-PCR assays have been used to detect POWV from CSF and autopsy tissue samples (172, 243–248). Because POWV is sympatric with other flaviviruses in both North America and the Russian Far East, RT-PCR results obtained by a broad-range assay need to be confirmed. Confirmation of PCR-based amplification can be accomplished by sequencing or mass spectrometry of the amplified fragment or inclusion of virus-specific probes in a quantitative assay (142, 224, 249, 250). Multiplexing of molecular detection methods to potentially identify multiple targets simultaneously is an approach to reduce the assay cost while trying to keep the clinical sensitivity for each target; therefore, molecular diagnostic methods with increased multiplexity are being continuously developed. These assays may rely on expensive laboratory infrastructure and may require some special skills (including staff bioinformaticians).

In microsphere-based molecular detection assays, virus-specific probes are captured onto the surface of beads, and the number of targets may be flexibly chosen based on diagnostic needs, although the number of targets to be detected simultaneously is usually no more than a few dozen. For example, a commercial molecular diagnostic panel can identify 21 different tick-borne protozoal, bacterial, and viral pathogens, including POWV (Luminex). High-density panviral and panmicrobial DNA probe-based solid-phase microarrays are known to target hundreds to thousands of viral pathogens or a mixture of viral, bacterial, and fungal pathogens and parasites of vertebrates, respectively. Some of these assays were reported to include POWV-specific capture DNA probes (251–254). Also, a more recent advancement in panvirus detection methods is the combination of viral metagenomics with high-throughput next-generation sequencing (NGS). This approach has led to the discovery of numerous novel viruses but has limited value in clinical settings due to the low sensitivity. This low sensitivity was recently demonstrated for a patient with encephalitis of an unknown etiology, whose CSF sample, collected during the acute phase, was found to contain only 10 POWV (lineage II)-specific sequence reads out of 2.4 million total sequence reads generated for the sample (225). Targeted enrichment of possible viral targets by using a mixture of virus-specific capture oligonucleotides has been demonstrated to increase the sensitivity of NGS-based detection of numerous medically important viruses from clinical specimens (255). This approach is, theoretically, able to detect all currently known viruses; however, it lacks the unbiased feature of viral metagenomics and may be specific for viruses targeted based on known genetic sequence information and some others that are very closely related to the targeted viruses. Despite the availability of these emerging new technologies, the clinical sensitivity of the most promising high-throughput methods awaits formal demonstration for a majority of targets, including the two lineages of POWV.

CONCLUSIONS AND PERSPECTIVES

POWV was discovered nearly 60 years ago, and research efforts continue to focus on understanding clinical disease and pathology, ecology, epidemiology, and viral genetic diversity. The development of sensitive and highly specific laboratory detection methods was beneficial to achieve progress in these research areas. However, serological assays suitable for routine laboratory use in hospitals are not commercially available, which hampers diagnosis in settings where these tests are most needed. Given that POWV can be cotransmitted with various human-pathogenic microbes and viruses, utilization of diagnostic panels that target multiple tick-borne pathogens most typical in an area of endemicity could be useful, with the added benefits to better understand disease outcomes during mixed infections and to consider choosing the most adequate treatment options.

The number of case reports of POWV illness sharply increased over the past decade in the United States, from <1 case per year pre-2000s to 10 cases per year since the mid-2000s. Based on these findings, some researchers consider POWV an emerging threat in North America, and research investigations of this once neglected virus infection gained new momentum in the past few years. From a medical perspective, new research data that indicate an expanding range for Ixodes scapularis, a major arthropod vector that aggressively attacks humans and transmits lineage II POWV, seem to be relevant, a finding based upon which an increased POWV prevalence in the United States is anticipated. Continued virus monitoring and disease surveillance will certainly help to establish if this expectation holds true over an extended period. Epidemiological surveillance will require support from enhanced field and laboratory studies to explore other possible hosts and vectors whose distributions overlap those of known POWV hosts and vectors and to assess the public health risk potentially posed by these additional species.

Despite the low public health burden of POWV infections currently, the considerable case fatality ratio and the high risk for short- or long-term sequelae characterized by various motor, sensory, and cognitive disfunctions in affected persons justify implementation of more research efforts to help understand disease pathogenesis and immunity and to explore new options in prevention and therapy. Prevention focuses on the avoidance of tick infestations by using personal protective measures and tick control. Vaccines against POWV are not available for use, and in light of the low disease incidence, it is questionable if vaccines would be cost-effective. Therapy for patients with POWV illness is palliative. Some patients with POWV neuroinvasive illness who were administered intravenous immunoglobulin or high-dose corticosteroids survived; however, whether these treatments have any beneficial effect on favorable disease outcomes remains unclear and needs additional confirmatory data. Antiviral treatment is not available, and data on the susceptibility of POWV to various antiviral drugs are scant. The lack of comprehensive information on antiviral approaches against POWV is somewhat inexplicable despite the growing database of antiviral drugs that act directly on flaviviral gene products and may be effective against these viruses. As an alternative, the emerging field of antiviral treatment using drugs that target host proteins required for various processes of the flavivirus cellular life cycle may also be better explored. Contrary to conventional antiviral drugs, whose routine use could readily select drug-resistant viral clones, an approach that targets cellular components required for the flavivirus life cycle may have the added advantage that resistance is less likely to develop given that host cell targets tend to evolve very slowly.

ACKNOWLEDGMENTS

We thank Kornélia Kurucz and György Szűcs for their assistance in preparation of illustrations. We are indebted to György Szűcs and Ferenc Jakab for the fruitful scientific discussions in relation to selected sections of the manuscript, and we appreciate the help of Ádám Tóth and Jon Gentsch for critical reading and editing of the text.

G.K. was supported by the University of Pécs in the frame of the Viral Pathogenesis Talent Centre program and also by the European Union, co-financed by European Social Fund grant no. EFOP-3.6.1.-16-2016-00004, entitled “Comprehensive Development for Implementing Smart Specialization Strategies at the University of Pécs.” K.B. was supported by the Momentum program. The funders had no role in collection or interpretation of data or the decision to submit the work for publication. Furthermore, there are no financial or other relationships that might lead to a conflict of interest.

  • Copyright © 2018 American Society for Microbiology.

All Rights Reserved.

REFERENCES

  1. 1.↵
    1. Dantas-Torres F,
    2. Chomel BB,
    3. Otranto D
    . 2012. Ticks and tick-borne diseases: a one health perspective. Trends Parasitol 28:437–446. doi:10.1016/j.pt.2012.07.003.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    1. Labuda M,
    2. Nuttall PA
    . 2004. Tick-borne viruses. Parasitology 129(Suppl):S221–S245. doi:10.1017/S0031182004005220.
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    1. Dobler G
    . 2010. Zoonotic tick-borne flaviviruses. Vet Microbiol 140:221–228. doi:10.1016/j.vetmic.2009.08.024.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Hinten SR,
    2. Beckett GA,
    3. Gensheimer KF,
    4. Pritchard E,
    5. Courtney TM,
    6. Sears SD,
    7. Woytowicz JM,
    8. Preston DG,
    9. Smith RP, Jr,
    10. Rand PW,
    11. Lacombe EH,
    12. Holman MS,
    13. Lubelczyk CB,
    14. Kelso PT,
    15. Beelen AP,
    16. Stobierski MG,
    17. Sotir MJ,
    18. Wong S,
    19. Ebel G,
    20. Kosoy O,
    21. Piesman J,
    22. Campbell GL,
    23. Marfin AA
    . 2008. Increased recognition of Powassan encephalitis in the United States, 1999–2005. Vector Borne Zoonotic Dis 8:733–740. doi:10.1089/vbz.2008.0022.
    OpenUrlCrossRefPubMedWeb of Science
  5. 5.↵
    1. Hermance ME,
    2. Thangamani S
    . 2017. Powassan virus: an emerging arbovirus of public health concern in North America. Vector Borne Zoonotic Dis 17:453–462. doi:10.1089/vbz.2017.2110.
    OpenUrlCrossRef
  6. 6.↵
    1. Simmonds P,
    2. Becher P,
    3. Bukh J,
    4. Gould EA,
    5. Meyers G,
    6. Monath T,
    7. Muerhoff S,
    8. Pletnev A,
    9. Rico-Hesse R,
    10. Smith DB,
    11. Stapleton JT
    , ICTV Report Consortium. 2017. ICTV virus taxonomy profile: Flaviviridae. J Gen Virol 98:2–3. doi:10.1099/jgv.0.000672.
    OpenUrlCrossRef
  7. 7.↵
    1. Pletnev A,
    2. Gould E,
    3. Heinz FX,
    4. Meyers G,
    5. Thiel HJ,
    6. Bukh J,
    7. Stiasny K,
    8. Collett MS,
    9. Becher P,
    10. Simmonds P,
    11. Rice CM,
    12. Monath TP
    . 2011. Flaviviridae, p 1003–1020. In King AMQ, Adams MJ, Carstens EB, Lefkowitz EJ (ed), Virus taxonomy. Classification and nomenclature of viruses. Ninth report of the International Committee on Taxonomy of Viruses. Elsevier Academic Press, San Diego, CA.
  8. 8.↵
    1. Blitvich BJ,
    2. Firth AE
    . 2015. Insect-specific flaviviruses: a systematic review of their discovery, host range, mode of transmission, superinfection exclusion potential and genomic organization. Viruses 7:1927–1959. doi:10.3390/v7041927.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Pierson TC,
    2. Diamond MS
    . 2013. Flaviviruses, p 747–794. In Knipe DM, Howley PM, Cohen JI, Griffin DE, Lamb RA, Martin MA, Racaniello VR, Roizman B (ed), Fields virology, 6th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
  10. 10.↵
    1. Calisher CH,
    2. Karabatsos N,
    3. Dalrymple JM,
    4. Shope RE,
    5. Porterfield JS,
    6. Westaway EG,
    7. Brandt WE
    . 1989. Antigenic relationships between flaviviruses as determined by cross-neutralization tests with polyclonal antisera. J Gen Virol 70:37–43. doi:10.1099/0022-1317-70-1-37.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    1. Lasala PR,
    2. Holbrook M
    . 2010. Tick-borne flaviviruses. Clin Lab Med 30:221–235. doi:10.1016/j.cll.2010.01.002.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Gould EA,
    2. de Lamballerie X,
    3. Zanotto PM,
    4. Holmes EC
    . 2001. Evolution, epidemiology, and dispersal of flaviviruses revealed by molecular phylogenies. Adv Virus Res 57:71–103. doi:10.1016/S0065-3527(01)57001-3.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Rey FA,
    2. Heinz FX,
    3. Mandl C,
    4. Kunz C,
    5. Harrison SC
    . 1995. The envelope glycoprotein from tick-borne encephalitis virus at 2 A resolution. Nature 375:291–298. doi:10.1038/375291a0.
    OpenUrlCrossRefPubMedWeb of Science
  14. 14.↵
    1. Zhang W,
    2. Chipman PR,
    3. Corver J,
    4. Johnson PR,
    5. Zhang Y,
    6. Mukhopadhyay S,
    7. Baker TS,
    8. Strauss JH,
    9. Rossmann MG,
    10. Kuhn RJ
    . 2003. Visualization of membrane protein domains by cryo-electron microscopy of dengue virus. Nat Struct Biol 10:907–912. doi:10.1038/nsb990.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    1. Grard G,
    2. Moureau G,
    3. Charrel RN,
    4. Lemasson JJ,
    5. Gonzalez JP,
    6. Gallian P,
    7. Gritsun TS,
    8. Holmes EC,
    9. Gould EA,
    10. de Lamballerie X
    . 2007. Genetic characterization of tick-borne flaviviruses: new insights into evolution, pathogenetic determinants and taxonomy. Virology 361:80–92. doi:10.1016/j.virol.2006.09.015.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    1. Saeedi BJ,
    2. Geiss BJ
    . 2013. Regulation of flavivirus RNA synthesis and capping. Wiley Interdiscip Rev RNA 4:723–735. doi:10.1002/wrna.1191.
    OpenUrlCrossRef
  17. 17.↵
    1. Chambers TJ,
    2. Hahn CS,
    3. Galler R,
    4. Rice CM
    . 1990. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol 44:649–688. doi:10.1146/annurev.mi.44.100190.003245.
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.↵
    1. Heinz FX,
    2. Allison SL
    . 2003. Flavivirus structure and membrane fusion. Adv Virus Res 59:63–97. doi:10.1016/S0065-3527(03)59003-0.
    OpenUrlCrossRefPubMedWeb of Science
  19. 19.↵
    1. Wallner G,
    2. Mandl CW,
    3. Kunz C,
    4. Heinz FX
    . 1995. The flavivirus 3ʹ-noncoding region: extensive size heterogeneity independent of evolutionary relationships among strains of tick-borne encephalitis virus. Virology 213:169–178. doi:10.1006/viro.1995.1557.
    OpenUrlCrossRefPubMedWeb of Science
  20. 20.↵
    1. Ng WC,
    2. Soto-Acosta R,
    3. Bradrick SS,
    4. Garcia-Blanco MA,
    5. Ooi EE
    . 2017. The 5ʹ and 3ʹ untranslated regions of the flaviviral genome. Viruses 9:E137. doi:10.3390/v9060137.
    OpenUrlCrossRef
  21. 21.↵
    1. Heinz FX,
    2. Stiasny K,
    3. Püschner-Auer G,
    4. Holzmann H,
    5. Allison SL,
    6. Mandl CW,
    7. Kunz C
    . 1994. Structural changes and functional control of the tick-borne encephalitis virus glycoprotein E by the heterodimeric association with protein prM. Virology 198:109–117. doi:10.1006/viro.1994.1013.
    OpenUrlCrossRefPubMedWeb of Science
  22. 22.↵
    1. Heinz FX,
    2. Auer G,
    3. Stiasny K,
    4. Holzmann H,
    5. Mandl C,
    6. Guirakhoo F,
    7. Kunz C
    . 1994. The interactions of the flavivirus envelope proteins: implications for virus entry and release. Arch Virol Suppl 9:339–348.
    OpenUrlPubMed
  23. 23.↵
    1. Stadler K,
    2. Allison SL,
    3. Schalich J,
    4. Heinz FX
    . 1997. Proteolytic activation of tick-borne encephalitis virus by furin. J Virol 71:8475–8481.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Heinz FX,
    2. Allison SL
    . 2001. The machinery for flavivirus fusion with host cell membranes. Curr Opin Microbiol 4:450–455. doi:10.1016/S1369-5274(00)00234-4.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    1. Rastogi M,
    2. Sharma N,
    3. Singh SK
    . 2016. Flavivirus NS1: a multifaceted enigmatic viral protein. Virol J 13:131. doi:10.1186/s12985-016-0590-7.
    OpenUrlCrossRef
  26. 26.↵
    1. Jacobs SC,
    2. Stephenson JR,
    3. Wilkinson GW
    . 1992. High-level expression of the tick-borne encephalitis virus NS1 protein by using an adenovirus-based vector: protection elicited in a murine model. J Virol 66:2086–2095.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Bollati M,
    2. Alvarez K,
    3. Assenberg R,
    4. Baronti C,
    5. Canard B,
    6. Cook S,
    7. Coutard B,
    8. Decroly E,
    9. de Lamballerie X,
    10. Gould EA,
    11. Grard G,
    12. Grimes JM,
    13. Hilgenfeld R,
    14. Jansson AM,
    15. Malet H,
    16. Mancini EJ,
    17. Mastrangelo E,
    18. Mattevi A,
    19. Milani M,
    20. Moureau G,
    21. Neyts J,
    22. Owens RJ,
    23. Ren J,
    24. Selisko B,
    25. Speroni S,
    26. Steuber H,
    27. Stuart DI,
    28. Unge T,
    29. Bolognesi M
    . 2010. Structure and functionality in flavivirus NS-proteins: perspectives for drug design. Antiviral Res 87:125–148. doi:10.1016/j.antiviral.2009.11.009.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    1. Best SM,
    2. Morris KL,
    3. Shannon JG,
    4. Robertson SJ,
    5. Mitzel DN,
    6. Park GS,
    7. Boer E,
    8. Wolfinbarger JB,
    9. Bloom ME
    . 2005. Inhibition of interferon-stimulated JAK-STAT signaling by a tick-borne flavivirus and identification of NS5 as an interferon antagonist. J Virol 79:12828–12839. doi:10.1128/JVI.79.20.12828-12839.2005.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Lin RJ,
    2. Chang BL,
    3. Yu HP,
    4. Liao CL,
    5. Lin YL
    . 2006. Blocking of interferon-induced Jak-Stat signaling by Japanese encephalitis virus NS5 through a protein tyrosine phosphatase-mediated mechanism. J Virol 80:5908–5918. doi:10.1128/JVI.02714-05.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Kroschewski H,
    2. Allison SL,
    3. Heinz FX,
    4. Mandl CW
    . 2003. Role of heparan sulfate for attachment and entry of tick-borne encephalitis virus. Virology 308:92–100. doi:10.1016/S0042-6822(02)00097-1.
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    1. Barkhash AV,
    2. Perelygin AA,
    3. Babenko VN,
    4. Brinton MA,
    5. Voevoda MI
    . 2012. Single nucleotide polymorphism in the promoter region of the CD209 gene is associated with human predisposition to severe forms of tick-borne encephalitis. Antiviral Res 93:64–68. doi:10.1016/j.antiviral.2011.10.017.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Protopopova EV,
    2. Sorokin AV,
    3. Konovalova SN,
    4. Kachko AV,
    5. Netesov SV,
    6. Loktev VB
    . 1999. Human laminin binding protein as a cell receptor for the tick-borne encephalitis virus. Zentralbl Bakteriol 289:632–638. doi:10.1016/S0934-8840(99)80021-8.
    OpenUrlCrossRef
  33. 33.↵
    1. Fernandez-Garcia MD,
    2. Mazzon M,
    3. Jacobs M,
    4. Amara A
    . 2009. Pathogenesis of flavivirus infections: using and abusing the host cell. Cell Host Microbe 5:318–328. doi:10.1016/j.chom.2009.04.001.
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    1. Overby AK,
    2. Popov VL,
    3. Niedrig M,
    4. Weber F
    . 2010. Tick-borne encephalitis virus delays interferon induction and hides its double-stranded RNA in intracellular membrane vesicles. J Virol 84:8470–8483. doi:10.1128/JVI.00176-10.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Ostfeld RS,
    2. Brunner JL
    . 2015. Climate change and Ixodes tick-borne diseases of humans. Philos Trans R Soc Lond B Biol Sci 370:20140051. doi:10.1098/rstb.2014.0051.
    OpenUrlCrossRefPubMed
  36. 36.↵
    1. Pfeffer M,
    2. Dobler G
    . 2010. Emergence of zoonotic arboviruses by animal trade and migration. Parasit Vectors 3:35. doi:10.1186/1756-3305-3-35.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Dantas-Torres F
    . 2015. Climate change, biodiversity, ticks and tick-borne diseases: the butterfly effect. Int J Parasitol Parasites Wildl 4:452–461. doi:10.1016/j.ijppaw.2015.07.001.
    OpenUrlCrossRef
  38. 38.↵
    1. Földvári G,
    2. Široký P,
    3. Szekeres S,
    4. Majoros G,
    5. Sprong H
    . 2016. Dermacentor reticulatus: a vector on the rise. Parasit Vectors 9:314. doi:10.1186/s13071-016-1599-x.
    OpenUrlCrossRef
  39. 39.↵
    1. Medlock JM,
    2. Hansford KM,
    3. Bormane A,
    4. Derdakova M,
    5. Estrada-Peña A,
    6. George JC,
    7. Golovljova I,
    8. Jaenson TG,
    9. Jensen JK,
    10. Jensen PM,
    11. Kazimirova M,
    12. Oteo JA,
    13. Papa A,
    14. Pfister K,
    15. Plantard O,
    16. Randolph SE,
    17. Rizzoli A,
    18. Santos-Silva MM,
    19. Sprong H,
    20. Vial L,
    21. Hendrickx G,
    22. Zeller H,
    23. Van Bortel W
    . 2013. Driving forces for changes in geographical distribution of Ixodes ricinus ticks in Europe. Parasit Vectors 6:1. doi:10.1186/1756-3305-6-1.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Waldenström J,
    2. Lundkvist A,
    3. Falk KI,
    4. Garpmo U,
    5. Bergström S,
    6. Lindegren G,
    7. Sjöstedt A,
    8. Mejlon H,
    9. Fransson T,
    10. Haemig PD,
    11. Olsen B
    . 2007. Migrating birds and tickborne encephalitis virus. Emerg Infect Dis 13:1215–1218. doi:10.3201/eid1308.061416.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Kovalev SY,
    2. Chernykh DN,
    3. Kokorev VS,
    4. Snitkovskaya TE,
    5. Romanenko VV
    . 2009. Origin and distribution of tick-borne encephalitis virus strains of the Siberian subtype in the Middle Urals, the north-west of Russia and the Baltic countries. J Gen Virol 90:2884–2892. doi:10.1099/vir.0.012419-0.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Korenberg EI,
    2. Kovalevskii YV
    . 1999. Main features of tick-borne encephalitis eco-epidemiology in Russia. Zentralbl Bakteriol 289:525–539. doi:10.1016/S0934-8840(99)80006-1.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Piesman J,
    2. Eisen L
    . 2008. Prevention of tick-borne diseases. Annu Rev Entomol 53:323–343. doi:10.1146/annurev.ento.53.103106.093429.
    OpenUrlCrossRefPubMedWeb of Science
  44. 44.↵
    1. Labuda M,
    2. Danielova V,
    3. Jones LD,
    4. Nuttall PA
    . 1993. Amplification of tick-borne encephalitis virus infection during co-feeding of ticks. Med Vet Entomol 7:339–342. doi:10.1111/j.1365-2915.1993.tb00702.x.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Labuda M,
    2. Jones LD,
    3. Williams T,
    4. Danielova V,
    5. Nuttall PA
    . 1993. Efficient transmission of tick-borne encephalitis virus between co-feeding ticks. J Med Entomol 30:295–299. doi:10.1093/jmedent/30.1.295.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Labuda M,
    2. Nuttall PA,
    3. Kozuch O,
    4. Elecková E,
    5. Williams T,
    6. Zuffová E,
    7. Sabó A
    . 1993. Non-viraemic transmission of tick-borne encephalitis virus: a mechanism for arbovirus survival in nature. Experientia 49:802–805. doi:10.1007/BF01923553.
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    1. Labuda M,
    2. Austyn JM,
    3. Zuffova E,
    4. Kozuch O,
    5. Fuchsberger N,
    6. Lysy J,
    7. Nuttall PA
    . 1996. Importance of localized skin infection in tick-borne encephalitis virus transmission. Virology 219:357–366. doi:10.1006/viro.1996.0261.
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    1. Labuda M,
    2. Kozuch O,
    3. Zuffová E,
    4. Elecková E,
    5. Hails RS,
    6. Nuttall PA
    . 1997. Tick-borne encephalitis virus transmission between ticks co-feeding on specific immune natural rodent hosts. Virology 235:138–143. doi:10.1006/viro.1997.8622.
    OpenUrlCrossRefPubMedWeb of Science
  49. 49.↵
    1. Randolph SE
    . 2011. Transmission of tick-borne pathogens between co-feeding ticks: Milan Labuda’s enduring paradigm. Ticks Tick Borne Dis 2:179–182. doi:10.1016/j.ttbdis.2011.07.004.
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. Havlíková S,
    2. Ličková M,
    3. Klempa B
    . 2013. Non-viraemic transmission of tick-borne viruses. Acta Virol 57:123–129. doi:10.4149/av_2013_02_123.
    OpenUrlCrossRefPubMed
  51. 51.↵
    1. Nuttall PA,
    2. Labuda M
    . 2003. Dynamics of infection in tick vectors and at the tick-host interface. Adv Virus Res 60:233–272. doi:10.1016/S0065-3527(03)60007-2.
    OpenUrlCrossRefPubMedWeb of Science
  52. 52.↵
    1. Nuttall PM,
    2. Labuda M
    . 2008. Saliva-assisted transmission of tick-borne pathogens, p 205–219. In Bowman AS, Nuttall PA (ed), Ticks: biology, disease and control. Cambridge University Press, Cambridge, United Kingdom.
  53. 53.↵
    1. Danielová V,
    2. Holubová J,
    3. Pejcoch M,
    4. Daniel M
    . 2002. Potential significance of transovarial transmission in the circulation of tick-borne encephalitis virus. Folia Parasitol (Praha) 49:323–325. doi:10.14411/fp.2002.060.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Randolph SE
    . 2004. Tick ecology: processes and patterns behind the epidemiological risk posed by ixodid ticks as vectors. Parasitology 129(Suppl):S37–S65. doi:10.1017/S0031182004004925.
    OpenUrlCrossRefPubMedWeb of Science
  55. 55.↵
    1. Achazi K,
    2. Růžek D,
    3. Donoso-Mantke O,
    4. Schlegel M,
    5. Ali HS,
    6. Wenk M,
    7. Schmidt-Chanasit J,
    8. Ohlmeyer L,
    9. Rühe F,
    10. Vor T,
    11. Kiffner C,
    12. Kallies R,
    13. Ulrich RG,
    14. Niedrig M
    . 2011. Rodents as sentinels for the prevalence of tick-borne encephalitis virus. Vector Borne Zoonotic Dis 11:641–647. doi:10.1089/vbz.2010.0236.
    OpenUrlCrossRefPubMedWeb of Science
  56. 56.↵
    1. Knap N,
    2. Korva M,
    3. Dolinšek V,
    4. Sekirnik M,
    5. Trilar T,
    6. Avšič-Županc T
    . 2012. Patterns of tick-borne encephalitis virus infection in rodents in Slovenia. Vector Borne Zoonotic Dis 12:236–242. doi:10.1089/vbz.2011.0728.
    OpenUrlCrossRefPubMedWeb of Science
  57. 57.↵
    1. Pintér R,
    2. Madai M,
    3. Horváth G,
    4. Németh V,
    5. Oldal M,
    6. Kemenesi G,
    7. Dallos B,
    8. Bányai K,
    9. Jakab F
    . 2014. Molecular detection and phylogenetic analysis of tick-borne encephalitis virus in rodents captured in the Transdanubian region of Hungary. Vector Borne Zoonotic Dis 14:621–624. doi:10.1089/vbz.2013.1479.
    OpenUrlCrossRef
  58. 58.↵
    1. Tonteri E,
    2. Jääskeläinen AE,
    3. Tikkakoski T,
    4. Voutilainen L,
    5. Niemimaa J,
    6. Henttonen H,
    7. Vaheri A,
    8. Vapalahti O
    . 2011. Tick-borne encephalitis virus in wild rodents in winter, Finland, 2008-2009. Emerg Infect Dis 17:72–75. doi:10.3201/eid1701.100051.
    OpenUrlCrossRefPubMed
  59. 59.↵
    1. Carpi G,
    2. Bertolotti L,
    3. Rosati S,
    4. Rizzoli A
    . 2009. Prevalence and genetic variability of tick-borne encephalitis virus in host-seeking Ixodes ricinus in northern Italy. J Gen Virol 90:2877–2883. doi:10.1099/vir.0.013367-0.
    OpenUrlCrossRefPubMedWeb of Science
  60. 60.↵
    1. Durmiši E,
    2. Knap N,
    3. Saksida A,
    4. Trilar T,
    5. Duh D,
    6. Avšič-Županc T
    . 2011. Prevalence and molecular characterization of tick-borne encephalitis virus in Ixodes ricinus ticks collected in Slovenia. Vector Borne Zoonotic Dis 11:659–664. doi:10.1089/vbz.2010.0054.
    OpenUrlCrossRefPubMedWeb of Science
  61. 61.↵
    1. Charrel RN,
    2. Attoui H,
    3. Butenko AM,
    4. Clegg JC,
    5. Deubel V,
    6. Frolova TV,
    7. Gould EA,
    8. Gritsun TS,
    9. Heinz FX,
    10. Labuda M,
    11. Lashkevich VA,
    12. Loktev V,
    13. Lundkvist A,
    14. Lvov DV,
    15. Mandl CW,
    16. Niedrig M,
    17. Papa A,
    18. Petrov VS,
    19. Plyusnin A,
    20. Randolph S,
    21. Süss J,
    22. Zlobin VI,
    23. de Lamballerie X
    . 2004. Tick-borne virus diseases of human interest in Europe. Clin Microbiol Infect 10:1040–1055. doi:10.1111/j.1469-0691.2004.01022.x.
    OpenUrlCrossRefPubMedWeb of Science
  62. 62.↵
    1. Jaenson TG,
    2. Jaenson DG,
    3. Eisen L,
    4. Petersson E,
    5. Lindgren E
    . 2012. Changes in the geographical distribution and abundance of the tick Ixodes ricinus during the past 30 years in Sweden. Parasit Vectors 5:8. doi:10.1186/1756-3305-5-8.
    OpenUrlCrossRefPubMed
  63. 63.↵
    1. Donoso Mantke O,
    2. Schädler R,
    3. Niedrig M
    . 2008. A survey on cases of tick-borne encephalitis in European countries. Euro Surveill 13(17):pii=18848. https://www.eurosurveillance.org/content/10.2807/ese.13.17.18848-en.
    OpenUrl
  64. 64.↵
    1. Kunze U
    , ISW-TBE. 2006. Tick-borne encephalitis—a European health challenge. Conference report of the 8th meeting of the International Scientific Working Group on Tick-Borne Encephalitis (ISW TBE). Wien Med Wochenschr 156:376–378. doi:10.1007/s10354-006-0318-1.
    OpenUrlCrossRefPubMed
  65. 65.↵
    1. Smorodintsev AA
    . 1958. Tick-borne spring-summer encephalitis. Prog Med Virol 1:210–247.
    OpenUrlPubMed
  66. 66.↵
    1. Süss J
    . 2008. Tick-borne encephalitis in Europe and beyond—the epidemiological situation as of 2007. Euro Surveill 13(26):pii=18916. https://www.eurosurveillance.org/content/10.2807/ese.13.26.18916-en.
    OpenUrl
  67. 67.↵
    1. Růžek D,
    2. Yakimenko VV,
    3. Karan LS,
    4. Tkachev SE
    . 2010. Omsk haemorrhagic fever. Lancet 376:2104–2113. doi:10.1016/S0140-6736(10)61120-8.
    OpenUrlCrossRefPubMed
  68. 68.↵
    1. Holbrook MR
    . 2012. Kyasanur Forest disease. Antiviral Res 96:353–362. doi:10.1016/j.antiviral.2012.10.005.
    OpenUrlCrossRefPubMed
  69. 69.↵
    1. Mehla R,
    2. Kumar SR,
    3. Yadav P,
    4. Barde PV,
    5. Yergolkar PN,
    6. Erickson BR,
    7. Carroll SA,
    8. Mishra AC,
    9. Nichol ST,
    10. Mourya DT
    . 2009. Recent ancestry of Kyasanur Forest disease virus. Emerg Infect Dis 15:1431–1437. doi:10.3201/eid1509.080759.
    OpenUrlCrossRefPubMed
  70. 70.↵
    1. Gilbert L
    . 2016. Louping ill virus in the UK: a review of the hosts, transmission and ecological consequences of control. Exp Appl Acarol 68:363–374. doi:10.1007/s10493-015-9952-x.
    OpenUrlCrossRef
  71. 71.↵
    1. Ebel GD
    . 2010. Update on Powassan virus: emergence of a North American tick-borne flavivirus. Annu Rev Entomol 55:95–110. doi:10.1146/annurev-ento-112408-085446.
    OpenUrlCrossRefPubMedWeb of Science
  72. 72.↵
    1. Alkadhi H,
    2. Kollias SS
    . 2000. MRI in tick-borne encephalitis. Neuroradiology 42:753–755. doi:10.1007/s002340000396.
    OpenUrlCrossRefPubMed
  73. 73.↵
    1. Bender A,
    2. Schulte-Altedorneburg G,
    3. Walther EU,
    4. Pfister H-W
    . 2005. Severe tick borne encephalitis with simultaneous brain stem, bithalamic, and spinal cord involvement documented by MRI. J Neurol Neurosurg Psychiatry 76:135–137. doi:10.1136/jnnp.2004.040469.
    OpenUrlAbstract/FREE Full Text
  74. 74.↵
    1. Gelpi E,
    2. Preusser M,
    3. Garzuly F,
    4. Holzmann H,
    5. Heinz FX,
    6. Budka H
    . 2005. Visualization of Central European tick-borne encephalitis infection in fatal human cases. J Neuropathol Exp Neurol 64:506–512. doi:10.1093/jnen/64.6.506.
    OpenUrlCrossRefPubMed
  75. 75.↵
    1. Pfefferkorn T,
    2. Feddersen B,
    3. Schulte-Altedorneburg G,
    4. Linn J,
    5. Pfister HW
    . 2007. Tick-borne encephalitis with polyradiculitis documented by MRI. Neurology 68:1232–1233. doi:10.1212/01.wnl.0000259065.58968.10.
    OpenUrlCrossRef
  76. 76.↵
    1. Santos RI,
    2. Hermance ME,
    3. Gelman BB,
    4. Thangamani S
    . 2016. Spinal cord ventral horns and lymphoid organ involvement in Powassan virus infection in a mouse model. Viruses 8:220. doi:10.3390/v8080220.
    OpenUrlCrossRef
  77. 77.↵
    1. Qattan I,
    2. Akbar N,
    3. Afif H,
    4. Azmah SA,
    5. Al-Khateeb T,
    6. Zaki A,
    7. Ai-Hamdan N,
    8. Fontaine RE
    . 1996. A novel flavivirus: Makkah region 1994–1996. Saudi Epidemiol Bull 3:1–3.
    OpenUrl
  78. 78.↵
    1. Zaki AM
    . 1997. Isolation of a flavivirus related to the tick-borne encephalitis complex from human cases in Saudi Arabia. Trans R Soc Trop Med Hyg 91:179–181. doi:10.1016/S0035-9203(97)90215-7.
    OpenUrlCrossRefPubMed
  79. 79.↵
    1. Alzahrani AG,
    2. Al Shaiban HM,
    3. Al Mazroa MA,
    4. Al-Hayani O,
    5. MacNeil A,
    6. Rollin PE,
    7. Memish ZA
    . 2010. Alkhurma hemorrhagic fever in humans, Najran, Saudi Arabia. Emerg Infect Dis 16:1882–1888. doi:10.3201/eid1612.100417.
    OpenUrlCrossRefPubMed
  80. 80.↵
    1. Murhekar MV,
    2. Kasabi GS,
    3. Mehendale SM,
    4. Mourya DT,
    5. Yadav PD,
    6. Tandale BV
    . 2015. On the transmission pattern of Kyasanur Forest disease (KFD) in India. Infect Dis Poverty 4:37. doi:10.1186/s40249-015-0066-9.
    OpenUrlCrossRef
  81. 81.↵
    1. Novitskiy VS
    . 1948. Pathologic anatomy of spring-summer fever in Omsk region. Proc Omsk Inst Epidemiol Microbiol Gig 13:97.
    OpenUrl
  82. 82.↵
    1. Hermance ME,
    2. Santos RI,
    3. Kelly BC,
    4. Valbuena G,
    5. Thangamani S
    . 2016. Immune cell targets of infection at the tick-skin interface during Powassan virus transmission. PLoS One 11:e0155889. doi:10.1371/journal.pone.0155889.
    OpenUrlCrossRef
  83. 83.↵
    1. Suthar MS,
    2. Aguirre S,
    3. Fernandez-Sesma A
    . 2013. Innate immune sensing of flaviviruses. PLoS Pathog 9:e1003541. doi:10.1371/journal.ppat.1003541.
    OpenUrlCrossRefPubMed
  84. 84.↵
    1. Bogovic P,
    2. Strle F
    . 2015. Tick-borne encephalitis: a review of epidemiology, clinical characteristics, and management. World J Clin Cases 3:430–441. doi:10.12998/wjcc.v3.i5.430.
    OpenUrlCrossRefPubMed
  85. 85.↵
    1. McMinn PC
    . 1997. The molecular basis of virulence of the encephalitogenic flaviviruses. J Gen Virol 78:2711–2722. doi:10.1099/0022-1317-78-11-2711.
    OpenUrlCrossRefPubMedWeb of Science
  86. 86.↵
    1. Bílý T,
    2. Palus M,
    3. Eyer L,
    4. Elsterová J,
    5. Vancová M,
    6. Růžek D
    . 2015. Electron tomography analysis of tick-borne encephalitis virus infection in human neurons. Sci Rep 5:10745. doi:10.1038/srep10745.
    OpenUrlCrossRefPubMed
  87. 87.↵
    1. Růzek D,
    2. Vancová M,
    3. Tesarová M,
    4. Ahantarig A,
    5. Kopecký J,
    6. Grubhoffer L
    . 2009. Morphological changes in human neural cells following tick-borne encephalitis virus infection. J Gen Virol 90:1649–1658. doi:10.1099/vir.0.010058-0.
    OpenUrlCrossRefPubMedWeb of Science
  88. 88.↵
    1. Potokar M,
    2. Korva M,
    3. Jorgačevski J,
    4. Avšič-Županc T,
    5. Zorec R
    . 2014. Tick-borne encephalitis virus infects rat astrocytes but does not affect their viability. PLoS One 9:e86219. doi:10.1371/journal.pone.0086219.
    OpenUrlCrossRef
  89. 89.↵
    1. Lindqvist R,
    2. Mundt F,
    3. Gilthorpe JD,
    4. Wölfel S,
    5. Gekara NO,
    6. Kröger A,
    7. Överby AK
    . 2016. Fast type I interferon response protects astrocytes from flavivirus infection and virus-induced cytopathic effects. J Neuroinflammation 13:277. doi:10.1186/s12974-016-0748-7.
    OpenUrlCrossRef
  90. 90.↵
    1. Tigabu B,
    2. Juelich T,
    3. Bertrand J,
    4. Holbrook MR
    . 2009. Clinical evaluation of highly pathogenic tick-borne flavivirus infection in the mouse model. J Med Virol 81:1261–1269. doi:10.1002/jmv.21524.
    OpenUrlCrossRefPubMed
  91. 91.↵
    1. Grygorczuk S,
    2. Osada J,
    3. Parczewski M,
    4. Moniuszko A,
    5. Świerzbińska R,
    6. Kondrusik M,
    7. Czupryna P,
    8. Dunaj J,
    9. Dąbrowska M,
    10. Pancewicz S
    . 2016. The expression of the chemokine receptor CCR5 in tick-borne encephalitis. J Neuroinflammation 13:45. doi:10.1186/s12974-016-0511-0.
    OpenUrlCrossRefPubMed
  92. 92.↵
    1. Kindberg E,
    2. Vene S,
    3. Mickiene A,
    4. Lundkvist Å,
    5. Lindquist L,
    6. Svensson L
    . 2011. A functional Toll-like receptor 3 gene (TLR3) may be a risk factor for tick-borne encephalitis virus (TBEV) infection. J Infect Dis 203:523–528. doi:10.1093/infdis/jiq082.
    OpenUrlCrossRefPubMed
  93. 93.↵
    1. Belikov SI,
    2. Leonova GN,
    3. Kondratov IG,
    4. Romanova EV,
    5. Pavlenko EV
    . 2010. Coding nucleotide sequences of tick-borne encephalitis virus strains isolated from human blood without clinical symptoms of infection. Genetika 46:356–363.
    OpenUrlPubMed
  94. 94.↵
    1. Belikov SI,
    2. Kondratov IG,
    3. Potapova UV,
    4. Leonova GN
    . 2014. The relationship between the structure of the tick-borne encephalitis virus strains and their pathogenic properties. PLoS One 9:e94946. doi:10.1371/journal.pone.0094946.
    OpenUrlCrossRef
  95. 95.↵
    1. Fialová A,
    2. Cimburek Z,
    3. Iezzi G,
    4. Kopecký J
    . 2010. Ixodes ricinus tick saliva modulates tick-borne encephalitis virus infection of dendritic cells. Microbes Infect 12:580–585. doi:10.1016/j.micinf.2010.03.015.
    OpenUrlCrossRefPubMed
  96. 96.↵
    1. Hermance ME,
    2. Thangamani S
    . 2014. Proinflammatory cytokines and chemokines at the skin interface during Powassan virus transmission. J Invest Dermatol 134:2280–2283. doi:10.1038/jid.2014.150.
    OpenUrlCrossRefPubMed
  97. 97.↵
    1. Sadler AJ,
    2. Williams BR
    . 2007. Structure and function of the protein kinase R. Curr Top Microbiol Immunol 316:253–292.
    OpenUrlCrossRefPubMedWeb of Science
  98. 98.↵
    1. Kawai T,
    2. Akira S
    . 2006. Innate immune recognition of viral infection. Nat Immunol 7:131–137. doi:10.1038/ni1303.
    OpenUrlCrossRefPubMedWeb of Science
  99. 99.↵
    1. Wu SJ,
    2. Grouard-Vogel G,
    3. Sun W,
    4. Mascola JR,
    5. Brachtel E,
    6. Putvatana R,
    7. Louder MK,
    8. Filgueira L,
    9. Marovich MA,
    10. Wong HK,
    11. Blauvelt A,
    12. Murphy GS,
    13. Robb ML,
    14. Innes BL,
    15. Birx DL,
    16. Hayes CG,
    17. Frankel SS
    . 2000. Human skin Langerhans cells are targets of dengue virus infection. Nat Med 6:816–820. doi:10.1038/77553.
    OpenUrlCrossRefPubMedWeb of Science
  100. 100.↵
    1. Robertson SJ,
    2. Mitzel DN,
    3. Taylor RT,
    4. Best SM,
    5. Bloom ME
    . 2009. Tick-borne flaviviruses: dissecting host immune responses and virus countermeasures. Immunol Res 43:172–186. doi:10.1007/s12026-008-8065-6.
    OpenUrlCrossRefPubMedWeb of Science
  101. 101.↵
    1. Robertson SJ,
    2. Lubick KJ,
    3. Freedman BA,
    4. Carmody AB,
    5. Best SM
    . 2014. Tick-borne flaviviruses antagonize both IRF-1 and type I IFN signaling to inhibit dendritic cell function. J Immunol 192:2744–2755. doi:10.4049/jimmunol.1302110.
    OpenUrlAbstract/FREE Full Text
  102. 102.↵
    1. Werme K,
    2. Wigerius M,
    3. Johansson M
    . 2008. Tick-borne encephalitis virus NS5 associates with membrane protein scribble and impairs interferon-stimulated JAK-STAT signalling. Cell Microbiol 10:696–712. doi:10.1111/j.1462-5822.2007.01076.x.
    OpenUrlCrossRefPubMed
  103. 103.↵
    1. Laurent-Rolle M,
    2. Boer EF,
    3. Lubick KJ,
    4. Wolfinbarger JB,
    5. Carmody AB,
    6. Rockx B,
    7. Liu W,
    8. Ashour J,
    9. Shupert WL,
    10. Holbrook MR,
    11. Barrett AD,
    12. Mason PW,
    13. Bloom ME,
    14. García-Sastre A,
    15. Khromykh AA,
    16. Best SM
    . 2010. The NS5 protein of the virulent West Nile virus NY99 strain is a potent antagonist of type I interferon-mediated JAK-STAT signaling. J Virol 84:3503–3515. doi:10.1128/JVI.01161-09.
    OpenUrlAbstract/FREE Full Text
  104. 104.↵
    1. Masson F,
    2. Mount AM,
    3. Wilson NS,
    4. Belz GT
    . 2008. Dendritic cells: driving the differentiation programme of T cells in viral infections. Immunol Cell Biol 86:333–342. doi:10.1038/icb.2008.15.
    OpenUrlCrossRefPubMed
  105. 105.↵
    1. Blom K,
    2. Braun M,
    3. Pakalniene J,
    4. Dailidyte L,
    5. Béziat V,
    6. Lampen MH,
    7. Klingström J,
    8. Lagerqvist N,
    9. Kjerstadius T,
    10. Michaëlsson J,
    11. Lindquist L,
    12. Ljunggren HG,
    13. Sandberg JK,
    14. Mickiene A,
    15. Gredmark-Russ S
    . 2015. Specificity and dynamics of effector and memory CD8 T cell responses in human tick-borne encephalitis virus infection. PLoS Pathog 11:e1004622. doi:10.1371/journal.ppat.1004622.
    OpenUrlCrossRefPubMed
  106. 106.↵
    1. Blom K,
    2. Braun M,
    3. Pakalniene J,
    4. Lunemann S,
    5. Enqvist M,
    6. Dailidyte L,
    7. Schaffer M,
    8. Lindquist L,
    9. Mickiene A,
    10. Michaëlsson J,
    11. Ljunggren HG,
    12. Gredmark-Russ S
    . 2016. NK cell responses to human tick-borne encephalitis virus infection. J Immunol 197:2762–2771. doi:10.4049/jimmunol.1600950.
    OpenUrlAbstract/FREE Full Text
  107. 107.↵
    1. Johnson BW
    . 2016. Neurotropic flaviviruses, p 229–258. In Reiss CS (ed), Neurotropic viral infections, 2nd ed. Springer, Cham, Switzerland.
  108. 108.↵
    World Health Organization. 2011. Vaccines against tick-borne encephalitis: WHO position paper. Wkly Epidemiol Rec 86:241–256.
    OpenUrlPubMed
  109. 109.↵
    1. Heinz FX,
    2. Stiasny K,
    3. Holzmann H,
    4. Grgic-Vitek M,
    5. Kriz B,
    6. Essl A,
    7. Kundi M
    . 2013. Vaccination and tick-borne encephalitis, central Europe. Emerg Infect Dis 19:69–76. doi:10.3201/eid1901.120458.
    OpenUrlCrossRefPubMed
  110. 110.↵
    1. Donoso Mantke O,
    2. Escadafal C,
    3. Niedrig M,
    4. Pfeffer M
    , on behalf of the Working Group for Tick-Borne Encephalitis Virus. 2011. Tick-borne encephalitis in Europe, 2007 to 2009. Euro Surveill 16(39):pii=19976. doi:10.2807/ese.16.39.19976-en.
    OpenUrlCrossRef
  111. 111.↵
    1. Schuler M,
    2. Zimmermann H,
    3. Altpeter E,
    4. Heininger U
    . 2014. Epidemiology of tick-borne encephalitis in Switzerland, 2005 to 2011. Euro Surveill 19(13):pii=20756. doi:10.2807/1560-7917.ES2014.19.13.20756.
    OpenUrlCrossRef
  112. 112.↵
    1. Heinz FX,
    2. Stiasny K,
    3. Holzmann H,
    4. Kundi M,
    5. Sixl W,
    6. Wenk M,
    7. Kainz W,
    8. Essl A,
    9. Kunz C
    . 2015. Emergence of tick-borne encephalitis in new endemic areas in Austria: 42 years of surveillance. Euro Surveill 20(13):pii=21077. doi:10.2807/1560-7917.ES2015.20.13.21077.
    OpenUrlCrossRef
  113. 113.↵
    1. Kluger G,
    2. Schöttler A,
    3. Waldvogel K,
    4. Nadal D,
    5. Hinrichs W,
    6. Wündisch GF,
    7. Laub MC
    . 1995. Tickborne encephalitis despite specific immunoglobulin prophylaxis. Lancet 346:1502. doi:10.1016/S0140-6736(95)92527-9.
    OpenUrlCrossRefPubMed
  114. 114.↵
    1. Waldvogel K,
    2. Bossart W,
    3. Huisman T,
    4. Boltshauser E,
    5. Nadal D
    . 1996. Severe tick-borne encephalitis following passive immunization. Eur J Pediatr 155:775–779. doi:10.1007/BF02002905.
    OpenUrlCrossRefPubMed
  115. 115.↵
    1. Chrdle A,
    2. Chmelík V,
    3. Růžek D
    . 2016. Tick-borne encephalitis: what travellers should know when visiting an endemic country. Hum Vaccin Immunother 12:2694–2699. doi:10.1080/21645515.2016.1218098.
    OpenUrlCrossRef
  116. 116.↵
    1. Bartel DP
    . 2009. MicroRNAs: target recognition and regulatory functions. Cell 136:215–233. doi:10.1016/j.cell.2009.01.002.
    OpenUrlCrossRefPubMedWeb of Science
  117. 117.↵
    1. Behura SK
    . 2007. Insect microRNAs: structure, function and evolution. Insect Biochem Mol Biol 37:3–9. doi:10.1016/j.ibmb.2006.10.006.
    OpenUrlCrossRefPubMedWeb of Science
  118. 118.↵
    1. Bonaldo MC,
    2. Mello SM,
    3. Trindade GF,
    4. Rangel AA,
    5. Duarte AS,
    6. Oliveira PJ,
    7. Freire MS,
    8. Kubelka CF,
    9. Galler R
    . 2007. Construction and characterization of recombinant flaviviruses bearing insertions between E and NS1 genes. Virol J 4:115. doi:10.1186/1743-422X-4-115.
    OpenUrlCrossRefPubMed
  119. 119.↵
    1. Teterina NL,
    2. Liu G,
    3. Maximova OA,
    4. Pletnev AG
    . 2014. Silencing of neurotropic flavivirus replication in the central nervous system by combining multiple microRNA target insertions in two distinct viral genome regions. Virology 456–457:247–258. doi:10.1016/j.virol.2014.04.001.
    OpenUrlCrossRef
  120. 120.↵
    1. Teterina NL,
    2. Maximova OA,
    3. Kenney H,
    4. Liu G,
    5. Pletnev AG
    . 2016. MicroRNA-based control of tick-borne flavivirus neuropathogenesis: challenges and perspectives. Antiviral Res 127:57–67. doi:10.1016/j.antiviral.2016.01.003.
    OpenUrlCrossRef
  121. 121.↵
    1. Heiss BL,
    2. Maximova OA,
    3. Thach DC,
    4. Speicher JM,
    5. Pletnev AG
    . 2012. MicroRNA targeting of neurotropic flavivirus: effective control of virus escape and reversion to neurovirulent phenotype. J Virol 86:5647–5659. doi:10.1128/JVI.07125-11.
    OpenUrlAbstract/FREE Full Text
  122. 122.↵
    1. Heiss BL,
    2. Maximova OA,
    3. Pletnev AG
    . 2011. Insertion of microRNA targets into the flavivirus genome alters its highly neurovirulent phenotype. J Virol 85:1464–1472. doi:10.1128/JVI.02091-10.
    OpenUrlAbstract/FREE Full Text
  123. 123.↵
    1. Rumyantsev AA,
    2. Goncalvez AP,
    3. Giel-Moloney M,
    4. Catalan J,
    5. Liu Y,
    6. Gao QS,
    7. Almond J,
    8. Kleanthous H,
    9. Pugachev KV
    . 2013. Single-dose vaccine against tick-borne encephalitis. Proc Natl Acad Sci U S A 110:13103–13108. doi:10.1073/pnas.1306245110.
    OpenUrlAbstract/FREE Full Text
  124. 124.↵
    1. Labuda M,
    2. Trimnell AR,
    3. Licková M,
    4. Kazimírová M,
    5. Davies GM,
    6. Lissina O,
    7. Hails RS,
    8. Nuttall PA
    . 2006. An antivector vaccine protects against a lethal vector-borne pathogen. PLoS Pathog 2:e27. doi:10.1371/journal.ppat.0020027.
    OpenUrlCrossRefPubMed
  125. 125.↵
    1. Havlíková S,
    2. Roller L,
    3. Koci J,
    4. Trimnell AR,
    5. Kazimírová M,
    6. Klempa B,
    7. Nuttall PA
    . 2009. Functional role of 64P, the candidate transmission-blocking vaccine antigen from the tick, Rhipicephalus appendiculatus. Int J Parasitol 39:1485–1494. doi:10.1016/j.ijpara.2009.05.005.
    OpenUrlCrossRefPubMed
  126. 126.↵
    1. de La Fuente J,
    2. Kocan KM,
    3. Contreras M
    . 2015. Prevention and control strategies for ticks and pathogen transmission. Rev Sci Tech 34:249–264. doi:10.20506/rst.34.1.2357.
    OpenUrlCrossRef
  127. 127.↵
    1. Flint M,
    2. McMullan LK,
    3. Dodd KA,
    4. Bird BH,
    5. Khristova ML,
    6. Nichol ST,
    7. Spiropoulou CF
    . 2014. Inhibitors of the tick-borne, hemorrhagic fever-associated flaviviruses. Antimicrob Agents Chemother 58:3206–3216. doi:10.1128/AAC.02393-14.
    OpenUrlAbstract/FREE Full Text
  128. 128.↵
    1. Lo MK,
    2. Shi P-Y,
    3. Chen Y-L,
    4. Flint M,
    5. Spiropoulou CF
    . 2016. In vitro antiviral activity of adenosine analog NITD008 against tick-borne flaviviruses. Antiviral Res 130:46–49. doi:10.1016/j.antiviral.2016.03.013.
    OpenUrlCrossRef
  129. 129.↵
    1. Eyer L,
    2. Valdés JJ,
    3. Gil VA,
    4. Nencka R,
    5. Hřebabecký H,
    6. Šála M,
    7. Salát J,
    8. Černý J,
    9. Palus M,
    10. De Clercq E,
    11. Růžek D
    . 2015. Nucleoside inhibitors of tick-borne encephalitis virus. Antimicrob Agents Chemother 59:5483–5493. doi:10.1128/AAC.00807-15.
    OpenUrlAbstract/FREE Full Text
  130. 130.↵
    1. McLean DM,
    2. Donohue WL
    . 1959. Powassan virus: isolation of virus from a fatal case of encephalitis. Can Med Assoc J 80:708–711.
    OpenUrlPubMed
  131. 131.↵
    1. Goldfield M,
    2. Austin SM,
    3. Black HC,
    4. Taylor BF,
    5. Altman R
    . 1973. A non-fatal human case of Powassan virus encephalitis. Am J Trop Med Hyg 22:78–81. doi:10.4269/ajtmh.1973.22.78.
    OpenUrlFREE Full Text
  132. 132.↵
    1. Leonova GN,
    2. Isachkova LM,
    3. Baranov NI,
    4. Krugliak SP
    . 1980. Role of Powassan virus in the etiological structure of tick-borne encephalitis in the Primorsky Kray. Vopr Virusol 1980(2):173–176.
    OpenUrl
  133. 133.↵
    1. Thomas LA,
    2. Kennedy RC,
    3. Eklund CM
    . 1960. Isolation of a virus closely related to Powassan virus from Dermacentor andersoni collected along North Cache la Poudre River, Colo. Proc Soc Exp Biol Med 104:355–359. doi:10.3181/00379727-104-25836.
    OpenUrlCrossRefPubMed
  134. 134.↵
    1. Leonova GN,
    2. Kondratov IG,
    3. Ternovoi VA,
    4. Romanova EV,
    5. Protopopova EV,
    6. Chausov EV,
    7. Pavlenko EV,
    8. Ryabchikova EI,
    9. Belikov SI,
    10. Loktev VB
    . 2009. Characterization of Powassan viruses from Far Eastern Russia. Arch Virol 154:811–820. doi:10.1007/s00705-009-0376-y.
    OpenUrlCrossRefPubMedWeb of Science
  135. 135.↵
    1. Hicar MD,
    2. Edwards K,
    3. Bloch K
    . 2011. Powassan virus infection presenting as acute disseminated encephalomyelitis in Tennessee. Pediatr Infect Dis J 30:86–88. doi:10.1097/INF.0b013e3181f2f492.
    OpenUrlCrossRefPubMed
  136. 136.↵
    Centers for Disease Control and Prevention. 2001. Outbreak of Powassan encephalitis—Maine and Vermont, 1999-2001. MMWR Morb Mortal Wkly Rep 50:761–764.
    OpenUrlPubMed
  137. 137.↵
    1. Johnson DK,
    2. Staples JE,
    3. Sotir MJ,
    4. Warshauer DM,
    5. Davis JP
    . 2010. Tickborne Powassan virus infections among Wisconsin residents. WMJ 109:91–97.
    OpenUrlPubMed
  138. 138.↵
    1. Wilson MS,
    2. Wherrett BA,
    3. Mahdy MS
    . 1979. Powassan virus meningoencephalitis: a case report. Can Med Assoc J 121:320–323.
    OpenUrlPubMed
  139. 139.↵
    1. Gholam BI,
    2. Puksa S,
    3. Provias JP
    . 1999. Powassan encephalitis: a case report with neuropathology and literature review. CMAJ 161:1419–1422.
    OpenUrlFREE Full Text
  140. 140.↵
    1. Woodall JP,
    2. Roz A
    . 1977. Experimental milk-borne transmission of Powassan virus in the goat. Am J Trop Med Hyg 26:190–192. doi:10.4269/ajtmh.1977.26.190.
    OpenUrlAbstract/FREE Full Text
  141. 141.↵
    1. Calisher CH
    . 1994. Medically important arboviruses of the United States and Canada. Clin Microbiol Rev 7:89–116. doi:10.1128/CMR.7.1.89.
    OpenUrlAbstract/FREE Full Text
  142. 142.↵
    1. Ebel GD,
    2. Kramer LD
    . 2004. Short report: duration of tick attachment required for transmission of Powassan virus by deer ticks. Am J Trop Med Hyg 71:268–271. doi:10.4269/ajtmh.2004.71.3.0700268.
    OpenUrlAbstract/FREE Full Text
  143. 143.↵
    1. Tutolo JW,
    2. Staples JE,
    3. Sosa L,
    4. Bennett N
    . 2017. Notes from the field: Powassan virus disease in an infant—Connecticut, 2016. MMWR Morb Mortal Wkly Rep 66:408–409. doi:10.15585/mmwr.mm6615a3.
    OpenUrlCrossRef
  144. 144.↵
    1. Cook MJ
    . 2015. Lyme borreliosis: a review of data on transmission time after tick attachment. Int J Gen Med 8:1–8. doi:10.2147/IJGM.S73791.
    OpenUrlCrossRef
  145. 145.↵
    1. El Khoury MY,
    2. Camargo JF,
    3. Wormser GP
    . 2013. Changing epidemiology of Powassan encephalitis in North America suggests the emergence of the deer tick virus subtype. Expert Rev Anti Infect Ther 11:983–985. doi:10.1586/14787210.2013.837805.
    OpenUrlCrossRef
  146. 146.↵
    1. Leonova GN,
    2. Sorokina MN,
    3. Krugliak SP
    . 1991. The clinico-epidemiological characteristics of Powassan encephalitis in the southern Soviet Far East. Zh Mikrobiol Epidemiol Immunobiol 1991(3):35–39.
    OpenUrl
  147. 147.↵
    1. Romero JR,
    2. Simonsen KA
    . 2008. Powassan encephalitis and Colorado tick fever. Infect Dis Clin North Am 22:545–559. doi:10.1016/j.idc.2008.03.001.
    OpenUrlCrossRefPubMedWeb of Science
  148. 148.↵
    Centers for Disease Control and Prevention. 2011. West Nile virus disease and other arboviral diseases—United States, 2010. MMWR Morb Mortal Wkly Rep 60:1009–1013.
    OpenUrlPubMed
  149. 149.↵
    Centers for Disease Control and Prevention. 2012. West Nile virus disease and other arboviral diseases—United States, 2011. MMWR Morb Mortal Wkly Rep 61:510–514.
    OpenUrlCrossRefPubMed
  150. 150.↵
    Centers for Disease Control and Prevention. 2013. West Nile virus and other arboviral diseases—United States, 2012. MMWR Morb Mortal Wkly Rep 62:513–517.
    OpenUrlPubMed
  151. 151.↵
    1. Lindsey NP,
    2. Lehman JA,
    3. Staples JE,
    4. Fischer M
    , Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, CDC. 2014. West Nile virus and other arboviral diseases—United States, 2013. MMWR Morb Mortal Wkly Rep 63:521–526.
    OpenUrlPubMed
  152. 152.↵
    1. Lindsey NP,
    2. Lehman JA,
    3. Staples JE,
    4. Fischer M
    . 2015. West Nile virus and other nationally notifiable arboviral diseases—United States, 2014. MMWR Morb Mortal Wkly Rep 64:929–934. doi:10.15585/mmwr.mm6434a1.
    OpenUrlCrossRefPubMed
  153. 153.↵
    1. Krow-Lucal E,
    2. Lindsey NP,
    3. Lehman J,
    4. Fischer M,
    5. Staples JE
    . 2017. West Nile virus and other nationally notifiable arboviral diseases—United States, 2015. MMWR Morb Mortal Wkly Rep 66:51–55. doi:10.15585/mmwr.mm6602a3.
    OpenUrlCrossRef
  154. 154.↵
    1. Sung S,
    2. Wurcel AG,
    3. Whittier S,
    4. Kulas K,
    5. Kramer LD,
    6. Flam R,
    7. Roberts JK,
    8. Tsiouris S
    . 2013. Powassan meningoencephalitis, New York, New York, USA. Emerg Infect Dis 19:1504–1506. doi:10.3201/eid1909.121846.
    OpenUrlCrossRef
  155. 155.↵
    1. Neitzel DF,
    2. Lynfield R,
    3. Smith K
    . 2013. Powassan virus encephalitis, Minnesota, USA. Emerg Infect Dis 19:686. doi:10.3201/eid1904.121651.
    OpenUrlCrossRef
  156. 156.↵
    1. Birge J,
    2. Sonnesyn S
    . 2012. Powassan virus encephalitis, Minnesota, USA. Emerg Infect Dis 18:1669–1671. doi:10.3201/eid1810.120621.
    OpenUrlCrossRefPubMed
  157. 157.↵
    1. Piantadosi A,
    2. Rubin DB,
    3. McQuillen DP,
    4. Hsu L,
    5. Lederer PA,
    6. Ashbaugh CD,
    7. Duffalo C,
    8. Duncan R,
    9. Thon J,
    10. Bhattacharyya S,
    11. Basgoz N,
    12. Feske SK,
    13. Lyons JL
    . 2016. Emerging cases of Powassan virus encephalitis in New England: clinical presentation, imaging, and review of the literature. Clin Infect Dis 62:707–713. doi:10.1093/cid/civ1005.
    OpenUrlCrossRefPubMed
  158. 158.↵
    1. Raval M,
    2. Singhal M,
    3. Guerrero D,
    4. Alonto A
    . 2012. Powassan virus infection: case series and literature review from a single institution. BMC Res Notes 5:594. doi:10.1186/1756-0500-5-594.
    OpenUrlCrossRefPubMed
  159. 159.↵
    1. Deardorff ER,
    2. Nofchissey RA,
    3. Cook JA,
    4. Hope AG,
    5. Tsvetkova A,
    6. Talbot SL,
    7. Ebel GD
    . 2013. Powassan virus in mammals, Alaska and New Mexico, U.S.A., and Russia, 2004-2007. Emerg Infect Dis 19:2012–2016. doi:10.3201/eid1912.130319.
    OpenUrlCrossRefPubMed
  160. 160.↵
    1. Anonymous
    . 1962. United States-Mexico Border Public Health Association—conference report. Public Health Rep 77:140–146. doi:10.2307/4591552.
    OpenUrlCrossRef
  161. 161.↵
    1. Rossier E,
    2. Harrison RJ,
    3. Lemieux B
    . 1974. A case of Powassan virus encephalitis. Can Med Assoc J 110:1173–1174.
    OpenUrlPubMed
  162. 162.↵
    1. Fitch WM,
    2. Artsob H
    . 1990. Powassan encephalitis in New Brunswick. Can Fam Physician 36:1289–1290.
    OpenUrlPubMed
  163. 163.↵
    1. Kettyls GD,
    2. Verrall VM,
    3. Wilton LD,
    4. Clapp JB,
    5. Clarke DA,
    6. Rublee JD
    . 1972. Arbovirus infections in man in British Columbia. Can Med Assoc J 106:1175–1179.
    OpenUrlPubMed
  164. 164.↵
    1. McLean DM,
    2. Macpherson LW,
    3. Walker SJ,
    4. Funk G
    . 1960. Powassan virus: surveys of human and animal sera. Am J Public Health Nations Health 50:1539–1544. doi:10.2105/AJPH.50.10.1539.
    OpenUrlCrossRefPubMed
  165. 165.↵
    1. McLean DM,
    2. Walker SJ,
    3. Macpherson LW,
    4. Scholten TH,
    5. Ronald K,
    6. Wyllie JC,
    7. McQueen EJ
    . 1961. Powassan virus: investigations of possible natural cycles of infection. J Infect Dis 109:19–23. doi:10.1093/infdis/109.1.19.
    OpenUrlCrossRefPubMed
  166. 166.↵
    1. Braue K,
    2. Palm J,
    3. Kemperman M,
    4. Neitzel D,
    5. Vetter S
    . 2012. Seroprevalence of Powassan virus in healthy central and northeastern Minnesota blood donors. Minnesota Department of Health, St Paul, MN. http://conservancy.umn.edu/bitstream/handle/11299/123361/Braue.pdf?sequence=1&isAllowed=y.
  167. 167.↵
    1. Frost HM,
    2. Schotthoefer AM,
    3. Thomm AM,
    4. Dupuis AP, II,
    5. Kehl SC,
    6. Kramer LD,
    7. Fritsche TR,
    8. Harrington YA,
    9. Knox KK
    . 2017. Serologic evidence of Powassan virus infection in patients with suspected Lyme disease. Emerg Infect Dis 23:1384–1388. doi:10.3201/eid2308.161971.
    OpenUrlCrossRef
  168. 168.↵
    1. Nofchissey RA,
    2. Deardorff ER,
    3. Blevins TM,
    4. Anishchenko M,
    5. Bosco-Lauth A,
    6. Berl E,
    7. Lubelczyk C,
    8. Mutebi JP,
    9. Brault AC,
    10. Ebel GD,
    11. Magnarelli LA
    . 2013. Seroprevalence of Powassan virus in New England deer, 1979-2010. Am J Trop Med Hyg 88:1159–1162. doi:10.4269/ajtmh.12-0586.
    OpenUrlAbstract/FREE Full Text
  169. 169.↵
    Centers for Disease Control and Prevention. 1999. Outbreak of West Nile-like viral encephalitis—New York, 1999. MMWR Morb Mortal Wkly Rep 48:845–849.
    OpenUrlPubMed
  170. 170.↵
    1. Lanciotti RS,
    2. Roehrig JT,
    3. Deubel V,
    4. Smith J,
    5. Parker M,
    6. Steele K,
    7. Crise B,
    8. Volpe KE,
    9. Crabtree MB,
    10. Scherret JH,
    11. Hall RA,
    12. MacKenzie JS,
    13. Cropp CB,
    14. Panigrahy B,
    15. Ostlund E,
    16. Schmitt B,
    17. Malkinson M,
    18. Banet C,
    19. Weissman J,
    20. Komar N,
    21. Savage HM,
    22. Stone W,
    23. McNamara T,
    24. Gubler DJ
    . 1999. Origin of the West Nile virus responsible for an outbreak of encephalitis in the northeastern United States. Science 286:2333–2337. doi:10.1126/science.286.5448.2333.
    OpenUrlAbstract/FREE Full Text
  171. 171.↵
    1. Eisen RJ,
    2. Eisen L
    . 2018. The blacklegged tick, Ixodes scapularis: an increasing public health concern. Trends Parasitol 34:295–309. doi:10.1016/j.pt.2017.12.006.
    OpenUrlCrossRef
  172. 172.↵
    1. Telford SR, III,
    2. Armstrong PM,
    3. Katavolos P,
    4. Foppa I,
    5. Garcia AS,
    6. Wilson ML,
    7. Spielman A
    . 1997. A new tick-borne encephalitis-like virus infecting New England deer ticks, Ixodes dammini. Emerg Infect Dis 3:165–170. doi:10.3201/eid0302.970209.
    OpenUrlCrossRefPubMedWeb of Science
  173. 173.↵
    1. Mandl CW,
    2. Holzmann H,
    3. Kunz C,
    4. Heinz FX
    . 1993. Complete genomic sequence of Powassan virus: evaluation of genetic elements in tick-borne versus mosquito-borne flaviviruses. Virology 194:173–184. doi:10.1006/viro.1993.1247.
    OpenUrlCrossRefPubMed
  174. 174.↵
    1. Kuno G,
    2. Artsob H,
    3. Karabatsos N,
    4. Tsuchiya KR,
    5. Chang GJ
    . 2001. Genomic sequencing of deer tick virus and phylogeny of Powassan-related viruses of North America. Am J Trop Med Hyg 65:671–676. doi:10.4269/ajtmh.2001.65.671.
    OpenUrlAbstract
  175. 175.↵
    1. Beasley DW,
    2. Suderman MT,
    3. Holbrook MR,
    4. Barrett AD
    . 2001. Nucleotide sequencing and serological evidence that the recently recognized deer tick virus is a genotype of Powassan virus. Virus Res 79:81–89. doi:10.1016/S0168-1702(01)00330-6.
    OpenUrlCrossRefPubMedWeb of Science
  176. 176.↵
    1. Pesko KN,
    2. Torres-Perez F,
    3. Hjelle BL,
    4. Ebel GD
    . 2010. Molecular epidemiology of Powassan virus in North America. J Gen Virol 91:2698–2705. doi:10.1099/vir.0.024232-0.
    OpenUrlCrossRefPubMedWeb of Science
  177. 177.↵
    1. Tavakoli NP,
    2. Wang H,
    3. Dupuis M,
    4. Hull R,
    5. Ebel GD,
    6. Gilmore EJ,
    7. Faust PL
    . 2009. Fatal case of deer tick virus encephalitis. N Engl J Med 360:2099–2107. doi:10.1056/NEJMoa0806326.
    OpenUrlCrossRefPubMedWeb of Science
  178. 178.↵
    1. El Khoury MY,
    2. Camargo JF,
    3. White JL,
    4. Backenson BP,
    5. Dupuis AP, II,
    6. Escuyer KL,
    7. Kramer L,
    8. St George K,
    9. Chatterjee D,
    10. Prusinski M,
    11. Wormser GP,
    12. Wong SJ
    . 2013. Potential role of deer tick virus in Powassan encephalitis cases in Lyme disease-endemic areas of New York, USA. Emerg Infect Dis 19:1926–1933. doi:10.3201/eid1912.130903.
    OpenUrlCrossRefPubMed
  179. 179.↵
    1. Ebel GD,
    2. Foppa I,
    3. Spielman A,
    4. Telford SR, II.
    1999. A focus of deer tick virus transmission in the northcentral united States. Emerg Infect Dis 5:570–574. doi:10.3201/eid0504.990423.
    OpenUrlCrossRefPubMedWeb of Science
  180. 180.↵
    1. Anderson JF,
    2. Armstrong PM
    . 2012. Prevalence and genetic characterization of Powassan virus strains infecting Ixodes scapularis in Connecticut. Am J Trop Med Hyg 87:754–759. doi:10.4269/ajtmh.2012.12-0294.
    OpenUrlAbstract/FREE Full Text
  181. 181.↵
    1. Tokarz R,
    2. Williams SH,
    3. Sameroff S,
    4. Sanchez Leon M,
    5. Jain K,
    6. Lipkin WI
    . 2014. Virome analysis of Amblyomma americanum, Dermacentor variabilis, and Ixodes scapularis ticks reveals novel highly divergent vertebrate and invertebrate viruses. J Virol 88:11480–11492. doi:10.1128/JVI.01858-14.
    OpenUrlAbstract/FREE Full Text
  182. 182.↵
    1. L’vov DK,
    2. Al’khovskiĭ SV,
    3. Shchelkanov MI,
    4. Deriabin PG,
    5. Gitel’man AK,
    6. Botikov AG,
    7. Aristova VA
    . 2014. Genetic characterisation of Powassan virus (POWV) isolated from Haemophysalis longicornis ticks in Primorye and two strains of Tick-borne encephalitis virus (TBEV) (Flaviviridae, Flavivirus): Alma-Arasan virus (AAV) isolated from Ixodes persulcatus ticks in Kazakhstan and Malyshevo virus isolated from Aedes vexans nipponii mosquitoes in Khabarovsk kray. Vopr Virusol 59:18–22.
    OpenUrl
  183. 183.↵
    1. Brackney DE,
    2. Nofchissey RA,
    3. Fitzpatrick KA,
    4. Brown IK,
    5. Ebel GD
    . 2008. Stable prevalence of Powassan virus in Ixodes scapularis in a northern Wisconsin focus. Am J Trop Med Hyg 79:971–973. doi:10.4269/ajtmh.2008.79.971.
    OpenUrlAbstract/FREE Full Text
  184. 184.↵
    1. Dupuis AP, II,
    2. Peters RJ,
    3. Prusinski MA,
    4. Falco RC,
    5. Ostfeld RS,
    6. Kramer LD
    . 2013. Isolation of deer tick virus (Powassan virus, lineage II) from Ixodes scapularis and detection of antibody in vertebrate hosts sampled in the Hudson Valley, New York State. Parasit Vectors 6:185. doi:10.1186/1756-3305-6-185.
    OpenUrlCrossRefPubMed
  185. 185.↵
    1. Ebel GD,
    2. Spielman A,
    3. Telford SR, III.
    2001. Phylogeny of North American Powassan virus. J Gen Virol 82:1657–1665. doi:10.1099/0022-1317-82-7-1657.
    OpenUrlCrossRefPubMedWeb of Science
  186. 186.↵
    1. Pettersson JH,
    2. Fiz-Palacios O
    . 2014. Dating the origin of the genus Flavivirus in the light of Beringian biogeography. J Gen Virol 95:1969–1982. doi:10.1099/vir.0.065227-0.
    OpenUrlCrossRefPubMed
  187. 187.↵
    1. Subbotina EL,
    2. Loktev VB
    . 2012. Molecular evolution of the tick-borne encephalitis and Powassan viruses. Mol Biol 46:75–84. doi:10.1134/S0026893311060148.
    OpenUrlCrossRef
  188. 188.↵
    1. Heinze DM,
    2. Gould EA,
    3. Forrester NL
    . 2012. Revisiting the clinal concept of evolution and dispersal for the tick-borne flaviviruses by using phylogenetic and biogeographic analyses. J Virol 86:8663–8671. doi:10.1128/JVI.01013-12.
    OpenUrlAbstract/FREE Full Text
  189. 189.↵
    1. Brackney DE,
    2. Brown IK,
    3. Nofchissey RA,
    4. Fitzpatrick KA,
    5. Ebel GD
    . 2010. Homogeneity of Powassan virus populations in naturally infected Ixodes scapularis. Virology 402:366–371. doi:10.1016/j.virol.2010.03.035.
    OpenUrlCrossRef
  190. 190.↵
    1. Grubaugh ND,
    2. Rückert C,
    3. Armstrong PM,
    4. Bransfield A,
    5. Anderson JF,
    6. Ebel GD,
    7. Brackney DE
    . 2016. Transmission bottlenecks and RNAi collectively influence tick-borne flavivirus evolution. Virus Evol 2:vew033. doi:10.1093/ve/vew033.
    OpenUrlCrossRef
  191. 191.↵
    1. McLean DM,
    2. McQueen EJ,
    3. Petite HE,
    4. Macpherson LW,
    5. Scholten TH,
    6. Ronald K
    . 1962. Powassan virus: field investigations in Northern Ontario, 1959 to 1961. Can Med Assoc J 86:971–974.
    OpenUrlPubMed
  192. 192.↵
    1. McLean DM,
    2. Larke RP
    . 1963. Powassan and Silverwater viruses: ecology of two Ontario arboviruses. Can Med Assoc J 88:182–185.
    OpenUrlPubMed
  193. 193.↵
    1. McLean DM,
    2. Cobb C,
    3. Gooderham SE,
    4. Smart CA,
    5. Wilson AG,
    6. Wilson WE
    . 1967. Powassan virus: persistence of virus activity during 1966. Can Med Assoc J 96:660–664.
    OpenUrlPubMed
  194. 194.↵
    1. McLean DM,
    2. Smith PA,
    3. Livingstone SE,
    4. Wilson WE,
    5. Wilson AG
    . 1966. Powassan virus: vernal spread during 1965. Can Med Assoc J 94:532–536.
    OpenUrlPubMed
  195. 195.↵
    1. McLean DM,
    2. Ladyman SR,
    3. Purvin-Good KW
    . 1968. Westward extension of Powassan virus prevalence. Can Med Assoc J 98:946–949.
    OpenUrlPubMed
  196. 196.↵
    1. Whitney E,
    2. Jamnback H
    . 1965. The first isolations of Powassan virus in New York state. Proc Soc Exp Biol Med 119:432–435. doi:10.3181/00379727-119-30202.
    OpenUrlCrossRefPubMed
  197. 197.↵
    1. Johnson HN
    . 1987. Isolation of Powassan virus from a spotted skunk in California. J Wildl Dis 23:152–153. doi:10.7589/0090-3558-23.1.152.
    OpenUrlCrossRefPubMed
  198. 198.↵
    1. Main AJ,
    2. Carey AB,
    3. Downs WG
    . 1979. Powassan virus in Ixodes cookei and Mustelidae in New England. J Wildl Dis 15:585–591. doi:10.7589/0090-3558-15.4.585.
    OpenUrlCrossRefPubMedWeb of Science
  199. 199.↵
    1. Ebel GD,
    2. Campbell EN,
    3. Goethert HK,
    4. Spielman A,
    5. Telford SR, III.
    2000. Enzootic transmission of deer tick virus in New England and Wisconsin sites. Am J Trop Med Hyg 63:36–42. doi:10.4269/ajtmh.2000.63.36.
    OpenUrlAbstract
  200. 200.↵
    1. Zarnke RL,
    2. Yuill TM
    . 1981. Powassan virus infection in snowshoe hares (Lepus americanus). J Wildl Dis 17:303–310. doi:10.7589/0090-3558-17.2.303.
    OpenUrlCrossRefPubMed
  201. 201.↵
    1. Timoney P
    . 1971. Powassan virus infection in the grey squirrel. Acta Virol 15:429.
    OpenUrlPubMedWeb of Science
  202. 202.↵
    1. McLean DM,
    2. Crawford MA,
    3. Ladyman SR,
    4. Peers RR,
    5. Purvin-Good KW
    . 1970. California encephalitis and Powassan virus activity in British Columbia, 1969. Am J Epidemiol 92:266–272. doi:10.1093/oxfordjournals.aje.a121206.
    OpenUrlCrossRefPubMed
  203. 203.↵
    1. McLean DM,
    2. Best JM,
    3. Mahalingam S,
    4. Chernesky MA,
    5. Wilson WE
    . 1964. Powassan virus: summer infection cycle, 1964. Can Med Assoc J 91:1360–1362.
    OpenUrlPubMed
  204. 204.↵
    1. McLean DM,
    2. Devos A,
    3. Quantz EJ
    . 1964. Powassan virus: field investigations during the summer of 1963. Am J Trop Med Hyg 13:747–753. doi:10.4269/ajtmh.1964.13.747.
    OpenUrlAbstract/FREE Full Text
  205. 205.↵
    1. Whitney E,
    2. Roz AP,
    3. Rayner GA,
    4. Deibel R
    . 1969. Serologic survey for arbovirus activity in deer sera from nine counties in New York State. Wildl Dis 5:392–397.
    OpenUrlPubMed
  206. 206.↵
    1. Whitney E,
    2. Jamnback H,
    3. Means RG,
    4. Watthews TH
    . 1968. Arthropod-borne-virus survey in St. Lawrence County, New York. Arbovirus reactivity in serum from amphibians, reptiles, birds, and mammals. Am J Trop Med Hyg 17:645–650. doi:10.4269/ajtmh.1968.17.645.
    OpenUrlAbstract/FREE Full Text
  207. 207.↵
    1. Artsob H,
    2. Spence L,
    3. Th’ng C,
    4. Lampotang V,
    5. Johnston D,
    6. MacInnes C,
    7. Matejka F,
    8. Voigt D,
    9. Watt I
    . 1986. Arbovirus infections in several Ontario mammals, 1975-1980. Can J Vet Res 50:42–46.
    OpenUrlPubMedWeb of Science
  208. 208.↵
    1. Pedersen K,
    2. Wang E,
    3. Weaver SC,
    4. Wolf PC,
    5. Randall AR,
    6. Van Why KR,
    7. Travassos Da Rosa APA,
    8. Gidlewski T
    . 2017. Serologic evidence of various arboviruses detected in white-tailed deer (Odocoileus virginianus) in the United States. Am J Trop Med Hyg 97:319–323. doi:10.4269/ajtmh.17-0180.
    OpenUrlCrossRef
  209. 209.↵
    1. Lvov DK,
    2. Shchelkanov MY,
    3. Alkhovsky SV,
    4. Deryabin PG
    . 2015. Zoonotic viruses of Northern Eurasia: taxonomy and ecology, p 135–392. Academic Press, Amsterdam, Netherlands.
  210. 210.↵
    1. Little PB,
    2. Thorsen J,
    3. Moore W,
    4. Weninger N
    . 1985. Powassan viral encephalitis: a review and experimental studies in the horse and rabbit. Vet Pathol 22:500–507. doi:10.1177/030098588502200510.
    OpenUrlCrossRefPubMed
  211. 211.↵
    1. Keane DP,
    2. Parent J,
    3. Little PB
    . 1987. California serogroup and Powassan virus infection of cats. Can J Microbiol 33:693–697. doi:10.1139/m87-121.
    OpenUrlCrossRefPubMedWeb of Science
  212. 212.↵
    1. Artsob H,
    2. Spence L,
    3. Surgeoner G,
    4. McCreadie J,
    5. Thorsen J,
    6. Th’ng C,
    7. Lampotang V
    . 1984. Isolation of Francisella tularensis and Powassan virus from ticks (Acari: Ixodidae) in Ontario, Canada. J Med Entomol 21:165–168. doi:10.1093/jmedent/21.2.165.
    OpenUrlCrossRefPubMed
  213. 213.↵
    1. McLean DM,
    2. Chernesky MA,
    3. Chernesky SJ,
    4. Goddard EJ,
    5. Ladyman SR,
    6. Peers RR,
    7. Purvin-Good KW
    . 1969. Arbovirus prevalence in the East Kootenay Region, 1968. Can Med Assoc J 100:320–326.
    OpenUrlPubMed
  214. 214.↵
    1. McCoy KD,
    2. Léger E,
    3. Dietrich M
    . 2013. Host specialization in ticks and transmission of tick-borne diseases: a review. Front Cell Infect Microbiol 3:57. doi:10.3389/fcimb.2013.00057.
    OpenUrlCrossRef
  215. 215.↵
    1. Costero A,
    2. Grayson MA
    . 1996. Experimental transmission of Powassan virus (Flaviviridae) by Ixodes scapularis ticks (Acari: Ixodidae). Am J Trop Med Hyg 55:536–546. doi:10.4269/ajtmh.1996.55.536.
    OpenUrlAbstract/FREE Full Text
  216. 216.↵
    1. Lvov DK,
    2. Leonova GN,
    3. Gromashevskiĭ VL,
    4. Belikova NP,
    5. Berezina LK
    . 1974. Isolation of the Powassan virus from Haemaphysalis neumanni Dönitz, 1905 ticks in the Maritime Territory. Vopr Virusol 1974(5):538–541.
    OpenUrl
  217. 217.↵
    1. Krugliak SP,
    2. Leonova GN
    . 1989. The significance of Ixodes ticks in the southern Far East in the circulation of Powassan virus. Vopr Virusol 34:358–362.
    OpenUrlPubMed
  218. 218.↵
    1. Leonova GN,
    2. Krugliak SP,
    3. Lozovskaia SA,
    4. Rybachuk VN
    . 1987. The role of wild murine rodents in the selection of different strains of tick-borne encephalitis and Powassan viruses. Vopr Virusol 32:591–595.
    OpenUrlPubMed
  219. 219.↵
    1. Kuno G
    . 2007. Host range specificity of flaviviruses: correlation with in vitro replication. J Med Entomol 44:93–101. doi:10.1093/jmedent/41.5.93.
    OpenUrlCrossRefPubMed
  220. 220.↵
    1. Lawrie CH,
    2. Uzcátegui NY,
    3. Armesto M,
    4. Bell-Sakyi L,
    5. Gould EA
    . 2004. Susceptibility of mosquito and tick cell lines to infection with various flaviviruses. Med Vet Entomol 18:268–274. doi:10.1111/j.0269-283X.2004.00505.x.
    OpenUrlCrossRefPubMed
  221. 221.↵
    1. Doughty CT,
    2. Yawetz S,
    3. Lyons J
    . 2017. Emerging causes of arbovirus encephalitis in North America: Powassan, Chikungunya, and Zika viruses. Curr Neurol Neurosci Rep 17:12. doi:10.1007/s11910-017-0724-3.
    OpenUrlCrossRef
  222. 222.↵
    1. Dhama K,
    2. Pawaiya RVS,
    3. Chakraborty S,
    4. Tiwari R,
    5. Verma AK
    . 2014. Powassan virus (POWV) infection in animals and humans: a review. Asian J Anim Vet Adv 9:177–189. doi:10.3923/ajava.2014.177.189.
    OpenUrlCrossRef
  223. 223.↵
    1. Partington MW,
    2. Thomson V,
    3. O’Shaughnessy MV
    . 1980. Powassan virus encephalitis in southeastern Ontario. Can Med Assoc J 123:603–606.
    OpenUrlPubMed
  224. 224.↵
    1. Cavanaugh CE,
    2. Muscat PL,
    3. Telford SR, III,
    4. Goethert H,
    5. Pendlebury W,
    6. Elias SP,
    7. Robich R,
    8. Welch M,
    9. Lubelczyk CB,
    10. Smith RP
    . 2017. Fatal deer tick virus infection in Maine. Clin Infect Dis 65:1043–1046. doi:10.1093/cid/cix435.
    OpenUrlCrossRef
  225. 225.↵
    1. Piantadosi A,
    2. Kanjilal S,
    3. Ganesh V,
    4. Khanna A,
    5. Hyle EP,
    6. Rosand J,
    7. Bold T,
    8. Metsky HC,
    9. Lemieux J,
    10. Leone MJ,
    11. Freimark L,
    12. Matranga CB,
    13. Adams G,
    14. McGrath G,
    15. Zamirpour S,
    16. Telford S, III,
    17. Rosenberg E,
    18. Cho T,
    19. Frosch MP,
    20. Goldberg MB,
    21. Mukerji SS,
    22. Sabeti PC
    . 2018. Rapid detection of Powassan virus in a patient with encephalitis by metagenomic sequencing. Clin Infect Dis 66:789–792. doi:10.1093/cid/cix792.
    OpenUrlCrossRef
  226. 226.↵
    1. Smith R,
    2. Woodall JP,
    3. Whitney E,
    4. Deibel R,
    5. Gross MA,
    6. Smith V,
    7. Bast TF
    . 1974. Powassan virus infection. A report of three human cases of encephalitis. Am J Dis Child 127:691–693. doi:10.1001/archpedi.1974.02110240077010.
    OpenUrlCrossRefPubMed
  227. 227.↵
    1. Conway D,
    2. Rossier E,
    3. Spence L,
    4. Artsob A
    . 1976. Powassan virus encephalitis with shoulder girdle involvement. Can Dis Wkly Rep 2:85–87.
    OpenUrl
  228. 228.↵
    1. Embil JA,
    2. Camfield P,
    3. Artsob H,
    4. Chase DP
    . 1983. Powassan virus encephalitis resembling herpes simplex encephalitis. Arch Intern Med 143:341–343. doi:10.1001/archinte.1983.00350020167030.
    OpenUrlCrossRefPubMedWeb of Science
  229. 229.↵
    1. Kolski H,
    2. Ford-Jones EL,
    3. Richardson S,
    4. Petric M,
    5. Nelson S,
    6. Jamieson F,
    7. Blaser S,
    8. Gold R,
    9. Otsubo H,
    10. Heurter H,
    11. MacGregor D
    . 1998. Etiology of acute childhood encephalitis at The Hospital for Sick Children, Toronto, 1994-1995. Clin Infect Dis 26:398–409. doi:10.1086/516301.
    OpenUrlCrossRefPubMedWeb of Science
  230. 230.↵
    1. Lessell S,
    2. Collins TE
    . 2003. Ophthalmoplegia in Powassan encephalitis. Neurology 60:1726–1727. doi:10.1212/01.WNL.0000064167.16083.02.
    OpenUrlFREE Full Text
  231. 231.↵
    Centers for Disease Control and Prevention. 1995. Arboviral disease—United States, 1994. MMWR Morb Mortal Wkly Rep 44:641–644.
    OpenUrlPubMed
  232. 232.↵
    1. Jackson AC
    . 1989. Leg weakness associated with Powassan virus infection—Ontario. Can Dis Wkly Rep 15:123–124.
    OpenUrlPubMed
  233. 233.↵
    1. Deibel R,
    2. Srihongse S,
    3. Woodall JP
    . 1979. Arboviruses in New York State: an attempt to determine the role of arboviruses in patients with viral encephalitis and meningitis. Am J Trop Med Hyg 28:577–582. doi:10.4269/ajtmh.1979.28.577.
    OpenUrlAbstract/FREE Full Text
  234. 234.↵
    1. Aliota MT,
    2. Dupuis AP, II,
    3. Wilczek MP,
    4. Peters RJ,
    5. Ostfeld RS,
    6. Kramer LD
    . 2014. The prevalence of zoonotic tick-borne pathogens in Ixodes scapularis collected in the Hudson Valley, New York State. Vector Borne Zoonotic Dis 14:245–250. doi:10.1089/vbz.2013.1475.
    OpenUrlCrossRefPubMed
  235. 235.↵
    1. Tokarz R,
    2. Tagliafierro T,
    3. Cucura DM,
    4. Rochlin I,
    5. Sameroff S,
    6. Lipkin WI
    . 2017. Detection of Anaplasma phagocytophilum, Babesia microti, Borrelia burgdorferi, Borrelia miyamotoi, and Powassan virus in ticks by a multiplex real-time reverse transcription-PCR assay. mSphere 2:e00151-17. doi:10.1128/mSphere.00151-17.
    OpenUrlAbstract/FREE Full Text
  236. 236.↵
    1. Johnson AJ,
    2. Martin DA,
    3. Karabatsos N,
    4. Roehrig JT
    . 2000. Detection of anti-arboviral immunoglobulin G by using a monoclonal antibody-based capture enzyme-linked immunosorbent assay. J Clin Microbiol 38:1827–1831.
    OpenUrlAbstract/FREE Full Text
  237. 237.↵
    1. Martin DA,
    2. Muth DA,
    3. Brown T,
    4. Johnson AJ,
    5. Karabatsos N,
    6. Roehrig JT
    . 2000. Standardization of immunoglobulin M capture enzyme-linked immunosorbent assays for routine diagnosis of arboviral infections. J Clin Microbiol 38:1823–1826.
    OpenUrlAbstract/FREE Full Text
  238. 238.↵
    1. Basile AJ,
    2. Horiuchi K,
    3. Panella AJ,
    4. Laven J,
    5. Kosoy O,
    6. Lanciotti RS,
    7. Venkateswaran N,
    8. Biggerstaff BJ
    . 2013. Multiplex microsphere immunoassays for the detection of IgM and IgG to arboviral diseases. PLoS One 8:e75670. doi:10.1371/journal.pone.0075670.
    OpenUrlCrossRef
  239. 239.↵
    1. Thomm AM,
    2. Schotthoefer AM,
    3. Dupuis AP, II,
    4. Kramer LD,
    5. Frost HM,
    6. Fritsche TR,
    7. Harrington YA,
    8. Knox KK,
    9. Kehl SC
    . 2018. Development and validation of a serologic test panel for detection of Powassan virus infection in U.S. patients residing in regions where Lyme disease is endemic. mSphere 3:e00467-17. doi:10.1128/mSphere.00467-17.
    OpenUrlAbstract/FREE Full Text
  240. 240.↵
    Centers for Disease Control and Prevention. 2008. False-positive results with a commercially available West Nile virus immunoglobulin M assay—United States, 2008. MMWR Morb Mortal Wkly Rep 58:458–460.
    OpenUrl
  241. 241.↵
    1. Johnson AJ,
    2. Noga AJ,
    3. Kosoy O,
    4. Lanciotti RS,
    5. Johnson AA,
    6. Biggerstaff BJ
    . 2005. Duplex microsphere-based immunoassay for detection of anti-West Nile virus and anti-St. Louis encephalitis virus immunoglobulin M antibodies. Clin Diagn Lab Immunol 12:566–574. doi:10.1128/CDLI.12.5.566-574.2005.
    OpenUrlCrossRefPubMed
  242. 242.↵
    1. Abdelwahab KS,
    2. Almeida JD,
    3. Doane FW,
    4. McLean DM
    . 1964. Powassan virus: morphology and cytopathology. Can Med Assoc J 90:1068–1072.
    OpenUrlPubMed
  243. 243.↵
    1. Patel P,
    2. Landt O,
    3. Kaiser M,
    4. Faye O,
    5. Koppe T,
    6. Lass U,
    7. Sall AA,
    8. Niedrig M
    . 2013. Development of one-step quantitative reverse transcription PCR for the rapid detection of flaviviruses. Virol J 10:58. doi:10.1186/1743-422X-10-58.
    OpenUrlCrossRefPubMed
  244. 244.↵
    1. Meiyu F,
    2. Huosheng C,
    3. Cuihua C,
    4. Xiaodong T,
    5. Lianhua J,
    6. Yifei P,
    7. Weijun C,
    8. Huiyu G
    . 1997. Detection of flaviviruses by reverse transcriptase-polymerase chain reaction with the universal primer set. Microbiol Immunol 41:209–213. doi:10.1111/j.1348-0421.1997.tb01192.x.
    OpenUrlCrossRefPubMedWeb of Science
  245. 245.↵
    1. Scaramozzino N,
    2. Crance J-M,
    3. Jouan A,
    4. DeBriel DA,
    5. Stoll F,
    6. Garin D
    . 2001. Comparison of flavivirus universal primer pairs and development of a rapid, highly sensitive heminested reverse transcription-PCR assay for detection of flavivirus targeted to a conserved region of the NS5 gene sequences. J Clin Microbiol 39:1922–1927. doi:10.1128/JCM.39.5.1922-1927.2001.
    OpenUrlAbstract/FREE Full Text
  246. 246.↵
    1. Whitby JE,
    2. Ni H,
    3. Whitby HE,
    4. Jennings AD,
    5. Bradley LM,
    6. Lee JM,
    7. Lloyd G,
    8. Stephenson JR,
    9. Barrett AD
    . 1993. Rapid detection of viruses of the tick-borne encephalitis virus complex by RT-PCR of viral RNA. J Virol Methods 45:103–114. doi:10.1016/0166-0934(93)90144-G.
    OpenUrlCrossRefPubMed
  247. 247.↵
    1. Gaunt MW,
    2. Gould EA
    . 2005. Rapid subgroup identification of the flaviviruses using degenerate primer E-gene RT-PCR and site specific restriction enzyme analysis. J Virol Methods 128:113–127. doi:10.1016/j.jviromet.2005.04.006.
    OpenUrlCrossRefPubMedWeb of Science
  248. 248.↵
    1. Domingo C,
    2. Patel P,
    3. Linke S,
    4. Achazi K,
    5. Niedrig M
    . 2011. Molecular diagnosis of flaviviruses. Future Virol 6:1059–1074. doi:10.2217/fvl.11.77.
    OpenUrlCrossRef
  249. 249.↵
    1. Grant-Klein RJ,
    2. Baldwin CD,
    3. Turell MJ,
    4. Rossi CA,
    5. Li F,
    6. Lovari R,
    7. Crowder CD,
    8. Matthews HE,
    9. Rounds MA,
    10. Eshoo MW,
    11. Blyn LB,
    12. Ecker DJ,
    13. Sampath R,
    14. Whitehouse CA
    . 2010. Rapid identification of vector-borne flaviviruses by mass spectrometry. Mol Cell Probes 24:219–228. doi:10.1016/j.mcp.2010.04.003.
    OpenUrlCrossRefPubMed
  250. 250.↵
    1. El Khoury MY,
    2. Hull RC,
    3. Bryant PW,
    4. Escuyer KL,
    5. St George K,
    6. Wong SJ,
    7. Nagaraja A,
    8. Kramer L,
    9. Dupuis AP,
    10. Purohit T,
    11. Shah T,
    12. Wormser GP
    . 2013. Diagnosis of acute deer tick virus encephalitis. Clin Infect Dis 56:e40–e47. doi:10.1093/cid/cis938.
    OpenUrlCrossRefPubMed
  251. 251.↵
    1. Khan MJ,
    2. Trabuco AC,
    3. Alfonso HL,
    4. Figueiredo ML,
    5. Batista WC,
    6. Badra SJ,
    7. Figueiredo LT,
    8. Lavrador MA,
    9. Aquino VH
    . 2016. DNA microarray platform for detection and surveillance of viruses transmitted by small mammals and arthropods. PLoS Negl Trop Dis 10:e0005017. doi:10.1371/journal.pntd.0005017.
    OpenUrlCrossRef
  252. 252.↵
    1. Vina-Rodriguez A,
    2. Sachse K,
    3. Ziegler U,
    4. Chaintoutis SC,
    5. Keller M,
    6. Groschup MH,
    7. Eiden M
    . 2017. A novel pan-flavivirus detection and identification assay based on RT-qPCR and microarray. Biomed Res Int 2017:4248756. doi:10.1155/2017/4248756.
    OpenUrlCrossRef
  253. 253.↵
    1. Wang D,
    2. Coscoy L,
    3. Zylberberg M,
    4. Avila PC,
    5. Boushey HA,
    6. Ganem D,
    7. DeRisi JL
    . 2002. Microarray-based detection and genotyping of viral pathogens. Proc Natl Acad Sci U S A 99:15687–15692. doi:10.1073/pnas.242579699.
    OpenUrlAbstract/FREE Full Text
  254. 254.↵
    1. Palacios G,
    2. Quan PL,
    3. Jabado OJ,
    4. Conlan S,
    5. Hirschberg DL,
    6. Liu Y,
    7. Zhai J,
    8. Renwick N,
    9. Hui J,
    10. Hegyi H,
    11. Grolla A,
    12. Strong JE,
    13. Towner JS,
    14. Geisbert TW,
    15. Jahrling PB,
    16. Büchen-Osmond C,
    17. Ellerbrok H,
    18. Sanchez-Seco MP,
    19. Lussier Y,
    20. Formenty P,
    21. Nichol MS,
    22. Feldmann H,
    23. Briese T,
    24. Lipkin WI
    . 2007. Panmicrobial oligonucleotide array for diagnosis of infectious diseases. Emerg Infect Dis 13:73–81. doi:10.3201/eid1301.060837.
    OpenUrlCrossRefPubMedWeb of Science
  255. 255.↵
    1. Wylie TN,
    2. Wylie KM,
    3. Herter BN,
    4. Storch GA
    . 2015. Enhanced virome sequencing using targeted sequence capture. Genome Res 25:1910–1920. doi:10.1101/gr.191049.115.
    OpenUrlAbstract/FREE Full Text

Author Bios


Embedded Image

Gábor Kemenesi obtained his M.Sc. degree and defended his Ph.D. at the Faculty of Sciences, University of Pécs, Hungary. After graduating at the University of Pécs, Hungary, he started to work with arthropod-borne disease in Europe, such as West Nile fever virus and Usutu virus. His research interests include highly pathogenic viral zoonoses and wildlife diseases with zoonotic potential. His current research projects are focusing on rodent-, bat-, and arthropod-borne infections, which occur across Eurasia and Africa. He is the deputy leader of the Biosafety Level 4 suit laboratory at the Szentágothai Research Centre, Pécs.


Embedded Image

Krisztián Bányai obtained his M.Sc. degree at the Faculty of Sciences, University of Pécs (then Janus Pannonius University of Pécs), and defended his Ph.D. degree at the Faculty of Medicine, University of Pécs. As a research fellow of the Association of Public Health Laboratories, he did his postdoctoral training at the Centers for Disease Control and Prevention, Atlanta, GA. Currently, he is the leader of the Lendület Pathogen Discovery Group at the Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary. His research activities include, but are not limited to, understanding arbovirus diversity and molecular epidemiology of zoonotic infections.

View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
Tick-Borne Flaviviruses, with a Focus on Powassan Virus
Gábor Kemenesi, Krisztián Bányai
Clinical Microbiology Reviews Dec 2018, 32 (1) e00106-17; DOI: 10.1128/CMR.00106-17

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Clinical Microbiology Reviews article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Tick-Borne Flaviviruses, with a Focus on Powassan Virus
(Your Name) has forwarded a page to you from Clinical Microbiology Reviews
(Your Name) thought you would be interested in this article in Clinical Microbiology Reviews.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Tick-Borne Flaviviruses, with a Focus on Powassan Virus
Gábor Kemenesi, Krisztián Bányai
Clinical Microbiology Reviews Dec 2018, 32 (1) e00106-17; DOI: 10.1128/CMR.00106-17
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • SUMMARY
    • INTRODUCTION
    • TICK-BORNE FLAVIVIRUSES
    • POWASSAN VIRUS
    • CONCLUSIONS AND PERSPECTIVES
    • ACKNOWLEDGMENTS
    • REFERENCES
    • Author Bios
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Powassan virus
arbovirus
viral encephalitis

Related Articles

Cited By...

About