Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Neurovirol. Author manuscript; available in PMC 2015 Dec 1.
Published in final edited form as:
PMCID: PMC4331079
NIHMSID: NIHMS633656
PMID: 25287260

Flaviviruses, an expanding threat in public health: focus on Dengue, West Nile, and Japanese encephalitis virus

Abstract

The flaviviruses Dengue, West Nile, and Japanese encephalitis represent three major mosquito-borne viruses worldwide. These pathogens impact the lives of millions of individuals and potentially could affect non-endemic areas already colonized by mosquito vectors. Unintentional transport of infected vectors (Aedes and Culex sp), traveling within endemic areas, rapid adaptation of the insects into new geographic locations, climate change, and lack of medical surveillance have greatly contributed to the increase in flaviviral infections worldwide. The mechanisms by which flaviviruses alter the immune and the central nervous system have only recently been examined despite the alarming number of infections, related deaths, and increasing global distribution. In this review, we will discuss the expansion of the geographic areas affected by flaviviruses, the potential threats to previously unaffected countries, the mechanisms of pathogenesis, and the potential therapeutic interventions to limit the devastating consequences of these viruses.

Keywords: vaccines, mosquito, fever, bioterrorism, brain

General Introduction

The genus Flavivirus is composed of approximately 73 arthropod-borne viruses, or arboviruses, that infect rodents, pigs, birds, non-human primates, humans, and other mammalian hosts. Several members of this virus family which include the dengue virus (DENV), Japanese encephalitis virus (JEV), West Nile virus (WNV), St. Louis encephalitis virus, and Yellow Fever virus are associated with important human diseases which are transmitted by arthropod vectors. Hepatitis C virus is a notable exception as while it is related to the other medially important flaviviruses, it is one of the few members of this viral family that is not vector-borne. Most of these viruses can cause a wide variety of clinical manifestations and complications such as undifferentiated fever, capillary leakage-hemorrhagic disease, and encephalitis which can potentially lead to death. Most flaviviruses are zoonotic and depend upon non-human animal vectors for their survival, replication, and dispersal with the exception of DENV which propagates mainly in humans. While the evolutionary event that led to the increased spread of the viruses is still unknown, population movements, rapid urbanization, and widespread deforestation have contributed to the expansion of the pathogens into previously non-endemic areas (Bhatt et al, 2013; Petersen and Marfin, 2005). In addition, burgeoning travel to endemic areas and the slow increase in global temperatures due to climate changes have allowed the expansion of WNV and DENV into new territories (Beatty et al, 2005; Caminade et al, 2012; Hansen, 2006; Rahmstorf et al, 2007). Despite the adaptation, wild reservoirs of DENV are still maintained in subtropical and tropical areas that support a mosquito-monkey-mosquito transmission cycle (Gubler, 2002). While most flaviviruses are endemic in tropical areas, the distribution of several members of the viral family such as WNV has extended to temperate areas including the United States of America within the past decade (Caminade et al, 2012).

Flavivirus epidemiology and global dispersal

Flaviviruses are important human pathogens that have plagued mankind, accounting for millions of mortality worldwide (Table 1). The first recorded epidemic of dengue-like disease was reported between 1779 and 1780 when outbreaks occurred in Asia, Africa, and North America (Gubler, 1998; Gubler, 2002). Since its emergence, four DENV closely related but genetically distinct serotypes have been identified (Zanotto et al, 1996). Each serotype is believed to have emerged from a common ancestor and evolved separately (Kawaguchi et al, 2003) despite possessing different antigenicity and causing different degrees of illness severities but often potentially leading to dengue hemorrhagic fever (DHF; currently being changed to severe dengue as indicated by WHO). Variations within each serotype have been genetically detected, though not limited to, within the envelope protein (E) and non-structural protein, NS5, within the viral genome. WNV is being transmitted within in the U.S. population since the first reported case in 1999 (see Table 1 for details). Since then, the total number of West Nile virus infection cases has been increasing annually within the United States of America(http://www.cdc.gov/westnile/resources/pdfs/cummulative/99_2013_cummulativeHumanCases.pdf). Currently, DENV is endemic in at least 100 countries throughout Asia, the Pacific, the Americas, Africa, and the Caribbean (Figure 1). Approximately one million infected individuals suffer from either dengue fever or dengue hemorrhagic fever annually with Dengue-related deaths estimated to occur between 1 to 5 percent of those infected as reported by WHO (http://whqlibdoc.who.int/publications/2009/9789241547871_eng.pdf) (see Table 1). DENV infections are often self-limiting and range from asymptomatic to relatively mild, undifferentiated illness with the patient eventually making full recovery. However, approximately 5% of symptomatic cases progress to a more severe disease whose manifestations include fever, myalgia, vomiting, and acute abdominal pain. The symptoms then progress further to hypotension, tachycardia, decreased peripheral perfusion, perisoreositis, and myocarditis (Krishnamurti et al, 2001; Malavige et al, 2004). In addition to these symptoms, hemorrhage develops approximately 7 days post-infection with no observed circulatory collapse (Krishnamurti et al, 2001). Factors influencing disease severity are not well understood. A predisposition to severe illness in secondary infections due to antibody-dependent enhancement of infection is often cited (Kliks et al, 1989).

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0001.jpg

Global distribution of DENV in relation to its arthropod vectors. DENV is endemic in parts of Asia, Australia, Africa, and Latin America (area between the solid lines) and slowly expanding to other parts of the globe. Currently, DENV is located within the distribution range of A. aegypti and A. albopictus (dotted line). The presences of these mosquito vectors in non-DENV endemic areas suggest the potential for the expansion of the virus into new regions if optimal conditions are present.

Table 1

Flavivirus epidemiology, infection, and complications.

VirusAt Risk IndividualsAnnually Reported New Infection CasesSymptomsNeurologic DiseaseMortality Rate
Japanese Encephalitis Virus (JEV)Children-Young Adult (0-15 years old)35,000-50,000 new cases (in JEV-endemic countries)Pyrexia, cephalalgia, vomiting neurologic dysfunction, muscle weakness, seizures, loss of motor functionParkinsonism Flaccid paralysis Coma Acute Encephalitis~25-30%
Dengue (DENV)No age distinctionone million cases (globally)Pyrexia, myalgia, vomiting, cephalalgia, abdominal pain, bleeding, low blood pressure, tachycardia, seizuresEncephalitis~1-5%
West Nile Virus (WNV)Adults and immunocompromised persons(≥ 50 years old at greater risk of developing more severe disease and complications)Ranging from 20-5674 cases annually between 1999-2012 in the U.S; reports of WNV infection and mortality rates reported in Canada and MexicoFever, cephalalgia, nausea, vomiting, myalgia, muscle weakness, lower back pain, loss of motor functionEncephalitis, Meningitis~3-15%

In the United States and its territories, DENV transmission primarily occurs in tropical and subtropical areas such as Puerto Rico, the US Virgin Islands, American Samoa, and the US-Affiliated Pacific Islands (Imrie et al, 2006; Mohammed et al, 2010a; Rigau-Perez et al, 2001; Tomashek et al, 2009), where dengue is endemic. Dengue has been found to be the most frequent cause of febrile illness among US travelers returning from Asia, Latin America and the Caribbean (Freedman et al, 2006; Mohammed et al, 2010b; Sharp et al, 2012). In addition, outbreaks of dengue occur sporadically in non-endemic areas where the mosquito vectors exist (Brunkard et al, 2007; Graham et al, 2011; Radke et al, 2012; Ramos et al, 2008), Hawaii (Effleret al, 2005), and Florida, (Radke et al, 2012). Meanwhile, WNV infections have been reported throughout the continental United States. While the risk of WNV infection within various age groups remain the same, the potential for developing neurologic complications due to WNV infection increases with age. In the past 10 years approximately 40,000 individuals have become infected with WNV in the U.S. (http://www.cdc.gov/westnile/resources/pdfs/cummulative/99_2013_cummulativeHumanCases.pdf) , of which ~20% developed neuroinvasive diseases (i.e. encephalitis and meningitis) with 12% fatality rate (Lindsey et al, 2010). Interestingly, ~80% of WNV-infected individuals are asymptomatic.

Within days following infection with JEV, the host begins to exhibit clinical manifestations of the disease beginning with cephalalgia, vomiting, and pyrexia which last approximately 1 week (Sarkari et al, 2012b). After which, neurologic disorders ensues hallmarked by Parkinsonism, flaccid paralysis, and coma. Up to one third of infected patients acquire acute encephalitis which can often lead to complications and patient fatality (Lee et al, 2012; Sarkari et al, 2012a; Sarkari et al, 2012b). Indeed, up to fifty percent of patients who contract the virus often die due to complications. Interestingly, patients who enter convalescence remain seropositive against JEV despite persistent viremia (Ravi et al, 1993). Currently, there are no reported JEV cases within the U.S.

Distribution of viral vectors

DENV is transmitted to humans by the mosquito vectors, Aedes aegypti and Aedes albopictus. Similarly, the global dissemination of WNV (Figure 2) and JEV (Figure 3) rely on the Culex species of mosquitoes for their dispersal, in particular C. quinquefasciatus. Unlike WNV, however, JEV is currently localized in parts of Asia and surrounding islands and the northern region of Queensland, Australia. These mosquitoes, in particular A. aegypti, have become widely distributed across tropical and subtropical areas, including vast areas of the United States (Figure 1and and2).2). The spread of the DENV in the U.S. is exacerbated by the expanded range of A. albopictus in recent years, reaching as far as New England (Figure 2) (Mousson et al, 2005). In addition, mosquito species capable of transmitting WNV have also been discovered to be as far north as Canada (www.hc-sc.gc.ca). The spread of WNV as well as DENV is facilitated by the dispersal of suitable arthropod vectors which has been accelerated by rapid urbanization, increased travel into endemic countries, and adaptation to climate changes. Together, these events are expected to contribute to increases in the number of individuals affected by these pathogens (Anders et al, 2011; Petersen and Marfin, 2005).

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0002.jpg

Distribution of WNV and DENV infections have been reported United States. To date, WNV infections (green states) have been documented throughout the United States with the exception of Alaska (gray). Texas, Florida, and New York are currently the only states in which both WNV and locally acquired DENV (red states) have been reported. The dotted line designates the northernmost distribution of the WNV mosquito vector, Culex sp. while the dashed line illustrates the northern-most territory of the DENV mosquito vectors, Aedes sp, in the United States.

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0003.jpg

Distribution of JEV. JEV is currently endemic within South, East, and South East Asia (highlighted in blue) and its surrounding islands indicated by the red border. To date, there are no reported cases of JEV outside of this region; however, the expansion of its mosquito vector, Culex sp, to other countries worldwide make to spread of this dangerous pathogen a significant threat.

Typically, flaviviral transmission into humans occurs within two weeks of viremia following the initial feeding from an infected host. After entering a naϊve mosquito in the blood meal, the virus will require an additional 8-12 days of incubation before it can be re-transmitted to another human. Once infected with the virus, the mosquito remains infectious for the remained of its life. Symptoms among infected human hosts typically develop within seven days after the mosquito bite and lasts between 3-14 days. While some individuals do not develop any significant symptoms, they can still successfully transmit these viruses to others via mosquitoes. DENV is unique among other flaviviruses as they are the only virus within the family which utilizes humans as its amplifying host. Among WNV and JEV, viral amplification have been documented to occur within various mammalian hosts including horses, sheep, pigs, and goats; in the case of JEV, the virus has also been reported in bats as detected by qPCR (Liu et al, 2013; Olaleye et al, 1990; Pauvolid-Correa et al, 2011; Peiris et al, 1992). The expansion of WNV and JEV across different geographical regions is believed to be mediated by the seasonal migrations of birds (Reed et al, 2003).

Dengue is endemic to many parts of the tropics and subtropics, with outbreaks closely correlated with the completion of the annual monsoon season as reported by the WHO. During this time, an increase in the Aedes sp. mosquito population is also typically observed. Furthermore, the risk of contracting DHF also increases as more humans become infected with the virus during this short time period. A combination of optimal environmental conditions that facilitate an increase in infected arthropod vectors, greater presence of individuals with no immunity to one of the four virus types (DENV 1-4), and an opportunity for infected vector-host contact are required for the onset of DENV and other flavivirus epidemics. Interestingly, despite the induction of lifelong protective immunity against one DENV serotype, protection against the virus is partial and transient against other serotypes (Chen et al, 2004). Secondary infection by a different DENV serotype often leads to increased severity and carries a higher risk of susceptibility to DHF and patient mortality. DHF is a life-threatening illness characterized by internal bleeding, serious brain compromise, organ failure, and eventually death. In some cases dengue shock syndrome is observed among DF and DHF patients. While it is generally accepted that tertiary and quartenary DENV infections are asymptomatic, the lack of a clinical data that accurately determine multiple DENV infections among affected individuals and the importance of ADE make the surveillance of the virus and its treatment important. There are currently no approved vaccines or therapies for the disease. Treatment of infected patients is mostly palliative, with an administration of fluids and an occasional blood transfusion in the event of severe hemorrhage. The lack of treatment for DENV and related flaviviruses (i.e WNV and JEV) make these diseases debilitating and deadly.

In the past, DHF cases in the United States were only observed among travelers who have returned from visits to dengue-endemic areas. However, data from the Centers for Disease Control (CDC, http://www.cdc.gov/dengue/) and the National Resources Defense Council (NRDC, http://www.nrdc.org/health/dengue/) indicate that localized cases of DENV infection have already been detected in Hawaii, Texas, Florida, and New York along with the mosquitoes that are responsible for harboring the virus (Figure 2). While the U.S population currently has no immunity to the virus due to infrequent interactions between infected individuals and viral vectors which are necessary for successful dissemination of DENV, the presence of DENV within the U.S. suggests the imminent risk of infection among millions of Americans. Furthermore, the rapid spread of the WNV throughout the U.S. suggests the possibility for DENV to become prevalent within the U.S. given the proper existing conditions.

Flavivirus genetics and life cycle

Flaviviruses are composed of a single-stranded positive-sense RNA genome (~11 kilobases) packaged into a 40-60 nm virion comprising of a spherical nucleocapsid core coated in an icosahedral envelope (Harris et al, 2006; Rodenhuis-Zybert et al, 2010). The entire flaviviral genome consists of 10 genes encoding a large polyprotein that is post-translationally cleaved by host and viral proteases to produce three structural (protein capsid, C; prM/M protein, and envelope protein, E) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) (Figure 4) (Leyssen et al, 2000). The NS region encodes for various important proteases, in particular NS3 and NS2B, which are important in the auto-processing of the virus, mediation of viral genome replication, and virus packaging (Clyde and Harris, 2006). The transcription of these genes is controlled by the 5’- and 3’-untranslated regions (UTR) flanking the viral genome (Appaiahgari and Vrati, 2012b; Leyssen et al, 2000; Lindenbach et al, 2007).

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0004.jpg

Flavivirus genome. The flavivirus genome consists of a single stranded positive-sense RNA encoding a polyprotein post-translationally cleaved by host proteases (sites designated by the blue arrows) and viral proteases (sites designated by green arrows). The site designated by the red arrow is cleaved by a yet to be identified protease. Processing of the polyprotein produces 3 structural (white boxes) and 7 non-structural genes (grey boxes). The prM protein is then later cleaved within the Golgi to release the M protein important for the maturation of the virus. Translation of viral RNA is controlled by the untranslated regions (UTR) located at the 5’ and 3’-ends of the RNA.

The mechanisms of flaviviral infection of the host cell and its life cycle are not fully understood. The current consensus is that endocytosis of the viral particle is important in the successful infection of the cell and the production of progeny viruses (Figure 5). Attachment of the DENV and other flaviviruses to cells utilizes multiple potential receptors that facilitate the attachment and internalization of the virus (i.e. CD14, R80, heparin sulfate, C-type lectin receptors, DC-SIGN, and mannose receptors) (Rodenhuis-Zybert et al, 2010); however, the exact mechanism by which DENV and other flaviviruses use these molecules for cellular internalization is still under investigation. The attached virus is internalized into an endosomal compartment which acidifies to facilitate the fusion of the viral envelope with the endosomal compartment (Figure 5). This fusion of the viral envelope is due to the rearrangement of the capsid proteins resulting in the release of the virus into the host cell (van der Schaar et al, 2008). The viral RNA is released into the host cytoplasm and transported to the endoplasmic reticulum (ER) where it undergoes two different fates: first, the positive sense RNA is translated to produce a polyprotein that is post-translationally cleaved into structural and non-structural proteins (listed above) or second, the genetic material is converted into a negative sense RNA by viral NS5 RNA dependent RNA polymerases (RdRp) and used to produce positive-stranded RNA copies (Figure 5). The viral genome is then packaged within the cytoplasm by the action of the protein C to form the nucleocapsid while the prM and E proteins heterodimerize within the lumen of the ER and initiate viral budding (Kuhn et al, 2002; Zhang et al, 2004). Nascent virion particles formed within the ER travel through the secretory pathway and into the Golgi apparatus. Changes in pH within the trans-Golgi network trigger the dissociation of the prM/E heterodimers activating the cellular endo-protease furin. Activation of this protease leads to the cleavage the prM protein to generate protein M (membrane associated) and the peptide pr (Rauscher et al, 1997; Yu et al, 2008; Zybert et al, 2008). The cleavage of this protein complex results in a mature, fully infectious virion.

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0005.jpg

Flavivirus life cycle. Following attachment to the host extracellular surface (1), the virus is endocytosed (2) and encapsulated inside an endosomal vacuole. Acidification of the endosomal compartment alters the E protein causing the fusion of the virus with the endosome (3) facilitating virion release into the intracellular compartment where it is unpackaged (4). The released viral genome undergoes two different fates: the viral genome is either transported to the endoplasmic reticulum where it is translated into a polyprotein (5a) or converted into a negative-sense RNA to make positive-sense RNA copies (5b). The large polyprotein is post-translationally processed (6) producing structural and non-structural components important for virus assembly and maturation. The viral genome is packaged into a capsid and transported to the Golgi where it is coated by the E/M protein complex (7) to produce a mature virion (8).

Immuno and central nervous system pathogenesis

Studies of DENV immunopathogenesis are highly limited due to the nature of the disease and lack of animal models that recapitulate the disease. However, results obtained from humanized mice show some similarities in the development and progression of the disease which is typically observed in humans. Thus, these models will play an important role in elucidating flaviviral pathogenesis now and in the future (Mota and Rico-Hesse, 2011). Epidemiological studies have identified some of the risk factors important for disease development and severity following flaviviral infection. These factors include age (younger people are most susceptible to DENV and JEV infection while older or immunocompromised individuals are affected by WNV), high body mass index, viral strain, gender, genetic variation of the major histocompatibility complex (MHC)-class I-related sequence B, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), tumor necrosis factor-α (TNF-α) and phospholipase C epsilon 1 genes, environmental conditions such as mosquito spread and temperature, and secondary infection with a different viral strain (in the case of dengue) (Anders et al, 2011; Khor et al, 2011; Nguyen et al, 2005; Simmons et al, 2012).

In DENV infection, the serotype-specific life immunity acquired following the initial infection by the virus does not provide complete protection to other infecting serotypes. In fact, subsequent DENV infections increase the development of more severe dengue illnesses such DHF (Halstead et al, 2002; Kliks et al, 1988). The increased disease severity during secondary DENV infections is potentially due to the role of memory cells and antibodies. This is the concept for antibody-dependent enhancement (ADE) of secondary dengue infections (Halstead and O'Rourke, 1977). During ADE, antibodies generated after the initial infection recognize different DENV serotypes during secondary infections and facilitate increased binding and internalization of the virus via the Fc receptors on leukocytes and blood-brain barrier (BBB) cells. The adherence and infection of the BBB by DENV leads to inflammatory cytokine and chemokine production (i.e TNF-α, MCP-1/CCL2, IL-2, IL-6, IL-8, IL-10, and IL-12) as well as other inflammatory factors that lead to BBB compromise and CNS dysfunction (Anderson et al, 2011; Chaturvedi et al, 2000; Chen, 2012; Gubler, 1998; Malavige et al, 2004; Priyadarshini et al; Restrepo et al, 2008a; Restrepo et al, 2008b). This provides potential targets for therapeutic intervention against succeeding DENV infections.

Among leukocyte populations, dendritic and natural killer (NK) cells are the first ones to become infected. The infection of these cells occurs through pathogen recognizing receptors, such as toll-like receptors and DC-SIGN, and elicit inflammatory signals that lead to a Th2 immune response and eventual vascular barrier dysfunction (Chaturvedi et al, 2000). Monocyte/macrophages and lymphoid cells (T and B cells) have also been described as major preferential targets for flaviviruses due to the presence of viral RNA or proteins in infected cells which have been collected from lymphoid tissues (Durbin et al, 2008; Jessie et al, 2004; King et al, 1999; Lin et al, 2002; Mentor and Kurane, 1997; Srikiatkhachorn et al, 2012). The high viral titers within the germinal centers of these tissues also suggest that these sites can harbor viral particles that can disseminate and infect other sites within the host. Interestingly, flaviviruses have developed strategies to circumvent the host immune response limiting clearance either by inhibiting IFN secretion (Munoz-Jordan et al, 2003; Umareddy et al, 2008) or by reducing antigen presentation through the limitation of MHC and co-stimulatory molecule expression (Palmer et al, 2005). The loss of these molecules will lead to an impairment of CD4+ T lymphocyte activation and CD8+ T lymphocyte response and thus leading to the loss of an immune response against the viruses and disease development (Aleyas et al, 2009; Aleyas et al, 2010; Chase et al, 2011).

The in vivo cellular targets of DENV and other flaviviruses in the CNS remain to be fully characterized; however, our laboratory and others have observed that primary human astrocytes and brain microvascular endothelial cells can be infected by DENV (Daep, Munoz-Jordan and Eugenin, unpublished data). Given the potential importance of astrocytes as HIV reservoirs within the central nervous system (CNS) (Eugenin and Berman, 2007; Eugenin et al, 2011), these cells may also function to maintain DENV reservoirs within the brain. To date, the full mechanism of flavivirus infection of the CNS remains poorly understood due to early studies which did not detect pathological signs of viral invasion within the CNS (Burke, 1968; Nathanson and Cole, 1970). Deeper examination of CNS pathology following DENV infection, however, will provide groundbreaking information that will elucidate the role of the blood brain barrier, leukocytes, and CNS inflammation following flaviviral infection in the development of dengue fever (DF) and DHF.

Older manuscripts indicate that DENV antigens can be found in several tissues including neurons in the cerebrum, Purkinje's and granular cells in the cerebellum, astrocyte, microglia and cells of the choroid plexus (Bhoopat et al, 1996a). However, earlier reports describing DHF pathology indicates no brain compromise and clearly stated that minimal damage was observed (Burke, 1968; Nathanson and Cole, 1970) despite the devastation of the CNS following infection by DENV and other flaviviruses. In addition, positive detection of flaviviral antigens without viral RNA (Jessie et al., 2004) indicated viral uptake by endocytosis and/or phagocytosis and not by productive infection that result in progeny virions. Recent improvements in research techniques could provide detection assays with increased sensitivity and improved accuracy. In agreement, recent publications have demonstrated the presence of dengue antigens in the CNS due to the detection of viral proteins, immunoglobulin, and RNA (Araujo et al, 2011; Lima et al, 2011; Miagostovich et al, 1997a; Miagostovich et al, 1997b). Viral infiltration and subsequent infection of the CNS have been linked directly to CNS viral replication. Thus, the mechanism of invasion of the CNS requires reexamination and further studies. Later in this review, we will discuss all these mechanisms.

Role of leukocytes in CNS disease

Although there is a general consensus that flaviviruses can infect mononuclear cells in vivo, only few cell types have been identified to be targeted for infection in vitro. Indeed, autopsy studies clearly indicated that presence of flaviviruses within the Langerhan's cells located in the skin (Clyde et al, 2006). However, the participation of these or other cell types in the flaviviral pathogenesis has yet to be fully explored. Below we will discuss the evidence of leukocyte infections by flaviviruses.

Dendritic Cells (DC)

Following a bite from an infected mosquito, flaviviruses including DENV, WNV and JEV are inoculated into the human body and immediately interact with resident DC found in the epidermis and dermis layers of the skin. These cells originate from bone marrow as CD34+ progenitors and later differentiate into lymphoid/plasmacytoid (pDC; IL-3- and CD40 ligand-dependent maturation) or myeloid lineage (mDC; GM-CSF-dependent maturation). Following entry of DENV into DC, it has been observed that the total number of circulating DC's decreases and unresponsive thereby altering the host immune response. Furthermore, in vitro experiments utilizing mature PBMC-derived DC's showed an altered profile of inflammatory production (Hober et al, 1996a; Hober et al, 1996b). However, infiltration of infected DC's into the lymph nodes activates CD4+ and CD8+ T lymphocytes and elicits an adaptive immune response against the virus. Thus, flaviviruses use DC's as gateway cells to infiltrate and successfully infect their human host.

Monocyte/macrophages

Monocytes are one of the natural hosts of DENV (Durbin et al, 2008) and are implicated in the pathogenesis of DF and DHF (Halstead, 1988; Kliks et al, 1989). Interestingly, depletion of monocytes from murine models resulted in a ten–fold increase in viral load, suggesting that monocytes are also important in controlling viral infection (Fink et al, 2009). DENV infection also accelerates the differentiation of monocytes into macrophage and facilitates cellular transmigration into the CNS where inflammatory cytokines, chemokines, viral proteins, and other inflammatory factors are produced. Inflammation within the CNS leads to compromise and loss of endothelial function and potentially BBB dysfunction. Indeed, DENV infected CD16+ monocytes produce IL-1β, TNFα, CCL2, CCL3, and CCL4 (Azeredo et al, 2010). These cytokines and chemokines have been shown to be tightly involved in the loss of BBB integrity and the development of in CNS disease in the context of HIV CNS infection (Roberts et al, 2012a). These observations are also consistent with other viral diseases that compromise the CNS; for instance, it has been reported that HIV-infected patients have an elevated CD14+CD16+ monocyte population which can transmigrate into the CNS in response to CCL2 and produce inflammatory factors that disrupt the BBB (Eugenin et al, 2006b; Williams et al, 2012). However, to date, there are very few studies describing the neuropathogenesis of flaviviruses in this cell linage.

Lymphocytes

The role of lymphocytes in disease development following flaviviral infection has not yet been fully characterized due to various observed conundrums within the disease pathogenesis. In vitro studies have shown that DENV can infect as well as activate both CD4+ and CD8+ T cells (Mentor and Kurane, 1997; Pang et al, 2007). Indeed, DENV-specific CD4+ and CD8+ T cells have been identified among patients with DF (Gagnon et al, 1996) with the latter lymphocyte population important for controlling viral spread and replication within the host. However, studies have shown that CD8+ cytotoxic lymphocytes also contribute to DF pathogenesis and thus negatively impacting the health of the infected individual. The arrival of CD8+ T cells at the infection sites initiates the destruction of infected cells facilitating further production of inflammatory factors that lead to CNS damage and vascular endothelial dysfunction, resulting in BBB permeability (Mongkolsapaya et al, 2003; Rothman, 2009; Screaton and Mongkolsapaya, 2006). By inducing the production of inflammatory cytokines, chemokines, and other immune factors (e.g. IFN-γ TNF-α, IL-1β, IL-2, IL-4, Il-6, IL-7, IL-8, IL-18, CCL2, interferons and several soluble proteins such as CD4, sTNFR and CD8) during primary and secondary infections, the severity of the disease escalates and leads to the vascular permeability observed during DHF (Azeredo et al, 2010; Davis et al, 2008a; Hober et al, 1993; Kurane et al, 1991a; Kurane et al, 1991b). This supports the idea that T cell-produced inflammatory molecules are key factors that facilitate disease development following flavivirus infection with some promoting vascular permeability. This results in compromised BBB integrity and CNS dysfunction. Interestingly, despite JEV, WNV, and DENV belonging in the flaviviral family and causing similar outcomes, the viruses differ in their effects on the host immune system. Indeed it has previously been shown that IFNγ activity is inhibited by DENV by targeting STAT2 degradation whereas JEV and WNV does this by inhibiting STAT1 (Ashour et al, 2009; Laurent-Rolle et al, 2010; Lin et al, 2006; Mazzon et al, 2009).

B lymphocytes infected by DENV have been detected in both the spleen and bone marrow (Durbin et al, 2008; King et al, 1999; Lin et al, 2002) with active viral replication verified by PCR and Western blot analyses (Lin et al, 2002). Similar to T lymphocytes, the exposure of B cells to DENV in vitro induced production of inflammatory cytokine and DENV-specific immunoglobulins, both of which have been shown to be important in DENV pathogenesis and DF development. Since antibody production is essential in amplifying infection and inflammation, this suggests that the dysregulation of B cells is critical because they normally produce immunity against the serotype-specific E protein antigen (Mathew et al, 2011). During secondary infection, B lymphocyte response is predominately cross-reactive to other serotypes with the produced heterologous antibodies having greater avidity for the virus than homologous antibodies. This increased avidity for the pathogen is important during ADE of viral infection as observed in DENV pathogenesis (Lin et al, 2002).

During ADE of virus infection, DENV utilizes memory antibodies generated from previous DENV infection to bind to Fc receptors located on the extracellular surface facilitating their entry into the host cell. Interestingly, vaccination against any DENV serotypes results in full cytotoxic activity, cell proliferation, and controlled secretion of regulatory cytokines (Dharakul et al, 1994; Kurane et al, 1989; Malavige et al, 2004). While vaccination with the right serotype can evoke an effective immune response, some B cells produce auto-antibodies that undermine immunity and contribute to DENV pathogenesis (Malavige et al, 2004). Thus, B cells are targeted for DENV infection and contribute to disease development.

Similar to DENV, JEV infection elevates the levels of inflammatory molecules and chemokines (e.g. IFN-α, IFN-γ, IL-6, IL-8, IL-10, CXCL19, CXCL10, CXCL11, TNF-α, MIF, VEGF, sCD4, sCD8, TNFR, sIL-2, IL-1RA, and TGF-β) within the host. In addition, the secretion of matrix metalloproteinase-9 (MMP-9) at sites of infection (Tung et al, 2010), especially within the CNS, has been shown to be important in facilitating the breakdown of the endothelium leading to plasma leakage and BBB destruction. Similarly, the activation of lymphocytes and increased number of CD14+ cells following JEV infection has been correlated with the severity of the disease (Paessler and Walker, 2013; Rothman, 2009). Identifying mechanisms behind these changes is essential in targeting and designing therapeutic interventions that limit the devastating consequences of flavivirus infection.

Blood-brain barrier (BBB) dysfunction and CNS compromise

The BBB is a physical and highly specialized barrier separating the peripheral circulation from the CNS. This barrier is composed of multiple cell types, including brain microvascular endothelial cells (BMVEC), astrocytic end feet, pericytes, and perivascular macrophages, all of which are in close contact to neurons, glial cells and microglia (Spindler and Hsu, 2012b). Due to the high expression of tight junction proteins, the BBB is normally impermeable to most peripheral molecules, cells and restricts the diffusion of ions and small molecules.

Several viruses alter the BBB integrity and function, including HIV-1, human T lymphotropic viruse 1 (HTLV-1), lymphocytic choriomeningitis (LCMV), WNV, and simian immunodeficiency virus (SIV). The mechanism of BBB compromise involves transmigration of leukocytes and these viruses into the CNS parenchyma leading to BBB disruption, inflammation, and leukocyte transmigration into the CNS (Buckner et al, 2006; Eugenin and Berman, 2003; Eugenin et al, 2006b; Spindler and Hsu, 2012a; Williams et al, 2012). HIV transmigration and BBB disruption is the best examined cases of CNS viral invasion. Our laboratory and others have demonstrated that HIV infection alters BBB function by three different mechanisms: 1) alteration of the migratory properties of leukocytes, 2) changes in BBB permeability and 3) secretion of factors that compromise leukocyte migration and BBB integrity. We and others have also shown that HIV infection of leukocytes alters the expression of several adhesion molecules involved in leukocyte transmigration and increases the levels of important chemokine receptors such as CCR2 which increases the sensitivity to CCL2, a key chemokine involved in the pathogenesis of HIV CNS disease (Eugenin et al, 2006b; Roberts et al, 2012b). In addition, we demonstrated that BBB permeability among neuroAIDS patients is compromised at two stages: first, during productive HIV infection of leukocytes and second, following increased expression of CCL2 in the CNS. This combination results in BBB disruption potentially due to enhanced secretion of MMP's and others factors that highly compromise the expression and function of key barrier tight junction proteins such as occludin, claudin-1 and ZO-1 (Roberts et al, 2012a; Roberts et al, 2012b). Some factors involved in HIV neuropathogenesis include cleaved forms of adhesion molecules, such as PrPc and PECAM-1, with the latter competing with the PECAM-1-PECAM-1 or PrPC-PrPC interactions between endothelial cells. This direct competition leads to destabilization of the endothelium, compromising BBB integrity thereby facilitating leukocyte migration into the CNS (Eugenin et al, 2006a; Roberts et al, 2012a). However, our experiments using primary BMVEC's, indicate that CCL2 binding to its corresponding receptor alone has profound effects on tight junction protein expression and localization, resulting in BBB compromise by altering β-catenin distribution and interaction with adherence junctions and PECAM-1 (Roberts et al, 2012a; Roberts et al, 2012b). Thus, HIV CNS invasion is a highly regulated process and involves several host and viral components. But whether these mechanism are similar in flaviviruses is totally unknown.

Among the best described cases of CNS infection involves WNV. Due to the close relation of WNV with DENV and JEV, we expected that the mechanism of neuropathogenesis of these viruses to be similar (Figure 6). WNV attacks the brain causing neurotropic effects that include encephalitis and fever. In mouse models, WNV-associated neurologic disease is characterized by BBB disruption, increased leukocyte infiltration, inflammation, and neuronal loss (Glass et al, 2005; Samuel and Diamond, 2006). Some of these effects have been associated with the loss of BBB function due to degradation of junctional proteins such as ZO-1, claudin-1, occludin, JAM-A, β-catenin, VE-cadherin by secreted metalloproteinases (MMP) including −1, −3 and −9 (Roe et al, 2012a; Roe et al, 2012b). In addition, WNV infects human BMVEC's which could lead to a compromised BBB by inflammatory mechanisms described previously (Verma et al, 2009). Experiments using WNV-infected endothelial cells indicate an up-regulation of MHC–I and II, ICAM-1, VCAM-1, E- and P-selectin (King et al, 2003) which can contribute to leukocyte adhesion to the BBB and leukocyte infiltration into the CNS. During WNV infection, CD19+B220-BST-2+ leukocytes have been described as a major leukocyte population that transmigrates into the CNS and contributes to the development of encephalitis (Brehin et al, 2008). However, the mechanisms by which these cells transmigrate into the CNS remain unknown.

An external file that holds a picture, illustration, etc.
Object name is nihms-633656-f0006.jpg

Infection of CNS by flaviviruses. (1) Infection of the CNS occurs either through the adherence of the virus to molecules present on the surface of brain microvascular endothelial cells (BMVEC's) or infiltration of infected monocytes across the BBB. Viral infiltration then leads to infection of the BBB and CNS cell populations. (2) Infection of human astrocytes leads to chemokine production facilitating further recruitment of monocytes and macrophages. (3) Neurons infected by flaviviruses undergo apoptosis and activates the resident microglia population which produces an inflammatory response. Production of inflammatory cytokines (e.g. TNF-α, IL1β, INF-γ and IL-4), chemokines (e.g. CCL2, CCL5, CXCL9, CXCL10), inflammatory enzymes (COX2) and matrix-metalloproteinases (MMPs) leads to degradation of the endothelial barrier and the release of inflammatory factors (5) recruiting CD4+ and CD8+ T lymphocytes into the CNS parenchyma. Infiltration of CD4+/CD8+ T lymphocytes leads to further inflammation and eventually CNS damage.

Flavivirus infection of the CNS, especially with DENV, has been reported by a few studies with contradictory results. Viral RNA has been detected in the cerebrospinal fluid obtained from encephalitic individuals (Kumar et al, 2008; Miagostovich et al, 1997b). In addition, DENV antigens have been detected in neurons, astrocytes, and microglia (Bhoopat et al, 1996b; Ramos et al, 1998). While in situ hybridization for viral particles in neurons, BMVEC's, and glial cells were negative (Jessie et al, 2004), perivascular macrophages were positive for flaviviral NS3 protein. Further studies of these and other CNS cells are required to clarify their role in the neuropathogenesis of DENV and other flaviviruses. Thus, it is unclear whether proteins or RNA are product of replication, viral uptake or both.

Mechanisms of neuronal compromise

For a long time, the neurologic involvement of flavivirus infection was not considered and was only associated with peripheral infection, such as loss and extravasation of fluid, hyponatremia, and systemic failure. However, some reports indicating the unequivocal presence of viral proteins and genetic viral material in the brain and CSF of infected individuals suggest otherwise (Araujo et al, 2012; Soares et al, 2006; Solomon et al, 2000). Thus, the nature and mechanisms of CNS injury induced by these viruses must be examined. Several members of the flavivirus family also induced neuronal apoptosis in association with post-mitotic neuronal apoptosis that cannot be regenerated by neuronal stem cells (Ogata et al, 1991; Pekosz et al, 1996). Experiments in cell lines indicate that DENV can directly infect neurons (Despres et al, 1998) resulting in permanent damage.

Despite the problems with animal models for flavivirus infections, DENV has been easily adapted to invade the CNS of rodents. Normally, newborn mice are insensitive to non-neuroadapted mouse DENV strains. By passaging the virus within a murine host, the virus is neuroadapted to the murine host with highly neurovirulent strains of the virus selected and amplified (Despres et al, 1998; Sabin, 1952). This selection process produces viruses that are capable of inducing neuronal apoptosis in infected mice, especially in the hippocampus and cerebral cortex where viral CNS replication has been observed (Despres et al, 1998). In addition, several DENV infection animal models display interesting effects on neuroinflammation characterized by up-regulation of important chemokines (e.g. CCL2, CCL5, CXCL1, and CXCL2) and inflammatory cytokines (e.g. TNF-α and IFN-γ) that facilitate leukocyte infiltration into the CNS (Amaral et al, 2011a; Amaral et al, 2011b).

Mice infected intracerebrally with DENV-3 show progressive meningo-encephalitis characterized by CNS infiltration of neutrophils and mononuclear cells by a mechanism that correlates with a chemokine-dependent mechanism that includes CCL2 and CXCL12 (Amaral et al, 2011a; Amaral et al, 2011b). These results show the importance of these cell types in the development of CNS disease during DENV infection. Interestingly, similar mechanisms of CNS damage have been observed in mouse models of hepatitis virus (JHMV) which resulted in animal mortality (Zhou et al, 2003). Neutrophils have a biphasic role during WNV infection, first, serving as a reservoir for viral replication and second, aiding in viral clearance (Bai et al, 2007). However, these conflicting roles of neutrophils during WNV pathogenesis have not been examined further.

It has been observed that WNV causes limbic seizures by a mechanism involving N-methyl-D-aspartic acid receptor (NMDA) activation. Blocking this receptor activation could therefore abrogate limbic seizures and prolong animal survival (Getts et al, 2007). In addition to the neuronal damage induced by WNV infection, infiltration of leukocytes (e.g. neutrophils and mononuclear cells) was detected in the CNS parenchyma, similar to what was seen during DENV-3 infection of the CNS. WNV-mediated recruitment of microglial precursors (i.e. Ly6c+ inflammatory monocytes) indicates that the virus can pass through the BBB by infected leukocyte transmigration into the CNS and induce local inflammation (Getts et al, 2008). Once in the CNS, the virus can infect and replicate within neurons and astrocytes, but not microglia (Cheeran et al, 2005; Diniz et al, 2006; Hussmann et al, 2013) despite data illustrating microglial activation and secretion of cytokines and chemokines that lead to CNS dysfunction.

Meanwhile, infection of CNS cells with JEV, especially neurons, results in massive CNS compromise, encephalitis, and death (Chen et al, 2004; German et al, 2006; Ghoshal et al, 2007). Apoptosis induced by JEV has been linked to three related mechanisms: 1) direct infection of neurons by the virus, 2) infection of other CNS cells such as microglia and astrocytes, and 3) general inflammation (Chen et al, 2004; Das and Basu, 2008; Das et al, 2008; Raung et al, 2005; Raung et al, 2007; Swarup et al, 2008). It has been shown that inflammatory cells play an important role in the onset, progression, and severity of JEV-mediated encephalitis (Khanna et al, 1991; Mathur et al, 1988; Singh et al, 2000). Furthermore, reports indicate that the levels of inflammation can be a predictor of patient outcome following infection (Ravi et al, 1997). However, compared to other flaviviruses, the extent of glial infection during JEV is unclear. It is known that JEV triggers neuronal apoptosis by inducing endoplasmic reticulum (ER) stress through p38 mitogen-activated protein kinase (MAPK)-dependent and death-related transcription factor CHOP (C/EBP homologous protein) mediated pathways (Su et al, 2002). This is in agreement with WNV which also uses CHOP and mitochondrial pathways to induce apoptosis (Chu and Ng, 2003; Medigeshi et al, 2007). Additionally, phosphatidylinositol 3-kinase (PI3K), AKT, superoxide, arachidonic acid, caspases and alterations in bcl-2/Bax also participate in apoptosis (Courageot et al, 2003; Jan et al, 2000; Lee et al, 2005; Lin et al, 1997; Parquet et al, 2001). Our work, in HIV, a clear neurotropic virus, indicates that despite the low numbers of HIV-infected astrocytes which support minimal to undetected replication, the bystander mechanism of cell death amplification can be used by the virus to induce apoptosis and inflammation using electrical and chemical synapses (Eugenin and Berman, 2007; Eugenin et al, 2011). Therefore, the number of virus infected cells or the levels of viral replication are not good indicators of cellular damage.

Cytochrome C is a mitochondrial protein that has been shown to play an important role in cellular respiration. In addition, cytochrome C participates in cellular apoptosis following cell damage or infection. DENV infection triggers neuronal apoptosis by activation of phospholipase A2 (PLA2), superoxide anion generation, cytochrome C release from the mitochondria, caspase-3 activation and NF-kB translocation (Jan et al, 2000). This is similar to our results which show that HIV infection of glial cells dysregulates cytochrome C. Despite the high amounts of cytotoxic intracellular cytochrome C, HIV-infected cells are protected from apoptosis (Eugenin and Berman, 2013). Thus, future studies are required to identify the role of these factors in apoptosis, the generation of viral reservoirs, and the detection of flaviviruses.

Therapeutic approaches to flaviviral infections

Current management of flavivirus infections

There are no effective antiviral treatments or vaccines against DENV and WNV with only one vaccine available against JEV. Thus, infected patients can only provide with palliative care following diagnosis. DHF patients exhibiting hemorrhage as determined by lowered hematocrit levels are provided blood transfusions to improve clinical outcome. Furthermore, there are no treatments available that prevent, abolish, or reverse the devastating CNS consequences of flavivirus infection. Thus, elucidating the mechanisms utilized by flaviviruses to compromise the CNS will lead to better design of successful therapeutics against these pathogens. To date, virus control has largely involved either the administration of prophylactic measures (e.g. vaccines) or the control of flaviviral arthropod vectors, namely the Culex and Aedes mosquito species. Indeed, given the rapid global spread of these arthropods, the latter is the primary approach indicated by the CDC and the WHO to contain the spread of DENV, WNV, and JEV. Introduction and spread of DENV and WNV, as well as the presence of suitable JEV vectors in non-endemic areas, will greatly increase the number of individuals who are threatened by these pathogens.

Vaccine development

Due to the lack of effective therapy against most flaviviruses, the focus on developing potential vaccines has escalated. Considering the increasing prevalence of WNV in the United States, as well as the globalization of DENV, development of successful vaccines is imperative for effectively preventing flaviviral-associated diseases (Chao et al, 2012; Ehrenfeld et al, 2009; Sutter et al, 2000). However, any such success requires further knowledge of flaviviral disease pathogenesis. Identifying viral and host components important for disease development may lead to new targets for vaccines and therapeutic agents against the viruses. (Heinz and Stiasny, 2012).

The flavivirus genome encodes for a polyprotein that is post-translationally cleaved into 10 structural and non-structural components (see details in Figure 5). Since all are important in the life cycle and pathogenesis of the virus, they have the capacity to be utilized for vaccine development. Vaccines for the three structural proteins C, prM, and E are already being developed with the most emphasis on multivalent vaccines, which contain multiple immunostimulatory antigenic epitopes that simultaneously generate protection against multiple viruses and serotypes (Brewoo et al, 2012; Durbin et al, 2006; Ishikawa et al, 2011; Johnson et al, 2002; Johnson et al, 2004). Numerous studies have successfully produced chimeric vaccines containing the viral envelope including the prM/E portion of the structural protein as well as regions encompassing important non-structural components (e.g. NS1 and NS5). The ADE mechanism facilitating secondary and other subsequent DENV infections is still a challenge that must be addressed in these vaccines. Indeed, the limited heterotypic protection against other DENV serotypes has slowed DENV vaccine development. Production of multivalent vaccines eliciting simultaneous immunogenic responses against all DENV serotypes may lead to successful prevention of DF and DHF. There has been modest success developing live-attenuated, intertypic DENV chimeras containing replacements of the C, prM, E and/or NS1 regions of DENV 4 with the corresponding regions within the DENV-1 or DENV-2 genome (Bray and Lai, 1991). Monkeys singly or mix inoculated with these chimeras successfully showed simultaneous seroprotection against DENV-1 and -2 while at the same time producing DENV 4-specific neutralizing antibodies (Bray et al, 1996). To date, however, there are no vaccines in use that can simultaneously target all four DENV serotypes. Therefore, creating chimeric, tetravalent vaccines protecting against all four DENV serotypes could lead to promising prophylactic therapies against this emerging virus and prevent the more severe disease, DHF. Sanofi Pasteur has shown exciting and groundbreaking results in the development of a tetravalent vaccine against all four DENV serotypes. Currently, the vaccine is in phase III clinical study which is being conducted in DENV endemic and non-endemic countries (www.dengue.info). Successfully producing tetravalent vaccines will alleviate the drawback of ADE-associated with DENV infection and protect individuals from heterotypic infection with other DENV serotypes.

IMOJEV, meanwhile, is a chimeric JEV vaccine that integrated the prM and E genes of SA14-14-2 (Liu et al, 2011) into the Yellow fever vaccine vector, YFV17D (Poland et al, 1981). Remarkably, the vaccine carries a 100% seroconversion rate with greater than 85% seroprotection six months post-vaccination with continued protection against heterologous serotypes up to five years post inoculation (Appaiahgari and Vrati, 2012a; Appaiahgari and Vrati, 2012b) as well as the ability to provide continuous protection after only a single immunization dose. More interesting is the reported cross-protection of IMOJEV against the JEV-related flaviviruses such as Murray Valley encephalitis virus and WNV-Kunjin strain, however, the mechanism for this cross-protection has not yet been identified (Lobigs et al, 2009). Finally, an important feature of IMOJEV is the incorporated high-fidelity RNA polymerase within the YF17D genome which decreases the likelihood that the virus will undergo genetic mutations and revert back into its virulent state (Pugachev et al, 2004). Together, these positive attributes make IMOJEV a highly promising prophylaxis against JEV, and with further development, against other related flaviviruses.

The promising results observed during the development of multivalent DENV vaccines have led to the design of other viral chimeras integrating genomic markers from JEV, WNV, or DENV within the previously successful Yellow Fever virus (YFV) vaccine backbone (Arroyo et al, 2004; Chambers et al, 2003; Chambers et al, 1999; Guirakhoo et al, 2000; Guirakhoo et al, 1999; Monath et al, 1999; Querec et al, 2006). Unlike WNV and DENV, there has been significant progress in JEV vaccine development since the initial observation of acquired protection against JEV among accidentally exposed laboratory workers (Hammon and Sather, 1973; Pulmanausahakul et al, 2011). Since then, there is now one vaccine currently licensed for use in the United States (IXIARO) along with three vaccines in JEV endemic countries (e.g IMOJEV, SA14-14-2 live attenuated vaccine, and the now discontinued JE-VAX). Following the discontinuation of JE-VAX due to some reported adverse reactions to the vaccine, IXIARO, a cell-culture based inactivated virus derived from the SA-14-14-2 strain, is the only inactivated vaccine being produced and administered today. Still, the relatively limited effectiveness of IXIARO compared with live-attenuated vaccines (~80-90%) (Halstead and Thomas, 2011), reported reactions to the inactivated vaccine (Nazareth et al, 1994; Ohtaki et al, 1995; Plesner, 2003; Plesner and Ronne, 1997), and the requirement of multiple doses for complete protection (Pugachev et al, 2003) have led to searches for more immunogenic vaccines with less adverse effects. Production of live-attenuated SA14-14-2 vaccine in 1988 provided a seroconversion of 80% after a single dose with subsequent immunizations producing efficacy rates of 95-99% (Hase et al, 1993; Hennessy et al, 1996; Wills et al, 1992; Xin et al, 1988). While not as effective as IMOJEV, further development of SA14-14-2 could lead to a more effective live-attenuated vaccine. SA14-14-2 is an attenuated strain of JEV-SA14 produced though serial passaging of the virus within hamster kidney cells. Due to the low cost of its production, as well as minimal side effects, live-attenuated SA14-14-2 is being administered more prevalently than inactivated vaccines (Schioler et al, 2007). Indeed, since its approval for public, over 200 million children living in JEV-endemic areas have been immunized with the live-attenuated vaccine and accounts for over 80% of the total number of JEV vaccinations (Appaiahgari and Vrati, 2012b; Liu et al, 2011). While there have been no reports regarding SA-14-14-2 mutational reversion to its virulent state, such a risk remains a potential drawback for this and other live-attenuated viruses.

The reversion of live-attenuated viruses to their original pathogenic state and the side effects, including hypersensitivity against components within inactivated vaccines, suggest that there is a need for new prophylactic treatments against JEV and other flaviviruses. Since the initial concept of using plasmids as immunogens two decades ago, the development of DNA vaccines has advanced dramatically leading to the production of immunogenic agents which are in various phases of clinical trials (Ferraro et al, 2011). For instance, DNA vaccines have started being developed as prophylactic therapies against a variety of human pathogens including HIV (Ramirez et al, 2013), Plasmodium falciparum (Chuang et al, 2013), and Mycobacterium tuberculosis (Okada et al, 2012) with promising results. Inoculations of the host with engineered viral protein expression plasmids are sufficient to elicit a robust cytotoxic CD8+ and helper CD4+ T lymphocyte response as well as the production of protective antibodies against the expressed immunogens (Alarcon et al, 1999). This exciting technology has some important implications especially in the development of DENV vaccines as it could provide concurrent protection against all DENV serotypes thereby limiting the adverse effects of ADE which current vaccines have been unable to circumvent. This novel approach is being applied to develop new flavivirus vaccines that target the surface exposed E and prM and NS1 proteins which were previously shown to be highly immunogenic and elicit a favorable immune response (Ahsan and Gore, 2011; Azevedo et al, 2013; Costa et al, 2007; Costa et al, 2006a; Costa et al, 2006b; Davis et al, 2008b; Kulkarni et al, 2012; Lu et al, 2013; Schneeweiss et al, 2011). A DNA vaccine targeting DENV (designated D1ME100) and WNV previously entered in phase 1 of clinical trials produced favorable results (Beckett et al, 2011). Both D1ME100 and WNV vaccines elicited strong, long-lasting immune response in all test subjects as quantified by virus-specific IgG and IgM production. Even more interesting, despite only being immunized against DENV1, D1ME100-vaccinated subjects showed significant IFNγ response following inoculation with E proteins derived from all four DENV serotypes (Beckett et al, 2011). Together, these data show great promise for DNA vaccines in the prevention of flavivirus infections as well as infections by any pathogen lacking available treatments or vaccines.

Viral inhibitors

In order to successfully infect a suitable host, a virus must attach to the host's extracellular surface and penetrate into the cytosol. There, the viral genome is unpackaged, transcribed, and translated. Progeny viruses are then processed and packaged by host and viral machineries to produce mature, infectious virions. While vaccines have been highly successful in preventing important human infections such as polio and smallpox, research for antiviral therapeutic agents (i.e. small molecule inhibitors) is still being pursued for one major reason: identifying homologous targets across related pathogens can lead to the design of pharmaceutical drugs that will help eradicate numerous related human diseases. Close dependence of the flaviviral life cycle for host cellular processes allowed researchers to utilize these host functions in their design of antiviral agents. Indeed, numerous cellular components have been targeted, including viral adherence to the host extracellular surface (De Burghgraeve et al; Schmidt et al, 2012), nucleic acid and protein synthesis (Noisakran et al, 2008; Oh et al, 2006; Paranjape and Harris, 2007; Qing et al, 2009), intracellular trafficking and signaling (Hirsch et al, 2005; Honget al, 2013), and the host immune response (Dikeakos et al, 2010; Mishra et al, 2009; Proudfootet al, 1999). The ADE of DENV infection, which has hindered the production of successful DENV vaccines, has also been the primary reason for the design of more effective viral inhibitors.

Only a few inhibitors are currently available that prevent flaviviral attachment and entry. Due to the importance of E protein in flaviviral entry and its numerous binding targets on host cell membranes, it has been the focus for viral entry inhibitors (Liao et al, 2010; Schmidt et al, 2012). Schmidt, et. al. has shown that a DENV E protein inhibitor, designated 1662G07, and its analogs could successfully prevent DENV 2 fusion with host endosomes at micromolar to sub-micromolar concentrations (IC90 = 0.75 – 2 μM) when tested in vitro (Schmidt et al, 2012). In addition, the inhibitor has been shown to be effective against DENV 1 and 2 serotypes.

LCTA-949, a replication inhibitor of hepatitis C (Obeid et al, 2011), is another compound being developed against flaviviral infections, in particular DENV (De Burghgraeve et al). Treatment with LCTA-949 (12.5 μM) was observed to prevent DENV entry into host cells by limiting viral attachment to the cell membrane; however, this inhibitory activity occurs only when added concurrently with the virus in vitro. This suggests that the binding affinity of the virus for the host cell membrane maybe greater than viral adherence to the inhibitor. Even more interesting is the ability of LCTA-949 to competitively inhibit the attachment of antibody-opsonized DENV on the host cell surface. This suggests the compound's potential inhibitory activity against ADE of DENV infection. Due to its incapacity to reverse pre-existing DENV infections, LCTA-949 must be developed further in order for it to become clinically relevant. Inhibition of other flaviviral pathogens (i.e. hepatitis C and Yellow Fever viruses) and interference with ADE-DENV infections may make this novel compound a viable therapeutic agent against flaviviral infections in the future.

Similar to virus attachment and entry, post-translational processing of the translated viral genome is important for a successful viral life cycle and production of mature, infectious virions. Indeed, host signalases and viral proteases (NS2b and NS3) have been shown to be required for cleavage of the flaviviral polyprotein into biologically active components (Appaiahgari and Vrati, 2012b; Leyssen et al, 2000). Inhibiting these proteases has been proposed to abate virus formation and maturation (Filocamo et al, 1999). The importance of viral proteases during flavivirus pathogenesis, as well as in other viral infections, makes them attractive targets for therapeutic intervention. For instance, one anti-HIV therapy involves the use of a protease inhibitor, Duranvir (Prezista; Janssen Therapeutics), which targets the HIV-1 PR protease important for the processing of the Gag/Gag-Polyprotein (Phung and Yeni, 2011). This same approach is being applied towards flaviviruses by targeting the NS3 protease (Chappell et al, 2008; Lescar et al, 2008; Leyssen et al, 2000). In fact, a protease inhibitor (Boceprevir; Merck) targeting the NS3 protease of HCV has yielded impressive results in clinical trials (Manns et al). During the study, HCV loads among treated patients were drastically reduced as compared with naïve subjects. The activity of Boceprevir has not yet been tested against other related flaviviruses. Current results, however, suggest that Boceprevir and other NS3-targeting protease inhibitors are excellent candidates for treatment against DENV, WNV, and JEV infections.

Another potential enzyme targeted for flavivirus therapy is guanylyltransferase (GTase) encoded within the NS5 regions of the flavivirus genome. It has previously been shown that GTases in concert with viral RNA triphosphatase, methytransferase, and nucleoside 2’-O-methyltransferase, regulate flavivirus genome translation and preserve viral RNA integrity by methylating the 5’-end UTR of the flavivirus genome. (Issur et al, 2009). This, together with its conservation among related flaviviruses, suggests that GTases could be targeted for therapeutic intervention against members of this virus family (Bollati et al; Egloff et al, 2002; Geiss et al, 2009). Indeed, inhibition of GTase activity using the compound (E)-[3-[5-[4-tert-butylbenzylidene-4-oxo-2-thioxo-1,3-thiazolidin-3-yl] propanoic acid (BG-323) prevented DENV and WNV replication (Stahla-Beek et al, 2012). Furthermore, this same study showed that BG-323 may also have anti-viral activity against the Yellow Fever virus. It is possible to speculate that given its broad activity of against flaviviruses, BG-323 could be further developed into a successful therapy against flavivirus infections; further, against the severe diseases DF and DHF.

Vector control: a potential effective way to protect the population

The spread of mosquito vectors is a critical event in the transmission cycle of DENV, WNV and JEV. Thus, preventing or reducing viral transmission depends upon either the control of these insect vectors or interruption of the human-vector interaction. Some of these measures included in the list below which provides a comprehensive approach in designing better policies and programs to detect, eliminate, and attack the critical stages of vector development and viral transmission.

-Elimination of potential habitats by controlling local environments facilitating the successful reproduction of the mosquitoes. These recommendations involve simple steps targeting the early stages of vector development such as better management of natural and man-made stagnant water sources as well as chemical and biological control methods of mosquito populations (e.g. insecticides, repellants, mosquito fish, and Bacillus thuringiensis). These simple steps will help reduce the presence of larvae and adult mosquito populations thereby limiting the potential contact between infected arthropod vectors and humans.

- Politics and social advocacies are essential factors which allow the implementation of policies educating individuals about the global threat of flaviviral dispersion. Designing better policies will provide the general population and health care workers important medical training and preparation to face these important viral threats. This is even more important in areas previously not endemic to DENV, WNV, or JEV as health workers are not typically trained to identify these infectious diseases and thus potentially confuse the infections with other flu-like maladies. Given the increasing global spread of these viruses and its mosquito vectors, dispersal of medical information as well as policies aimed at preventing further spread of the pathogens is necessary.

- A better communication between health organizations and the population is essential in preventing disease spread. The spread of WNV, DENV, and JEV may be influenced by the lack of information about viral transmission, disease identification, disease treatment, and infection statistics. These problems are exacerbated among resource poor countries where flaviviruses typically found. Lack of resources and health services could impede the public diffusion of educational information important in controlling either the viruses or its insect vectors. Proper education would help alleviate this problem as it will lead to productive discussions regarding the control of these pathogens subsequently preventing viral transmission and the diseases associated with the infection.

Concluding remarks and future directions

The emergence of WNV in the United States, the world-wide expansion of DENV, the continuous threats of JEV, and the rapid dispersal of suitable mosquito vectors make research involving these pathogens imperative. It is clear that drastic global changes are affecting the spread of flaviviruses and their vectors. Further, with the rapidly expanding human population and the encroachment of these viruses in previously non-endemic areas, a global health problem involving these viruses is imminent. Thus, understanding how these viruses cause human diseases such as DHF is imperative. In addition, the ever increasing threats of new infections make the development of novel, effective vaccines and therapeutic agents vital. The sparse, conflicting information available regarding the pathogenesis of these viruses and the cell types, tissues, and organs they infect, however, make it difficult to produce successful drugs against any of these pathogens. Thus, until successful treatments and more effective vaccines are available, the best method of eliminating new infections in public and medical institutions is to stop viral vector spread and prevent becoming bitten by mosquitoes through the copious use of repellents.

The mechanisms of CNS dysfunction induced by flavivirus infection are poorly explored or ignored. The cell targets of flaviviruses and the replication pattern used by these viruses are still a matter of debate and require further investigation. Identifying how flaviviruses alter host cell physiology will provide the basis for understanding their pathogenesis and will subsequently lead to the design of more effective therapeutic agents against the viruses within the CNS. Recent advancements in research techniques and animal models and our data using primary human astrocytes and BMVEC's provided critical evidence suggesting that flaviviruses are indeed neurotropic. Further, identifying how these viruses regulate leukocyte activation and CNS transmigration as well as dysregulation of BBB function leading to CNS compromise and human disease is critically important.

Acknowledgements

This work was supported by funds from the Public Health Research Institute (PHRI) and National Institutes of Mental Health grant MH096625. We thank the PHRI shared facilities at Rutgers University, Newark, NJ (http://phri.org/facilities/facil_imaging.asp). We also thank Jonathan Guito for critically reading and proof-reading this manuscript.

Footnotes

The authors declare that they have no conflict of interest.

References

  • Ahsan MF, Gore MM. Comparison of immune response generated against Japanese encephalitis virus envelope protein expressed by DNA vaccines under macrophage associated versus ubiquitous expression promoters. Virol J. 2011;8:382. [PMC free article] [PubMed] [Google Scholar]
  • Alarcon JB, Waine GW, McManus DP. DNA vaccines: technology and application as anti-parasite and anti-microbial agents. Adv Parasitol. 1999;42:343–410. [PubMed] [Google Scholar]
  • Aleyas AG, George JA, Han YW, Rahman MM, Kim SJ, Han SB, Kim BS, Kim K, Eo SK. Functional modulation of dendritic cells and macrophages by Japanese encephalitis virus through MyD88 adaptor molecule-dependent and -independent pathways. J Immunol. 2009;183:2462–74. [PubMed] [Google Scholar]
  • Aleyas AG, Han YW, George JA, Kim B, Kim K, Lee CK, Eo SK. Multifront assault on antigen presentation by Japanese encephalitis virus subverts CD8+ T cell responses. J Immunol. 2010;185:1429–41. [PubMed] [Google Scholar]
  • Amaral DC, Rachid MA, Vilela MC, Campos RD, Ferreira GP, Rodrigues DH, Lacerda-Queiroz N, Miranda AS, Costa VV, Campos MA, Kroon EG, Teixeira MM, Teixeira AL. Intracerebral infection with dengue-3 virus induces meningoencephalitis and behavioral changes that precede lethality in mice. J Neuroinflammation. 2011a;8:23. [PMC free article] [PubMed] [Google Scholar]
  • Amaral DC, Rachid MA, Vilela MC, Campos RD, Ferreira GP, Rodrigues DH, Lacerda-Queiroz N, Miranda AS, Costa VV, Campos MA, Kroon EG, Teixeira MM, Teixeira AL. Intracerebral infection with dengue-3 virus induces meningoencephalitis and behavioral changes that precede lethality in mice. Journal of neuroinflammation. 2011b;8:23. [PMC free article] [PubMed] [Google Scholar]
  • Anders KL, Nguyet NM, Chau NV, Hung NT, Thuy TT, Lien le B, Farrar J, Wills B, Hien TT, Simmons CP. Epidemiological factors associated with dengue shock syndrome and mortality in hospitalized dengue patients in Ho Chi Minh City, Vietnam. Am J Trop Med Hyg. 2011;84:127–34. [PMC free article] [PubMed] [Google Scholar]
  • Anderson KB, Gibbons RV, Thomas SJ, Rothman AL, Nisalak A, Berkelman RL, Libraty DH, Endy TP. Preexisting Japanese encephalitis virus neutralizing antibodies and increased symptomatic dengue illness in a school-based cohort in Thailand. PLoS Negl Trop Dis. 2011;5:e1311. [PMC free article] [PubMed] [Google Scholar]
  • Appaiahgari MB, Vrati S. Clinical development of IMOJEV (R)--a recombinant Japanese encephalitis chimeric vaccine (JE-CV). Expert opinion on biological therapy. 2012a;12:1251–63. [PubMed] [Google Scholar]
  • Appaiahgari MB, Vrati S. Clinical development of IMOJEV (R)--a recombinant Japanese encephalitis chimeric vaccine (JE-CV). Expert Opin Biol Ther. 2012b;12:1251–63. [PubMed] [Google Scholar]
  • Araujo F, Nogueira R, Araujo Mde S, Perdigao A, Cavalcanti L, Brilhante R, Rocha M, Vilar DF, Holanda SS, Braga Dde M, Sidrim J. Dengue in patients with central nervous system manifestations, Brazil. Emerging infectious diseases. 2012;18:677–9. [PMC free article] [PubMed] [Google Scholar]
  • Araujo FM, Brilhante RS, Cavalcanti LP, Rocha MF, Cordeiro RA, Perdigao AC, Miralles IS, Araujo LC, Araujo RM, Lima EG, Sidrim JJ. Detection of the dengue non-structural 1 antigen in cerebral spinal fluid samples using a commercially available enzyme-linked immunosorbent assay. Journal of virological methods. 2011;177:128–31. [PubMed] [Google Scholar]
  • Arroyo J, Miller C, Catalan J, Myers GA, Ratterree MS, Trent DW, Monath TP. ChimeriVax-West Nile virus live-attenuated vaccine: preclinical evaluation of safety, immunogenicity, and efficacy. J Virol. 2004;78:12497–507. [PMC free article] [PubMed] [Google Scholar]
  • Ashour J, Laurent-Rolle M, Shi PY, Garcia-Sastre A. NS5 of dengue virus mediates STAT2 binding and degradation. J Virol. 2009;83:5408–18. [PMC free article] [PubMed] [Google Scholar]
  • Azeredo EL, Neves-Souza PC, Alvarenga AR, Reis SR, Torrentes-Carvalho A, Zagne SM, Nogueira RM, Oliveira-Pinto LM, Kubelka CF. Differential regulation of toll-like receptor-2, toll-like receptor-4, CD16 and human leucocyte antigen-DR on peripheral blood monocytes during mild and severe dengue fever. Immunology. 2010;130:202–16. [PMC free article] [PubMed] [Google Scholar]
  • Azevedo AS, Goncalves AJ, Archer M, Freire MS, Galler R, Alves AM. The Synergistic Effect of Combined Immunization with a DNA Vaccine and Chimeric Yellow Fever/Dengue Virus Leads to Strong Protection against Dengue. PLoS One. 2013;8:e58357. [PMC free article] [PubMed] [Google Scholar]
  • Bai F, Town T, Pradhan D, Cox J, Ashish, Ledizet M, Anderson JF, Flavell RA, Krueger JK, Koski RA, Fikrig E. Antiviral peptides targeting the west nile virus envelope protein. Journal of virology. 2007;81:2047–55. [PMC free article] [PubMed] [Google Scholar]
  • Beatty ME, Vorndam V, Hunsperger EA, Munoz JL, Clark GG. Travel-Associated Dengue Infections - United States, 2001--2004. Morbidity and Mortality Weekly Report. 2005;54:556–558. [PubMed] [Google Scholar]
  • Beckett CG, Tjaden J, Burgess T, Danko JR, Tamminga C, Simmons M, Wu SJ, Sun P, Kochel T, Raviprakash K, Hayes CG, Porter KR. Evaluation of a prototype dengue-1 DNA vaccine in a Phase 1 clinical trial. Vaccine. 2011;29:960–8. [PubMed] [Google Scholar]
  • Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein JS, Hoen AG, Sankoh O, Myers MF, George DB, Jaenisch T, Wint GR, Simmons CP, Scott TW, Farrar JJ, Hay SI. The global distribution and burden of dengue. Nature. 2013;496:504–7. [PMC free article] [PubMed] [Google Scholar]
  • Bhoopat L, Bhamarapravati N, Attasiri C, Yoksarn S, Chaiwun B, Khunamornpong S, Sirisanthana V. Immunohistochemical characterization of a new monoclonal antibody reactive with dengue virus-infected cells in frozen tissue using immunoperoxidase technique. Asian Pacific journal of allergy and immunology / launched by the Allergy and Immunology Society of Thailand. 1996a;14:107–13. [PubMed] [Google Scholar]
  • Bhoopat L, Bhamarapravati N, Attasiri C, Yoksarn S, Chaiwun B, Khunamornpong S, Sirisanthana V. Immunohistochemical characterization of a new monoclonal antibody reactive with dengue virus-infected cells in frozen tissue using immunoperoxidase technique. Asian Pac J Allergy Immunol. 1996b;14:107–13. [PubMed] [Google Scholar]
  • Bollati M, Alvarez K, Assenberg R, Baronti C, Canard B, Cook S, Coutard B, Decroly E, de Lamballerie X, Gould EA, Grard G, Grimes JM, Hilgenfeld R, Jansson AM, Malet H, Mancini EJ, Mastrangelo E, Mattevi A, Milani M, Moureau G, Neyts J, Owens RJ, Ren J, Selisko B, Speroni S, Steuber H, Stuart DI, Unge T. Bolognesi M Structure and functionality in flavivirus NS-proteins: perspectives for drug design. Antiviral Res. 87:125–48. [PMC free article] [PubMed] [Google Scholar]
  • Bray M, Lai CJ. Construction of intertypic chimeric dengue viruses by substitution of structural protein genes. Proc Natl Acad Sci U S A. 1991;88:10342–6. [PMC free article] [PubMed] [Google Scholar]
  • Bray M, Men R, Lai CJ. Monkeys immunized with intertypic chimeric dengue viruses are protected against wild-type virus challenge. J Virol. 1996;70:4162–6. [PMC free article] [PubMed] [Google Scholar]
  • Brehin AC, Mouries J, Frenkiel MP, Dadaglio G, Despres P, Lafon M, Couderc T. Dynamics of immune cell recruitment during West Nile encephalitis and identification of a new CD19+B220-BST-2+ leukocyte population. J Immunol. 2008;180:6760–7. [PubMed] [Google Scholar]
  • Brewoo JN, Kinney RM, Powell TD, Arguello JJ, Silengo SJ, Partidos CD, Huang CY, Stinchcomb DT, Osorio JE. Immunogenicity and efficacy of chimeric dengue vaccine (DENVax) formulations in interferon-deficient AG129 mice. Vaccine. 2012;30:1513–20. [PMC free article] [PubMed] [Google Scholar]
  • Brunkard JM, Robles Lopez JL, Ramirez J, Cifuentes E, Rothenberg SJ, Hunsperger EA, Moore CG, Brussolo RM, Villarreal NA, Haddad BM. Dengue Fever Seroprevalence and Risk Factors, Texas-Mexico Border, 2004. Emerg Infect Dis. 2007;13:1477–1483. [PMC free article] [PubMed] [Google Scholar]
  • Buckner CM, Luers AJ, Calderon TM, Eugenin EA, Berman JW. Neuroimmunity and the blood-brain barrier: molecular regulation of leukocyte transmigration and viral entry into the nervous system with a focus on neuroAIDS. J Neuroimmune Pharmacol. 2006;1:160–81. [PMC free article] [PubMed] [Google Scholar]
  • Burke T. Dengue haemorrhagic fever: a pathological study. Trans R Soc Trop Med Hyg. 1968;62:682–92. [PubMed] [Google Scholar]
  • Caminade C, Medlock JM, Ducheyne E, McIntyre KM, Leach S, Baylis M, Morse AP. Suitability of European climate for the Asian tiger mosquito Aedes albopictus: recent trends and future scenarios. J R Soc Interface. 2012;9:2708–17. [PMC free article] [PubMed] [Google Scholar]
  • Chambers TJ, Liang Y, Droll DA, Schlesinger JJ, Davidson AD, Wright PJ, Jiang X. Yellow fever virus/dengue-2 virus and yellow fever virus/dengue-4 virus chimeras: biological characterization, immunogenicity, and protection against dengue encephalitis in the mouse model. J Virol. 2003;77:3655–68. [PMC free article] [PubMed] [Google Scholar]
  • Chambers TJ, Nestorowicz A, Mason PW, Rice CM. Yellow fever/Japanese encephalitis chimeric viruses: construction and biological properties. J Virol. 1999;73:3095–101. [PMC free article] [PubMed] [Google Scholar]
  • Chao DL, Halstead SB, Halloran ME, Longini IM., Jr Controlling dengue with vaccines in Thailand. PLoS Negl Trop Dis. 2012;6:e1876. [PMC free article] [PubMed] [Google Scholar]
  • Chappell KJ, Stoermer MJ, Fairlie DP, Young PR. West Nile Virus NS2B/NS3 protease as an antiviral target. Curr Med Chem. 2008;15:2771–84. [PubMed] [Google Scholar]
  • Chase AJ, Medina FA, Munoz-Jordan JL. Impairment of CD4+ T cell polarization by dengue virus-infected dendritic cells. J Infect Dis. 2011;203:1763–74. [PubMed] [Google Scholar]
  • Chaturvedi UC, Agarwal R, Elbishbishi EA, Mustafa AS. Cytokine cascade in dengue hemorrhagic fever: implications for pathogenesis. FEMS Immunol Med Microbiol. 2000;28:183–8. [PubMed] [Google Scholar]
  • Cheeran MC, Hu S, Sheng WS, Rashid A, Peterson PK, Lokensgard JR. Differential responses of human brain cells to West Nile virus infection. J Neurovirol. 2005;11:512–24. [PubMed] [Google Scholar]
  • Chen CJ, Chen JH, Chen SY, Liao SL, Raung SL. Upregulation of RANTES gene expression in neuroglia by Japanese encephalitis virus infection. J Virol. 2004;78:12107–19. [PMC free article] [PubMed] [Google Scholar]
  • Chen SP. Molecular phylogenetic and evolutionary analysis of Japanese encephalitis virus in China. Epidemiol Infect. 2012;140:1637–43. [PubMed] [Google Scholar]
  • Chu JJ, Ng ML. Characterization of a 105-kDa plasma membrane associated glycoprotein that is involved in West Nile virus binding and infection. Virology. 2003;312:458–69. [PubMed] [Google Scholar]
  • Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, Tamminga C, Patterson N, Guerrero M, Bennett JW, McGrath S, Ganeshan H, Belmonte M, Farooq F, Abot E, Banania JG, Huang J, Newcomer R, Rein L, Litilit D, Richie NO, Wood C, Murphy J, Sauerwein R, Hermsen CC, McCoy AJ, Kamau E, Cummings J, Komisar J, Sutamihardja A, Shi M, Epstein JE, Maiolatesi S, Tosh D, Limbach K, Angov E, Bergmann-Leitner E, Bruder JT, Doolan DL, King CR, Carucci D, Dutta S, Soisson L, Diggs C, Hollingdale MR, Ockenhouse CF, Richie TL. DNA Prime/Adenovirus Boost Malaria Vaccine Encoding P. falciparum CSP and AMA1 Induces Sterile Protection Associated with Cell-Mediated Immunity. PLoS One. 2013;8:e55571. [PMC free article] [PubMed] [Google Scholar]
  • Clyde K, Harris E. RNA secondary structure in the coding region of dengue virus type 2 directs translation start codon selection and is required for viral replication. J Virol. 2006;80:2170–82. [PMC free article] [PubMed] [Google Scholar]
  • Clyde K, Kyle JL, Harris E. Recent advances in deciphering viral and host determinants of dengue virus replication and pathogenesis. J Virol. 2006;80:11418–31. [PMC free article] [PubMed] [Google Scholar]
  • Costa SM, Azevedo AS, Paes MV, Sarges FS, Freire MS, Alves AM. DNA vaccines against dengue virus based on the ns1 gene: the influence of different signal sequences on the protein expression and its correlation to the immune response elicited in mice. Virology. 2007;358:413–23. [PubMed] [Google Scholar]
  • Costa SM, Freire MS, Alves AM. DNA vaccine against the non-structural 1 protein (NS1) of dengue 2 virus. Vaccine. 2006a;24:4562–4. [PubMed] [Google Scholar]
  • Costa SM, Paes MV, Barreto DF, Pinhao AT, Barth OM, Queiroz JL, Armoa GR, Freire MS, Alves AM. Protection against dengue type 2 virus induced in mice immunized with a DNA plasmid encoding the non-structural 1 (NS1) gene fused to the tissue plasminogen activator signal sequence. Vaccine. 2006b;24:195–205. [PubMed] [Google Scholar]
  • Courageot MP, Catteau A, Despres P. Mechanisms of dengue virus-induced cell death. Adv Virus Res. 2003;60:157–86. [PubMed] [Google Scholar]
  • Das S, Basu A. Japanese encephalitis virus infects neural progenitor cells and decreases their proliferation. J Neurochem. 2008;106:1624–36. [PubMed] [Google Scholar]
  • Das S, Mishra MK, Ghosh J, Basu A. Japanese Encephalitis Virus infection induces IL-18 and IL-1beta in microglia and astrocytes: correlation with in vitro cytokine responsiveness of glial cells and subsequent neuronal death. J Neuroimmunol. 2008;195:60–72. [PubMed] [Google Scholar]
  • Davis AM, Hagan KA, Matthews LA, Bajwa G, Gill MA, Gale M, Jr., Farrar JD. Blockade of virus infection by human CD4+ T cells via a cytokine relay network. J Immunol. 2008a;180:6923–32. [PMC free article] [PubMed] [Google Scholar]
  • Davis MR, Langan JN, Johnson YJ, Ritchie BW, Van Bonn W. West Nile virus seroconversion in penguins after vaccination with a killed virus vaccine or a DNA vaccine. J Zoo Wildl Med. 2008b;39:582–9. [PubMed] [Google Scholar]
  • De Burghgraeve T, Kaptein SJ, Ayala-Nunez NV, Mondotte JA, Pastorino B, Printsevskaya SS, de Lamballerie X, Jacobs M, Preobrazhenskaya M, Gamarnik AV, Smit JM, Neyts J. An analogue of the antibiotic teicoplanin prevents flavivirus entry in vitro. PLoS One. 7:e37244. [PMC free article] [PubMed] [Google Scholar]
  • Despres P, Frenkiel MP, Ceccaldi PE, Duarte Dos Santos C, Deubel V. Apoptosis in the mouse central nervous system in response to infection with mouse-neurovirulent dengue viruses. J Virol. 1998;72:823–9. [PMC free article] [PubMed] [Google Scholar]
  • Dharakul T, Kurane I, Bhamarapravati N, Yoksan S, Vaughn DW, Hoke CH, Ennis FA. Dengue virus-specific memory T cell responses in human volunteers receiving a live attenuated dengue virus type 2 candidate vaccine. J Infect Dis. 1994;170:27–33. [PubMed] [Google Scholar]
  • Dikeakos JD, Atkins KM, Thomas L, Emert-Sedlak L, Byeon IJ, Jung J, Ahn J, Wortman MD, Kukull B, Saito M, Koizumi H, Williamson DM, Hiyoshi M, Barklis E, Takiguchi M, Suzu S, Gronenborn AM, Smithgall TE, Thomas G. Small molecule inhibition of HIV-1-induced MHC-I down-regulation identifies a temporally regulated switch in Nef action. Mol Biol Cell. 2010;21:3279–92. [PMC free article] [PubMed] [Google Scholar]
  • Diniz JA, Da Rosa AP, Guzman H, Xu F, Xiao SY, Popov VL, Vasconcelos PF, Tesh RB. West Nile virus infection of primary mouse neuronal and neuroglial cells: the role of astrocytes in chronic infection. Am J Trop Med Hyg. 2006;75:691–6. [PubMed] [Google Scholar]
  • Durbin AP, McArthur JH, Marron JA, Blaney JE, Thumar B, Wanionek K, Murphy BR, Whitehead SS. rDEN2/4Delta30(ME), a live attenuated chimeric dengue serotype 2 vaccine is safe and highly immunogenic in healthy dengue-naive adults. Hum Vaccin. 2006;2:255–60. [PubMed] [Google Scholar]
  • Durbin AP, Vargas MJ, Wanionek K, Hammond SN, Gordon A, Rocha C, Balmaseda A, Harris E. Phenotyping of peripheral blood mononuclear cells during acute dengue illness demonstrates infection and increased activation of monocytes in severe cases compared to classic dengue fever. Virology. 2008;376:429–35. [PMC free article] [PubMed] [Google Scholar]
  • Effler PV, Pang L, Kitsutani P, Vorndam V, Nakata M, Ayers T, Elm J, Tom T, Reiter P, Rigau-Perez JG, Hayes JM, Mills K, Napier M, Clark GG, Gubler DJ. Dengue fever, Hawaii, 2001-2002. Emerg Infect Dis. 2005;11:742–9. [PMC free article] [PubMed] [Google Scholar]
  • Egloff MP, Benarroch D, Selisko B, Romette JL, Canard B. An RNA cap (nucleoside-2'-O-)-methyltransferase in the flavivirus RNA polymerase NS5: crystal structure and functional characterization. EMBO J. 2002;21:2757–68. [PMC free article] [PubMed] [Google Scholar]
  • Ehrenfeld E, Modlin J, Chumakov K. Future of polio vaccines. Expert Rev Vaccines. 2009;8:899–905. [PMC free article] [PubMed] [Google Scholar]
  • Eugenin EA, Berman JW. Chemokine-dependent mechanisms of leukocyte trafficking across a model of the blood-brain barrier. Methods. 2003;29:351–61. [PubMed] [Google Scholar]
  • Eugenin EA, Berman JW. Gap junctions mediate human immunodeficiency virus-bystander killing in astrocytes. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2007;27:12844–50. [PMC free article] [PubMed] [Google Scholar]
  • Eugenin EA, Berman JW. Cytochrome C dysregulation induced by HIV infection of astrocytes results in bystander apoptosis of uninfected astrocytes by an IP and Calcium dependent mechanism. J Neurochem. 2013 [PMC free article] [PubMed] [Google Scholar]
  • Eugenin EA, Clements JE, Zink MC, Berman JW. Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2011;31:9456–65. [PMC free article] [PubMed] [Google Scholar]
  • Eugenin EA, Gamss R, Buckner C, Buono D, Klein RS, Schoenbaum EE, Calderon TM, Berman JW. Shedding of PECAM-1 during HIV infection: a potential role for soluble PECAM-1 in the pathogenesis of NeuroAIDS. J Leukoc Biol. 2006a;79:444–52. [PMC free article] [PubMed] [Google Scholar]
  • Eugenin EA, Osiecki K, Lopez L, Goldstein H, Calderon TM, Berman JW. CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigration of human immunodeficiency virus (HIV)-infected leukocytes across the blood-brain barrier: a potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci. 2006b;26:1098–106. [PMC free article] [PubMed] [Google Scholar]
  • Ferraro B, Morrow MP, Hutnick NA, Shin TH, Lucke CE, Weiner DB. Clinical applications of DNA vaccines: current progress. Clin Infect Dis. 2011;53:296–302. [PMC free article] [PubMed] [Google Scholar]
  • Filocamo G, Pacini L, Nardi C, Bartholomew L, Scaturro M, Delmastro P, Tramontano A, De Francesco R, Migliaccio G. Selection of functional variants of the NS3-NS4A protease of hepatitis C virus by using chimeric sindbis viruses. J Virol. 1999;73:561–75. [PMC free article] [PubMed] [Google Scholar]
  • Fink K, Ng C, Nkenfou C, Vasudevan SG, van Rooijen N, Schul W. Depletion of macrophages in mice results in higher dengue virus titers and highlights the role of macrophages for virus control. Eur J Immunol. 2009;39:2809–21. [PubMed] [Google Scholar]
  • Freedman DO, Weld LH, Kozarsky PE, Fisk T, Robins R, von Sonnenburg F, Keystone JS, Pandey P, Cetron MS, GeoSentinel Surveillance N. Spectrum of disease and relation to place of exposure among ill returned travelers. N Engl J Med. 2006;354:119–30. [PubMed] [Google Scholar]
  • Gagnon SJ, Zeng W, Kurane I, Ennis FA. Identification of two epitopes on the dengue 4 virus capsid protein recognized by a serotype-specific and a panel of serotype-cross-reactive human CD4+ cytotoxic T-lymphocyte clones. J Virol. 1996;70:141–7. [PMC free article] [PubMed] [Google Scholar]
  • Geiss BJ, Thompson AA, Andrews AJ, Sons RL, Gari HH, Keenan SM, Peersen OB. Analysis of flavivirus NS5 methyltransferase cap binding. J Mol Biol. 2009;385:1643–54. [PMC free article] [PubMed] [Google Scholar]
  • German AC, Myint KS, Mai NT, Pomeroy I, Phu NH, Tzartos J, Winter P, Collett J, Farrar J, Barrett A, Kipar A, Esiri MM, Solomon T. A preliminary neuropathological study of Japanese encephalitis in humans and a mouse model. Trans R Soc Trop Med Hyg. 2006;100:1135–45. [PubMed] [Google Scholar]
  • Getts DR, Matsumoto I, Muller M, Getts MT, Radford J, Shrestha B, Campbell IL, King NJ. Role of IFN-gamma in an experimental murine model of West Nile virus-induced seizures. J Neurochem. 2007;103:1019–30. [PubMed] [Google Scholar]
  • Getts DR, Terry RL, Getts MT, Muller M, Rana S, Shrestha B, Radford J, Van Rooijen N, Campbell IL, King NJ. Ly6c+ “inflammatory monocytes” are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J Exp Med. 2008;205:2319–37. [PMC free article] [PubMed] [Google Scholar]
  • Ghoshal A, Das S, Ghosh S, Mishra MK, Sharma V, Koli P, Sen E, Basu A. Proinflammatory mediators released by activated microglia induces neuronal death in Japanese encephalitis. Glia. 2007;55:483–96. [PubMed] [Google Scholar]
  • Glass WG, Lim JK, Cholera R, Pletnev AG, Gao JL, Murphy PM. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection. J Exp Med. 2005;202:1087–98. [PMC free article] [PubMed] [Google Scholar]
  • Graham AS, Pruszynski CA, Hribar LJ, DeMay DJ, Tambasco AN, Hartley AE, Fussell EM, Michael SF, Isern S. Mosquito-associated dengue virus, Key West, Florida, USA, 2010. Emerg Infect Dis. 2011;17:2074–5. [PMC free article] [PubMed] [Google Scholar]
  • Gubler DJ. Dengue and dengue hemorrhagic fever. Clin Microbiol Rev. 1998;11:480–96. [PMC free article] [PubMed] [Google Scholar]
  • Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends Microbiol. 2002;10:100–3. [PubMed] [Google Scholar]
  • Guirakhoo F, Weltzin R, Chambers TJ, Zhang ZX, Soike K, Ratterree M, Arroyo J, Georgakopoulos K, Catalan J, Monath TP. Recombinant chimeric yellow fever-dengue type 2 virus is immunogenic and protective in nonhuman primates. J Virol. 2000;74:5477–85. [PMC free article] [PubMed] [Google Scholar]
  • Guirakhoo F, Zhang ZX, Chambers TJ, Delagrave S, Arroyo J, Barrett AD, Monath TP. Immunogenicity, genetic stability, and protective efficacy of a recombinant, chimeric yellow fever-Japanese encephalitis virus (ChimeriVax-JE) as a live, attenuated vaccine candidate against Japanese encephalitis. Virology. 1999;257:363–72. [PubMed] [Google Scholar]
  • Halstead SB. Pathogenesis of dengue: challenges to molecular biology. Science. 1988;239:476–81. [PubMed] [Google Scholar]
  • Halstead SB, Lan NT, Myint TT, Shwe TN, Nisalak A, Kalyanarooj S, Nimmannitya S, Soegijanto S, Vaughn DW, Endy TP. Dengue hemorrhagic fever in infants: research opportunities ignored. Emerg Infect Dis. 2002;8:1474–9. [PMC free article] [PubMed] [Google Scholar]
  • Halstead SB, O'Rourke EJ. Antibody-enhanced dengue virus infection in primate leukocytes. Nature. 1977;265:739–41. [PubMed] [Google Scholar]
  • Halstead SB, Thomas SJ. New Japanese encephalitis vaccines: alternatives to production in mouse brain. Expert Rev Vaccines. 2011;10:355–64. [PubMed] [Google Scholar]
  • Hammon WM, Sather GE. Passive immunity for arbovirus infection. I. Artificially induced prophylaxis in man and mouse for Japanese (B) encephalitis. Am J Trop Med Hyg. 1973;22:524–34. [PubMed] [Google Scholar]
  • Hansen JE. Avoiding climate change. Science. 2006;311:469–70. [PubMed] [Google Scholar]
  • Harris E, Holden KL, Edgil D, Polacek C, Clyde K. Molecular biology of flaviviruses. Novartis Found Symp. 2006;277:23–39. discussion 40, 71-3, 251-3. [PubMed] [Google Scholar]
  • Hase T, Dubois DR, Summers PL, Downs MB, Ussery MA. Comparison of replication rates and pathogenicities between the SA14 parent and SA14-14-2 vaccine strains of Japanese encephalitis virus in mouse brain neurons. Arch Virol. 1993;130:131–43. [PubMed] [Google Scholar]
  • Heinz FX, Stiasny K. Flaviviruses and flavivirus vaccines. Vaccine. 2012;30:4301–6. [PubMed] [Google Scholar]
  • Hennessy S, Liu Z, Tsai TF, Strom BL, Wan CM, Liu HL, Wu TX, Yu HJ, Liu QM, Karabatsos N, Bilker WB, Halstead SB. Effectiveness of live-attenuated Japanese encephalitis vaccine (SA14-14-2): a case-control study. Lancet. 1996;347:1583–6. [PubMed] [Google Scholar]
  • Hirsch AJ, Medigeshi GR, Meyers HL, DeFilippis V, Fruh K, Briese T, Lipkin WI, Nelson JA. The Src family kinase c-Yes is required for maturation of West Nile virus particles. J Virol. 2005;79:11943–51. [PMC free article] [PubMed] [Google Scholar]
  • Hober D, Delannoy AS, Benyoucef S, De Groote D, Wattre P. High levels of sTNFR p75 and TNF alpha in dengue-infected patients. Microbiol Immunol. 1996a;40:569–73. [PubMed] [Google Scholar]
  • Hober D, Poli L, Roblin B, Gestas P, Chungue E, Granic G, Imbert P, Pecarere JL, Vergez-Pascal R, Wattre P, et al. Serum levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and interleukin-1 beta (IL-1 beta) in dengue-infected patients. Am J Trop Med Hyg. 1993;48:324–31. [PubMed] [Google Scholar]
  • Hober D, Shen L, Benyoucef S, De Groote D, Deubel V, Wattre P. Enhanced TNF alpha production by monocytic-like cells exposed to dengue virus antigens. Immunol Lett. 1996b;53:115–20. [PubMed] [Google Scholar]
  • Hong W, Zhang R, Di Z, He Y, Zhao Z, Hu J, Wu Y, Li W, Cao Z. Design of histidine-rich peptides with enhanced bioavailability and inhibitory activity against hepatitis C virus. Biomaterials. 2013;34:3511–22. [PMC free article] [PubMed] [Google Scholar]
  • Hussmann KL, Samuel MA, Kim KS, Diamond MS, Fredericksen BL. Differential Replication of Pathogenic and Nonpathogenic Strains of West Nile Virus within Astrocytes. Journal of virology. 2013;87:2814–22. [PMC free article] [PubMed] [Google Scholar]
  • Imrie A, Zhao Z, Bennett SN, Kitsutani P, Laille M, Effler P. Molecular epidemiology of dengue in the Pacific: introduction of two distinct strains of dengue virus type-1 [corrected] into Hawaii. Ann Trop Med Parasitol. 2006;100:327–36. [PubMed] [Google Scholar]
  • Ishikawa T, Wang G, Widman DG, Infante E, Winkelmann ER, Bourne N, Mason PW. Enhancing the utility of a prM/E-expressing chimeric vaccine for Japanese encephalitis by addition of the JEV NS1 gene. Vaccine. 2011;29:7444–55. [PubMed] [Google Scholar]
  • Issur M, Geiss BJ, Bougie I, Picard-Jean F, Despins S, Mayette J, Hobdey SE, Bisaillon M. The flavivirus NS5 protein is a true RNA guanylyltransferase that catalyzes a two-step reaction to form the RNA cap structure. RNA. 2009;15:2340–50. [PMC free article] [PubMed] [Google Scholar]
  • Jan JT, Chen BH, Ma SH, Liu CI, Tsai HP, Wu HC, Jiang SY, Yang KD, Shaio MF. Potential dengue virus-triggered apoptotic pathway in human neuroblastoma cells: arachidonic acid, superoxide anion, and NF-kappaB are sequentially involved. J Virol. 2000;74:8680–91. [PMC free article] [PubMed] [Google Scholar]
  • Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis. 2004;189:1411–8. [PubMed] [Google Scholar]
  • Johnson BW, Chambers TV, Crabtree MB, Bhatt TR, Guirakhoo F, Monath TP, Miller BR. Growth characteristics of ChimeriVax-DEN2 vaccine virus in Aedes aegypti and Aedes albopictus mosquitoes. Am J Trop Med Hyg. 2002;67:260–5. [PubMed] [Google Scholar]
  • Johnson BW, Chambers TV, Crabtree MB, Guirakhoo F, Monath TP, Miller BR. Analysis of the replication kinetics of the ChimeriVax-DEN 1, 2, 3, 4 tetravalent virus mixture in Aedes aegypti by real-time reverse transcriptase-polymerase chain reaction. Am J Trop Med Hyg. 2004;70:89–97. [PubMed] [Google Scholar]
  • Kawaguchi I, Sasaki A, Boots M. Why are dengue virus serotypes so distantly related? Enhancement and limiting serotype similarity between dengue virus strains. Proc Biol Sci. 2003;270:2241–7. [PMC free article] [PubMed] [Google Scholar]
  • Khanna N, Agnihotri M, Mathur A, Chaturvedi UC. Neutrophil chemotactic factor produced by Japanese encephalitis virus stimulated macrophages. Clin Exp Immunol. 1991;86:299–303. [PMC free article] [PubMed] [Google Scholar]
  • Khor CC, Chau TN, Pang J, Davila S, Long HT, Ong RT, Dunstan SJ, Wills B, Farrar J, Van Tram T, Gan TT, Binh NT, Tri le T, Lien le B, Tuan NM, Tham NT, Lanh MN, Nguyet NM, Hieu NT, Van NVCN, Thuy TT, Tan DE, Sakuntabhai A, Teo YY, Hibberd ML, Simmons CP. Genome-wide association study identifies susceptibility loci for dengue shock syndrome at MICB and PLCE1. Nat Genet. 2011;43:1139–41. [PMC free article] [PubMed] [Google Scholar]
  • King AD, Nisalak A, Kalayanrooj S, Myint KS, Pattanapanyasat K, Nimmannitya S, Innis BL. B cells are the principal circulating mononuclear cells infected by dengue virus. Southeast Asian J Trop Med Public Health. 1999;30:718–28. [PubMed] [Google Scholar]
  • King NJ, Shrestha B, Kesson AM. Immune modulation by flaviviruses. Adv Virus Res. 2003;60:121–55. [PubMed] [Google Scholar]
  • Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that maternal dengue antibodies are important in the development of dengue hemorrhagic fever in infants. Am J Trop Med Hyg. 1988;38:411–9. [PubMed] [Google Scholar]
  • Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke DS. Antibody-dependent enhancement of dengue virus growth in human monocytes as a risk factor for dengue hemorrhagic fever. Am J Trop Med Hyg. 1989;40:444–51. [PubMed] [Google Scholar]
  • Krishnamurti C, Kalayanarooj S, Cutting MA, Peat RA, Rothwell SW, Reid TJ, Green S, Nisalak A, Endy TP, Vaughn DW, Nimmannitya S, Innis BL. Mechanisms of hemorrhage in dengue without circulatory collapse. Am J Trop Med Hyg. 2001;65:840–7. [PubMed] [Google Scholar]
  • Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, Lenches E, Jones CT, Mukhopadhyay S, Chipman PR, Strauss EG, Baker TS, Strauss JH. Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell. 2002;108:717–25. [PMC free article] [PubMed] [Google Scholar]
  • Kulkarni R, Sapkal G, Gore M. Evaluation of Japanese encephalitis virus polytope DNA vaccine candidate in BALB/c mice. Virus Res. 2012;170:118–25. [PubMed] [Google Scholar]
  • Kumar R, Tripathi S, Tambe JJ, Arora V, Srivastava A, Nag VL. Dengue encephalopathy in children in Northern India: clinical features and comparison with non dengue. J Neurol Sci. 2008;269:41–8. [PubMed] [Google Scholar]
  • Kurane I, Brinton MA, Samson AL, Ennis FA. Dengue virus-specific, human CD4+ CD8− cytotoxic T-cell clones: multiple patterns of virus cross-reactivity recognized by NS3-specific T-cell clones. J Virol. 1991a;65:1823–8. [PMC free article] [PubMed] [Google Scholar]
  • Kurane I, Innis BL, Nimmannitya S, Nisalak A, Meager A, Janus J, Ennis FA. Activation of T lymphocytes in dengue virus infections. High levels of soluble interleukin 2 receptor, soluble CD4, soluble CD8, interleukin 2, and interferon-gamma in sera of children with dengue. J Clin Invest. 1991b;88:1473–80. [PMC free article] [PubMed] [Google Scholar]
  • Kurane I, Innis BL, Nisalak A, Hoke C, Nimmannitya S, Meager A, Ennis FA. Human T cell responses to dengue virus antigens. Proliferative responses and interferon gamma production. J Clin Invest. 1989;83:506–13. [PMC free article] [PubMed] [Google Scholar]
  • Laurent-Rolle M, Boer EF, Lubick KJ, Wolfinbarger JB, Carmody AB, Rockx B, Liu W, Ashour J, Shupert WL, Holbrook MR, Barrett AD, Mason PW, Bloom ME, Garcia-Sastre A, Khromykh AA, Best SM. 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. 2010;84:3503–15. [PMC free article] [PubMed] [Google Scholar]
  • Lee CJ, Liao CL, Lin YL. Flavivirus activates phosphatidylinositol 3-kinase signaling to block caspase-dependent apoptotic cell death at the early stage of virus infection. J Virol. 2005;79:8388–99. [PMC free article] [PubMed] [Google Scholar]
  • Lee DW, Choe YJ, Kim JH, Song KM, Cho H, Bae GR, Lee JK. Epidemiology of Japanese encephalitis in South Korea, 2007-2010. Int J Infect Dis. 2012;16:e448–52. [PubMed] [Google Scholar]
  • Lescar J, Luo D, Xu T, Sampath A, Lim SP, Canard B, Vasudevan SG. Towards the design of antiviral inhibitors against flaviviruses: the case for the multifunctional NS3 protein from Dengue virus as a target. Antiviral Res. 2008;80:94–101. [PubMed] [Google Scholar]
  • Leyssen P, De Clercq E, Neyts J. Perspectives for the treatment of infections with Flaviviridae. Clin Microbiol Rev. 2000;13:67–82. table of contents. [PMC free article] [PubMed] [Google Scholar]
  • Liao M, Sanchez-San Martin C, Zheng A, Kielian M. In vitro reconstitution reveals key intermediate states of trimer formation by the dengue virus membrane fusion protein. J Virol. 2010;84:5730–40. [PMC free article] [PubMed] [Google Scholar]
  • Lima DM, de Paula SO, Franca RF, Palma PV, Morais FR, Gomes-Ruiz AC, de Aquino MT, da Fonseca BA. A DNA vaccine candidate encoding the structural prM/E proteins elicits a strong immune response and protects mice against dengue-4 virus infection. Vaccine. 2011;29:831–8. [PubMed] [Google Scholar]
  • Lin RJ, Chang BL, Yu HP, Liao CL, Lin YL. Blocking of interferon-induced Jak-Stat signaling by Japanese encephalitis virus NS5 through a protein tyrosine phosphatase-mediated mechanism. J Virol. 2006;80:5908–18. [PMC free article] [PubMed] [Google Scholar]
  • Lin YL, Huang YL, Ma SH, Yeh CT, Chiou SY, Chen LK, Liao CL. Inhibition of Japanese encephalitis virus infection by nitric oxide: antiviral effect of nitric oxide on RNA virus replication. J Virol. 1997;71:5227–35. [PMC free article] [PubMed] [Google Scholar]
  • Lin YW, Wang KJ, Lei HY, Lin YS, Yeh TM, Liu HS, Liu CC, Chen SH. Virus replication and cytokine production in dengue virus-infected human B lymphocytes. J Virol. 2002;76:12242–9. [PMC free article] [PubMed] [Google Scholar]
  • Lindenbach BD, Pragai BM, Montserret R, Beran RK, Pyle AM, Penin F, Rice CM. The C terminus of hepatitis C virus NS4A encodes an electrostatic switch that regulates NS5A hyperphosphorylation and viral replication. J Virol. 2007;81:8905–18. [PMC free article] [PubMed] [Google Scholar]
  • Lindsey NP, Staples JE, Lehman JA, Fischer M. Surveillance for human West Nile virus disease - United States, 1999-2008. MMWR Surveill Summ. 2010;59:1–17. [PubMed] [Google Scholar]
  • Liu S, Li X, Chen Z, Chen Y, Zhang Q, Liao Y, Zhou J, Ke X, Ma L, Xiao J, Wu Y, Zheng X, Li J, Chen Q. Comparison of genomic and amino acid sequences of eight Japanese encephalitis virus isolates from bats. Arch Virol. 2013 [PMC free article] [PubMed] [Google Scholar]
  • Liu X, Yu Y, Li M, Liang G, Wang H, Jia L, Dong G. Study on the protective efficacy of SA14-14-2 attenuated Japanese encephalitis against different JE virus isolates circulating in China. Vaccine. 2011;29:2127–30. [PubMed] [Google Scholar]
  • Lobigs M, Larena M, Alsharifi M, Lee E, Pavy M. Live chimeric and inactivated Japanese encephalitis virus vaccines differ in their cross-protective values against Murray Valley encephalitis virus. J Virol. 2009;83:2436–45. [PMC free article] [PubMed] [Google Scholar]
  • Lu H, Xu XF, Gao N, Fan DY, Wang J, An J. Preliminary evaluation of DNA vaccine candidates encoding dengue-2 prM/E and NS1: their immunity and protective efficacy in mice. Mol Immunol. 2013;54:109–14. [PubMed] [Google Scholar]
  • Malavige GN, Fernando S, Fernando DJ, Seneviratne SL. Dengue viral infections. Postgrad Med J. 2004;80:588–601. [PMC free article] [PubMed] [Google Scholar]
  • Manns MP, Bourliere M, Benhamou Y, Pol S, Bonacini M, Trepo C, Wright D, Berg T, Calleja JL, White PW, Stern JO, Steinmann G, Yong CL, Kukolj G, Scherer J, Boecher WO. Potency, safety, and pharmacokinetics of the NS3/4A protease inhibitor BI201335 in patients with chronic HCV genotype-1 infection. J Hepatol. 54:1114–22. [PubMed] [Google Scholar]
  • Mathew A, West K, Kalayanarooj S, Gibbons RV, Srikiatkhachorn A, Green S, Libraty D, Jaiswal S, Rothman AL. B-cell responses during primary and secondary dengue virus infections in humans. J Infect Dis. 2011;204:1514–22. [PMC free article] [PubMed] [Google Scholar]
  • Mathur A, Bharadwaj M, Kulshreshtha R, Rawat S, Jain A, Chaturvedi UC. Immunopathological study of spleen during Japanese encephalitis virus infection in mice. Br J Exp Pathol. 1988;69:423–32. [PMC free article] [PubMed] [Google Scholar]
  • Mazzon M, Jones M, Davidson A, Chain B, Jacobs M. Dengue virus NS5 inhibits interferon-alpha signaling by blocking signal transducer and activator of transcription 2 phosphorylation. J Infect Dis. 2009;200:1261–70. [PubMed] [Google Scholar]
  • Medigeshi GR, Lancaster AM, Hirsch AJ, Briese T, Lipkin WI, Defilippis V, Fruh K, Mason PW, Nikolich-Zugich J, Nelson JA. West Nile virus infection activates the unfolded protein response, leading to CHOP induction and apoptosis. J Virol. 2007;81:10849–60. [PMC free article] [PubMed] [Google Scholar]
  • Mentor NA, Kurane I. Dengue virus infection of human T lymphocytes. Acta Virol. 1997;41:175–6. [PubMed] [Google Scholar]
  • Miagostovich MP, dos Santos FB, de Araujo ES, Dias J, Schatzmayr HG, Nogueira RM. Diagnosis of dengue by using reverse transcriptase-polymerase chain reaction. Mem Inst Oswaldo Cruz. 1997a;92:595–9. [PubMed] [Google Scholar]
  • Miagostovich MP, Ramos RG, Nicol AF, Nogueira RM, Cuzzi-Maya T, Oliveira AV, Marchevsky RS, Mesquita RP, Schatzmayr HG. Retrospective study on dengue fatal cases. Clin Neuropathol. 1997b;16:204–8. [PubMed] [Google Scholar]
  • Mishra MK, Ghosh D, Duseja R, Basu A. Antioxidant potential of Minocycline in Japanese Encephalitis Virus infection in murine neuroblastoma cells: correlation with membrane fluidity and cell death. Neurochem Int. 2009;54:464–70. [PubMed] [Google Scholar]
  • Mohammed H, Ramos M, Armstrong J, Munoz-Jordan J, Arnold-Lewis KO, Ayala A, Clark GG, Tull ES, Beatty ME. An outbreak of dengue fever in St. Croix (US Virgin Islands), 2005. PLoS One. 2010a;5:e13729. [PMC free article] [PubMed] [Google Scholar]
  • Mohammed HP, Ramos MM, Rivera A, Johansson M, Munoz-Jordan JL, Sun W, Tomashek KM. Travel-associated dengue infections in the United States, 1996 to 2005. J Travel Med. 2010b;17:8–14. [PubMed] [Google Scholar]
  • Monath TP, Soike K, Levenbook I, Zhang ZX, Arroyo J, Delagrave S, Myers G, Barrett AD, Shope RE, Ratterree M, Chambers TJ, Guirakhoo F. Recombinant, chimaeric live, attenuated vaccine (ChimeriVax) incorporating the envelope genes of Japanese encephalitis (SA14-14-2) virus and the capsid and nonstructural genes of yellow fever (17D) virus is safe, immunogenic and protective in non-human primates. Vaccine. 1999;17:1869–82. [PubMed] [Google Scholar]
  • Mongkolsapaya J, Dejnirattisai W, Xu XN, Vasanawathana S, Tangthawornchaikul N, Chairunsri A, Sawasdivorn S, Duangchinda T, Dong T, Rowland-Jones S, Yenchitsomanus PT, McMichael A, Malasit P, Screaton G. Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nat Med. 2003;9:921–7. [PubMed] [Google Scholar]
  • Mota J, Rico-Hesse R. Dengue virus tropism in humanized mice recapitulates human dengue fever. PLoS One. 2011;6:e20762. [PMC free article] [PubMed] [Google Scholar]
  • Mousson L, Dauga C, Garrigues T, Schaffner F, Vazeille M, Failloux AB. Phylogeography of Aedes (Stegomyia) aegypti (L.) and Aedes (Stegomyia) albopictus (Skuse) (Diptera: Culicidae) based on mitochondrial DNA variations. Genet Res. 2005;86:1–11. [PubMed] [Google Scholar]
  • Munoz-Jordan JL, Sanchez-Burgos GG, Laurent-Rolle M, Garcia-Sastre A. Inhibition of interferon signaling by dengue virus. Proc Natl Acad Sci U S A. 2003;100:14333–8. [PMC free article] [PubMed] [Google Scholar]
  • Nathanson N, Cole GA. Immunosuppression and experimental virus infection of the nervous system. Adv Virus Res. 1970;16:397–448. [PMC free article] [PubMed] [Google Scholar]
  • Nazareth B, Levin J, Johnson H, Begg N. Systemic allergic reactions to Japanese encephalitis vaccines. Vaccine. 1994;12:666. [PubMed] [Google Scholar]
  • Nguyen TH, Nguyen TL, Lei HY, Lin YS, Le BL, Huang KJ, Lin CF, Do QH, Vu TQ, Lam TM, Yeh TM, Huang JH, Liu CC, Halstead SB. Association between sex, nutritional status, severity of dengue hemorrhagic fever, and immune status in infants with dengue hemorrhagic fever. Am J Trop Med Hyg. 2005;72:370–4. [PubMed] [Google Scholar]
  • Noisakran S, Dechtawewat T, Avirutnan P, Kinoshita T, Siripanyaphinyo U, Puttikhunt C, Kasinrerk W, Malasit P, Sittisombut N. Association of dengue virus NS1 protein with lipid rafts. J Gen Virol. 2008;89:2492–500. [PubMed] [Google Scholar]
  • Obeid S, Printsevskaya SS, Olsufyeva EN, Dallmeier K, Durantel D, Zoulim F, Preobrazhenskaya MN, Neyts J, Paeshuyse J. Inhibition of hepatitis C virus replication by semi-synthetic derivatives of glycopeptide antibiotics. J Antimicrob Chemother. 2011;66:1287–94. [PMC free article] [PubMed] [Google Scholar]
  • Ogata A, Nagashima K, Hall WW, Ichikawa M, Kimura-Kuroda J, Yasui K. Japanese encephalitis virus neurotropism is dependent on the degree of neuronal maturity. J Virol. 1991;65:880–6. [PMC free article] [PubMed] [Google Scholar]
  • Oh W, Yang MR, Lee EW, Park KM, Pyo S, Yang JS, Lee HW, Song J. Jab1 mediates cytoplasmic localization and degradation of West Nile virus capsid protein. J Biol Chem. 2006;281:30166–74. [PubMed] [Google Scholar]
  • Ohtaki E, Matsuishi T, Hirano Y, Maekawa K. Acute disseminated encephalomyelitis after treatment with Japanese B encephalitis vaccine (Nakayama-Yoken and Beijing strains). J Neurol Neurosurg Psychiatry. 1995;59:316–7. [PMC free article] [PubMed] [Google Scholar]
  • Okada M, Kita Y, Nakajima T, Hashimoto S, Nakatani H, Nishimatsu S, Nishida Y, Kanamaru N, Kaneda Y, Takamori Y, McMurray D, Tan EV, Cang ML, Saunderson P, Dela Cruz EC. The study of novel DNA vaccines against tuberculosis: Induction of pathogen-specific CTL in the mouse and monkey models of tuberculosis. Hum Vaccin Immunother. 2012;9 [PMC free article] [PubMed] [Google Scholar]
  • Olaleye OD, Omilabu SA, Ilomechina EN, Fagbami AH. A survey for haemagglutination-inhibiting antibody to West Nile virus in human and animal sera in Nigeria. Comp Immunol Microbiol Infect Dis. 1990;13:35–9. [PubMed] [Google Scholar]
  • Paessler S, Walker DH. Pathogenesis of the viral hemorrhagic fevers. Annual review of pathology. 2013;8:411–40. [PubMed] [Google Scholar]
  • Palmer DR, Sun P, Celluzzi C, Bisbing J, Pang S, Sun W, Marovich MA, Burgess T. Differential effects of dengue virus on infected and bystander dendritic cells. J Virol. 2005;79:2432–9. [PMC free article] [PubMed] [Google Scholar]
  • Pang T, Cardosa MJ, Guzman MG. Of cascades and perfect storms: the immunopathogenesis of dengue haemorrhagic fever-dengue shock syndrome (DHF/DSS). Immunol Cell Biol. 2007;85:43–5. [PubMed] [Google Scholar]
  • Paranjape SM, Harris E. Y box-binding protein-1 binds to the dengue virus 3'-untranslated region and mediates antiviral effects. J Biol Chem. 2007;282:30497–508. [PubMed] [Google Scholar]
  • Parquet MC, Kumatori A, Hasebe F, Morita K, Igarashi A. West Nile virus-induced bax-dependent apoptosis. FEBS Lett. 2001;500:17–24. [PubMed] [Google Scholar]
  • Pauvolid-Correa A, Morales MA, Levis S, Figueiredo LT, Couto-Lima D, Campos Z, Nogueira MF, da Silva EE, Nogueira RM, Schatzmayr HG. Neutralising antibodies for West Nile virus in horses from Brazilian Pantanal. Mem Inst Oswaldo Cruz. 2011;106:467–74. [PubMed] [Google Scholar]
  • Peiris JS, Amerasinghe FP, Amerasinghe PH, Ratnayake CB, Karunaratne SH, Tsai TF. Japanese encephalitis in Sri Lanka--the study of an epidemic: vector incrimination, porcine infection and human disease. Trans R Soc Trop Med Hyg. 1992;86:307–13. [PubMed] [Google Scholar]
  • Pekosz A, Phillips J, Pleasure D, Merry D, Gonzalez-Scarano F. Induction of apoptosis by La Crosse virus infection and role of neuronal differentiation and human bcl-2 expression in its prevention. J Virol. 1996;70:5329–35. [PMC free article] [PubMed] [Google Scholar]
  • Petersen LR, Marfin AA. Shifting epidemiology of Flaviviridae. J Travel Med. 2005;12(Suppl 1):S3–11. [PubMed] [Google Scholar]
  • Phung BC, Yeni P. Darunavir: an effective protease inhibitor for HIV-infected patients. Expert Rev Anti Infect Ther. 2011;9:631–43. [PubMed] [Google Scholar]
  • Plesner AM. Allergic reactions to Japanese encephalitis vaccine. Immunol Allergy Clin North Am. 2003;23:665–97. [PubMed] [Google Scholar]
  • Plesner AM, Ronne T. Allergic mucocutaneous reactions to Japanese encephalitis vaccine. Vaccine. 1997;15:1239–43. [PubMed] [Google Scholar]
  • Poland JD, Calisher CH, Monath TP, Downs WG, Murphy K. Persistence of neutralizing antibody 30-35 years after immunization with 17D yellow fever vaccine. Bull World Health Organ. 1981;59:895–900. [PMC free article] [PubMed] [Google Scholar]
  • Priyadarshini D, Gadia RR, Tripathy A, Gurukumar KR, Bhagat A, Patwardhan S, Mokashi N, Vaidya D, Shah PS, Cecilia D. Clinical findings and pro-inflammatory cytokines in dengue patients in Western India: a facility-based study. PLoS One. 5:e8709. [PMC free article] [PubMed] [Google Scholar]
  • Proudfoot AE, Wells TN, Clapham PR. Chemokine receptors--future therapeutic targets for HIV? Biochem Pharmacol. 1999;57:451–63. [PubMed] [Google Scholar]
  • Pugachev KV, Guirakhoo F, Mitchell F, Ocran SW, Parsons M, Johnson BW, Kosoy OL, Lanciotti RS, Roehrig JT, Trent DW, Monath TP. Construction of yellow fever/St. Louis encephalitis chimeric virus and the use of chimeras as a diagnostic tool. Am J Trop Med Hyg. 2004;71:639–45. [PubMed] [Google Scholar]
  • Pugachev KV, Guirakhoo F, Trent DW, Monath TP. Traditional and novel approaches to flavivirus vaccines. Int J Parasitol. 2003;33:567–82. [PubMed] [Google Scholar]
  • Pulmanausahakul R, Roytrakul S, Auewarakul P, Smith DR. Chikungunya in Southeast Asia: understanding the emergence and finding solutions. International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases. 2011;15:e671–6. [PubMed] [Google Scholar]
  • Qing M, Yang F, Zhang B, Zou G, Robida JM, Yuan Z, Tang H, Shi PY. Cyclosporine inhibits flavivirus replication through blocking the interaction between host cyclophilins and viral NS5 protein. Antimicrob Agents Chemother. 2009;53:3226–35. [PMC free article] [PubMed] [Google Scholar]
  • Querec T, Bennouna S, Alkan S, Laouar Y, Gorden K, Flavell R, Akira S, Ahmed R, Pulendran B. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J Exp Med. 2006;203:413–24. [PMC free article] [PubMed] [Google Scholar]
  • Radke EG, Gregory CJ, Kintziger KW, Sauber-Schatz EK, Hunsperger EA, Gallagher GR, Barber JM, Biggerstaff BJ, Stanek DR, Tomashek KM, Blackmore CG. Dengue outbreak in Key West, Florida, USA, 2009. Emerg Infect Dis. 2012;18:135–7. [PMC free article] [PubMed] [Google Scholar]
  • Rahmstorf S, Cazenave A, Church JA, Hansen JE, Keeling RF, Parker DE, Somerville RC. Recent climate observations compared to projections. Science. 2007;316:709. [PubMed] [Google Scholar]
  • Ramirez LA, Arango T, Boyer J. Therapeutic and prophylactic DNA vaccines for HIV-1. Expert Opin Biol Ther. 2013;13:563–73. [PubMed] [Google Scholar]
  • Ramos C, Sanchez G, Pando RH, Baquera J, Hernandez D, Mota J, Ramos J, Flores A, Llausas E. Dengue virus in the brain of a fatal case of hemorrhagic dengue fever. J Neurovirol. 1998;4:465–8. [PubMed] [Google Scholar]
  • Ramos MM, Mohammed H, Zielinski-Gutierrez E, Hayden MH, Lopez JL, Fournier M, Trujillo AR, Burton R, Brunkard JM, Anaya-Lopez L, Banicki AA, Morales PK, Smith B, Munoz-Jordan JL, Waterman SH. Epidemic Dengue and Dengue Hemorrhagic Fever at the Texas-Mexico Border: Results of a Household-based Seroepidemiologic Survey, December 2005. Am J Trop Med Hyg. 2008;78:364–369. [PubMed] [Google Scholar]
  • Raung SL, Chen SY, Liao SL, Chen JH, Chen CJ. Tyrosine kinase inhibitors attenuate Japanese encephalitis virus-induced neurotoxicity. Biochem Biophys Res Commun. 2005;327:399–406. [PubMed] [Google Scholar]
  • Raung SL, Chen SY, Liao SL, Chen JH, Chen CJ. Japanese encephalitis virus infection stimulates Src tyrosine kinase in neuron/glia. Neurosci Lett. 2007;419:263–8. [PubMed] [Google Scholar]
  • Rauscher S, Flamm C, Mandl CW, Heinz FX, Stadler PF. Secondary structure of the 3'-noncoding region of flavivirus genomes: comparative analysis of base pairing probabilities. RNA. 1997;3:779–91. [PMC free article] [PubMed] [Google Scholar]
  • Ravi V, Desai AS, Shenoy PK, Satishchandra P, Chandramuki A, Gourie-Devi M. Persistence of Japanese encephalitis virus in the human nervous system. J Med Virol. 1993;40:326–9. [PubMed] [Google Scholar]
  • Ravi V, Parida S, Desai A, Chandramuki A, Gourie-Devi M, Grau GE. Correlation of tumor necrosis factor levels in the serum and cerebrospinal fluid with clinical outcome in Japanese encephalitis patients. J Med Virol. 1997;51:132–6. [PubMed] [Google Scholar]
  • Reed KD, Meece JK, Henkel JS, Shukla SK. Birds, migration and emerging zoonoses: west nile virus, lyme disease, influenza A and enteropathogens. Clin Med Res. 2003;1:5–12. [PMC free article] [PubMed] [Google Scholar]
  • Restrepo BN, Isaza DM, Salazar CL, Ramirez R, Ospina M, Alvarez LG. Serum levels of interleukin-6, tumor necrosis factor-alpha and interferon-gamma in infants with and without dengue. Rev Soc Bras Med Trop. 2008a;41:6–10. [PubMed] [Google Scholar]
  • Restrepo BN, Ramirez RE, Arboleda M, Alvarez G, Ospina M, Diaz FJ. Serum levels of cytokines in two ethnic groups with dengue virus infection. Am J Trop Med Hyg. 2008b;79:673–7. [PubMed] [Google Scholar]
  • Rigau-Perez JG, Vorndam AV, Clark GG. The dengue and dengue hemorrhagic fever epidemic in Puerto Rico, 1994-1995. Am J Trop Med Hyg. 2001;64:67–74. [PubMed] [Google Scholar]
  • Roberts TK, Eugenin EA, Lopez L, Romero IA, Weksler BB, Couraud PO, Berman JW. CCL2 disrupts the adherens junction: implications for neuroinflammation. Lab Invest. 2012a;92:1213–33. [PMC free article] [PubMed] [Google Scholar]
  • Roberts TK, Eugenin EA, Lopez L, Romero IA, Weksler BB, Couraud PO, Berman JW. CCL2 disrupts the adherens junction: implications for neuroinflammation. Laboratory investigation; a journal of technical methods and pathology. 2012b;92:1213–33. [PMC free article] [PubMed] [Google Scholar]
  • Rodenhuis-Zybert IA, Wilschut J, Smit JM. Dengue virus life cycle: viral and host factors modulating infectivity. Cell Mol Life Sci. 2010;67:2773–86. [PMC free article] [PubMed] [Google Scholar]
  • Roe K, Kumar M, Lum S, Orillo B, Nerurkar VR, Verma S. West Nile virus-induced disruption of the blood-brain barrier in mice is characterized by the degradation of the junctional complex proteins and increase in multiple matrix metalloproteinases. The Journal of general virology. 2012a;93:1193–203. [PMC free article] [PubMed] [Google Scholar]
  • Roe K, Kumar M, Lum S, Orillo B, Nerurkar VR, Verma S. West Nile virus-induced disruption of the blood-brain barrier in mice is characterized by the degradation of the junctional complex proteins and increase in multiple matrix metalloproteinases. J Gen Virol. 2012b;93:1193–203. [PMC free article] [PubMed] [Google Scholar]
  • Rothman AL. T lymphocyte responses to heterologous secondary dengue virus infections. Ann N Y Acad Sci. 2009;1171(Suppl 1):E36–41. [PubMed] [Google Scholar]
  • Sabin AB. Research on dengue during World War II. Am J Trop Med Hyg. 1952;1:30–50. [PubMed] [Google Scholar]
  • Samuel MA, Diamond MS. Pathogenesis of West Nile Virus infection: a balance between virulence, innate and adaptive immunity, and viral evasion. J Virol. 2006;80:9349–60. [PMC free article] [PubMed] [Google Scholar]
  • Sarkari NB, Thacker AK, Barthwal SP, Mishra VK, Prapann S, Srivastava D, Sarkari M. Japanese encephalitis (JE) part II: 14 years' follow-up of survivors. J Neurol. 2012a;259:58–69. [PubMed] [Google Scholar]
  • Sarkari NB, Thacker AK, Barthwal SP, Mishra VK, Prapann S, Srivastava D, Sarkari M. Japanese encephalitis (JE). Part I: clinical profile of 1,282 adult acute cases of four epidemics. J Neurol. 2012b;259:47–57. [PubMed] [Google Scholar]
  • Schioler KL, Samuel M, Wai KL. Vaccines for preventing Japanese encephalitis. Cochrane Database Syst Rev. 2007:CD004263. [PMC free article] [PubMed] [Google Scholar]
  • Schmidt AG, Lee K, Yang PL, Harrison SC. Small-molecule inhibitors of dengue-virus entry. PLoS Pathog. 2012;8:e1002627. [PMC free article] [PubMed] [Google Scholar]
  • Schneeweiss A, Chabierski S, Salomo M, Delaroque N, Al-Robaiy S, Grunwald T, Burki K, Liebert UG, Ulbert S. A DNA vaccine encoding the E protein of West Nile virus is protective and can be boosted by recombinant domain DIII. Vaccine. 2011;29:6352–7. [PubMed] [Google Scholar]
  • Screaton G, Mongkolsapaya J. T cell responses and dengue haemorrhagic fever. Novartis Found Symp. 2006;277:164–71. discussion 171-6, 251-3. [PubMed] [Google Scholar]
  • Sharp TM, Pillai P, Hunsperger E, Santiago GA, Anderson T, Vap T, Collinson J, Buss BF, Safranek TJ, Sotir MJ, Jentes ES, Munoz-Jordan JL, Arguello DF. A cluster of dengue cases in American missionaries returning from Haiti, 2010. Am J Trop Med Hyg. 2012;86:16–22. [PMC free article] [PubMed] [Google Scholar]
  • Simmons CP, Farrar JJ, Nguyen v V, Wills B. Dengue. N Engl J Med. 2012;366:1423–32. [PubMed] [Google Scholar]
  • Singh A, Kulshreshtha R, Mathur A. An enzyme immunoassay for detection of Japanese encephalitis virus-induced chemotactic cytokine. J Biosci. 2000;25:47–55. [PubMed] [Google Scholar]
  • Soares CN, Faria LC, Peralta JM, de Freitas MR, Puccioni-Sohler M. Dengue infection: neurological manifestations and cerebrospinal fluid (CSF) analysis. J Neurol Sci. 2006;249:19–24. [PubMed] [Google Scholar]
  • Solomon T, Dung NM, Vaughn DW, Kneen R, Thao LT, Raengsakulrach B, Loan HT, Day NP, Farrar J, Myint KS, Warrell MJ, James WS, Nisalak A, White NJ. Neurological manifestations of dengue infection. Lancet. 2000;355:1053–9. [PubMed] [Google Scholar]
  • Spindler KR, Hsu TH. Viral disruption of the blood-brain barrier. Trends in microbiology. 2012a;20:282–90. [PMC free article] [PubMed] [Google Scholar]
  • Spindler KR, Hsu TH. Viral disruption of the blood-brain barrier. Trends Microbiol. 2012b;20:282–90. [PMC free article] [PubMed] [Google Scholar]
  • Srikiatkhachorn A, Wichit S, Gibbons RV, Green S, Libraty DH, Endy TP, Ennis FA, Kalayanarooj S, Rothman AL. Dengue viral RNA levels in peripheral blood mononuclear cells are associated with disease severity and preexisting dengue immune status. PLoS One. 2012;7:e51335. [PMC free article] [PubMed] [Google Scholar]
  • Stahla-Beek HJ, April DG, Saeedi BJ, Hannah AM, Keenan SM, Geiss BJ. Identification of a novel antiviral inhibitor of the flavivirus guanylyltransferase enzyme. J Virol. 2012;86:8730–9. [PMC free article] [PubMed] [Google Scholar]
  • Su HL, Liao CL, Lin YL. Japanese encephalitis virus infection initiates endoplasmic reticulum stress and an unfolded protein response. J Virol. 2002;76:4162–71. [PMC free article] [PubMed] [Google Scholar]
  • Sutter RW, Prevots DR, Cochi SL. Poliovirus vaccines. Progress toward global poliomyelitis eradication and changing routine immunization recommendations in the United States. Pediatr Clin North Am. 2000;47:287–308. [PubMed] [Google Scholar]
  • Swarup V, Ghosh J, Das S, Basu A. Tumor necrosis factor receptor-associated death domain mediated neuronal death contributes to the glial activation and subsequent neuroinflammation in Japanese encephalitis. Neurochem Int. 2008;52:1310–21. [PubMed] [Google Scholar]
  • Tomashek KM, Rivera A, Munoz-Jordan JL, Hunsperger E, Santiago L, Padro O, Garcia E, Sun W. Description of a large island-wide outbreak of dengue in Puerto Rico, 2007. Am J Trop Med Hyg. 2009;81:467–74. [PubMed] [Google Scholar]
  • Tung WH, Tsai HW, Lee IT, Hsieh HL, Chen WJ, Chen YL, Yang CM. Japanese encephalitis virus induces matrix metalloproteinase-9 in rat brain astrocytes via NF-kappaB signalling dependent on MAPKs and reactive oxygen species. Br J Pharmacol. 2010;161:1566–83. [PMC free article] [PubMed] [Google Scholar]
  • Umareddy I, Tang KF, Vasudevan SG, Devi S, Hibberd ML, Gu F. Dengue virus regulates type I interferon signalling in a strain-dependent manner in human cell lines. J Gen Virol. 2008;89:3052–62. [PubMed] [Google Scholar]
  • van der Schaar HM, Rust MJ, Chen C, van der Ende-Metselaar H, Wilschut J, Zhuang X, Smit JM. Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells. PLoS Pathog. 2008;4:e1000244. [PMC free article] [PubMed] [Google Scholar]
  • Verma S, Lo Y, Chapagain M, Lum S, Kumar M, Gurjav U, Luo H, Nakatsuka A, Nerurkar VR. West Nile virus infection modulates human brain microvascular endothelial cells tight junction proteins and cell adhesion molecules: Transmigration across the in vitro blood-brain barrier. Virology. 2009;385:425–33. [PMC free article] [PubMed] [Google Scholar]
  • Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. Journal of leukocyte biology. 2012;91:401–15. [PMC free article] [PubMed] [Google Scholar]
  • Wills MR, Sil BK, Cao JX, Yu YX, Barrett AD. Antigenic characterization of the live attenuated Japanese encephalitis vaccine virus SA14-14-2: a comparison with isolates of the virus covering a wide geographic area. Vaccine. 1992;10:861–72. [PubMed] [Google Scholar]
  • Xin YY, Ming ZG, Peng GY, Jian A, Min LH. Safety of a live-attenuated Japanese encephalitis virus vaccine (SA14-14-2) for children. Am J Trop Med Hyg. 1988;39:214–7. [PubMed] [Google Scholar]
  • Yu IM, Zhang W, Holdaway HA, Li L, Kostyuchenko VA, Chipman PR, Kuhn RJ, Rossmann MG, Chen J. Structure of the immature dengue virus at low pH primes proteolytic maturation. Science. 2008;319:1834–7. [PubMed] [Google Scholar]
  • Zanotto PM, Gould EA, Gao GF, Harvey PH, Holmes EC. Population dynamics of flaviviruses revealed by molecular phylogenies. Proc Natl Acad Sci U S A. 1996;93:548–53. [PMC free article] [PubMed] [Google Scholar]
  • Zhang Y, Zhang W, Ogata S, Clements D, Strauss JH, Baker TS, Kuhn RJ, Rossmann MG. Conformational changes of the flavivirus E glycoprotein. Structure. 2004;12:1607–18. [PMC free article] [PubMed] [Google Scholar]
  • Zhou J, Stohlman SA, Hinton DR, Marten NW. Neutrophils promote mononuclear cell infiltration during viral-induced encephalitis. J Immunol. 2003;170:3331–6. [PubMed] [Google Scholar]
  • Zybert IA, van der Ende-Metselaar H, Wilschut J, Smit JM. Functional importance of dengue virus maturation: infectious properties of immature virions. J Gen Virol. 2008;89:3047–51. [PubMed] [Google Scholar]
-