Download University of Groningen Dengue and Chikungunya virus van

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Common cold wikipedia , lookup

Anti-nuclear antibody wikipedia , lookup

Infection control wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Neonatal infection wikipedia , lookup

Childhood immunizations in the United States wikipedia , lookup

Immunomics wikipedia , lookup

Infection wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Innate immune system wikipedia , lookup

Ebola virus disease wikipedia , lookup

Norovirus wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Chickenpox wikipedia , lookup

Orthohantavirus wikipedia , lookup

Molecular mimicry wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Human cytomegalovirus wikipedia , lookup

Marburg virus disease wikipedia , lookup

West Nile fever wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Hepatitis B wikipedia , lookup

Transcript
University of Groningen
Dengue and Chikungunya virus
van Duijl-Richter, Mareike
IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to
cite from it. Please check the document version below.
Document Version
Publisher's PDF, also known as Version of record
Publication date:
2016
Link to publication in University of Groningen/UMCG research database
Citation for published version (APA):
van Duijl-Richter, M. (2016). Dengue and Chikungunya virus: Cell entry mechanisms and the impact of
antibodies on infectivity [Groningen]: University of Groningen
Copyright
Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the
author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).
Take-down policy
If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately
and investigate your claim.
Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the
number of authors shown on this cover page is limited to 10 maximum.
Download date: 17-06-2017
Chapter
2
Dengue Virus Life Cycle
and Pathogenesis
Chapter 2
2
18
DENV Life Cycle and Pathogenesis
Dengue Virus Life Cycle and Pathogenesis
Introduction
Dengue virus (DENV) – mainly transmitted by the Aedes aegypti mosquito
– is the most common mosquito-borne viral infection worldwide. DENV causes an
estimated 390 million infections per year, of which about 100 million are symptomatic
[1]. Each of the four serotypes of DENV (designated DENV 1-4) can cause disease
with symptoms including fever, rash, headache, and musculoskeletal pain [2].
Approximately 500,000 to 1 million individuals however develop more severe disease
including abdominal pain, bleedings, organ impairment, hematological aberrations,
and plasma leakage. Severe dengue can be fatal if not properly treated, and each year
about 25,000 patients die from dengue infection [3,4].
Severe dengue is almost exclusively seen during secondary infection with a
heterotypic DENV serotype and during primary infection of infants with declining
levels of maternal antibodies [5-7]. This observation points towards a pathogenic role
of the immune system in severe disease development. In contrast, upon homotypic
re-infection, the newly infecting DENV is effectively neutralized and individuals do
not develop disease [8]. Also, third and tertiary infections usually do not lead to
disease symptoms [9]. In this chapter, the viral structure and life cycle of DENV
will be described, followed by a review of the host cell tropism and the mechanisms
controlling disease outcome.
2
Viral structure & life cycle
Dengue virus (DENV) belongs to the flavivirus genus within the Flaviviridae
family. Other flaviviruses are for example West Nile virus (WNV), Yellow Fever virus
(YFV), Japanese encephalitis virus (JEV), Tick-borne encephalitis virus (TBEV), and
the emerging Zika virus (ZIKV) [10,11]. DENV virions are small spherical particles
with a diameter of approximately 50 nm. Each virion contains a single-stranded,
positive-sensed RNA genome of 10.8kB in length, which is packaged by the capsid
(C) protein to form the nucleocapsid. The nucleocapsid is surrounded by a hostcell derived lipid bilayer in which 180 copies of the membrane protein (M) and
envelope protein (E) are anchored. The E ectodomain consists of three structural
domains (DI, DII and DIII). The E proteins are arranged as homodimers in a headto-tail orientation. The homodimers are lying tangential to the viral membrane in a
herringbone-like pattern, thereby giving the particle a “smooth” surface (figure 1
a,b) [12-15].
The first step in the viral life cycle (figure 2) involves binding of the E
glycoprotein to a receptor or attachment factor on the host target cell. The putative
19
Chapter 2
2
Figure 1 DENV envelope proteins on the mature virion
The E protein consists of three functional domains (DI, DII & DIII). Receptor-binding is presumably
mediated by DIII (blue). The fusion loop (green) is located at the tip of DII (yellow). DI is depicted in
red. On the mature virions, E homodimers lie flat on the membrane in a head-to-tail orientation, giving the particle a smooth appearance. Adapted from [39] and reprinted with permission from Elsevier
receptor binding domain has been allocated to E DIII [16-19]. Upon binding, DENV
particles enter the host cell via clathrin-mediated endocytosis. The internalized
virions are delivered to Rab5-positive early endosomes, which further mature into
Rab7-positive late endosomes [20,21]. Upon exposure of the virus to the acidic lumen
of the endosome, the E protein homodimers dissociate, which results in the exposure
of the fusion loop situated on DII (figure 1, green). The hydrophobic fusion peptide
subsequently inserts into the endosomal membrane and E homotrimers are formed.
Then, DIII folds back towards the fusion loop, which forces the opposing outer
leaflets of the membranes together. The outer membranes subsequently merge, which
is referred to as hemifusion. This short-lived intermediate is followed by complete
fusion and expansion of the fusion pore. Finally, the nucleocapsid is released to the
cytosol [12,22-24]. It has been suggested that fusion mainly occurs from within
Rab7-positive late endosomes [20], although contrasting reports have been published
considering the location of membrane fusion [25]. In addition to low pH, negatively
charged lipids were shown to support membrane fusion of DENV. This observation
strongly points towards fusion from within late endosomal compartments as
negatively charged lipids are abundantly present within these compartments [26].
Upon the release of the nucleocapsid to the cytosol, the capsid proteins
dissociate from the viral genome. The viral genome is translated at the rough
endoplasmic reticulum (ER) into a single polyprotein and processed into 3 structural
proteins (prM, E and C) and 7 non-structural (NS1, NS2A, NS2B, NS3, NS4A, NS4B,
and NS5) proteins. The ectodomains of prM and E, NS1 and parts of NS4A and NS4B
are facing the ER lumen whereas the other proteins are present at the cytosolic side of
the ER. NS4A, presumably together with other viral and host proteins, subsequently
triggers membrane rearrangements and vesicle formation by ER membrane
20
DENV Life Cycle and Pathogenesis
invagination. These vesicles were found to contain NS proteins, viral RNA, and host
cell factors and serve as sites for viral RNA replication. Newly synthesized viral RNA
is released to the cytosol and associates with C proteins to form the nucleocapsid.
The nucleocapsid subsequently buds into the ER lumen, thereby acquiring a lipid
membrane in which prM and E proteins are anchored. The newly assembled virus
particles have a “spiky” appearance with prM and E heterodimers arranged as 60
trimeric spikes on the viral surface [27-29] (figure 3).
2
The virions are then transported via the Golgi and Trans Golgi Network
(TGN) to the plasma membrane of the cell. During exocytosis the virus particles
further mature. The acidic lumen of the TGN triggers a global conformational
re-arrangement of the viral spike proteins leading to dissociation of the prM-E
heterodimers and formation of E homodimers which are capped by prM. This
Figure 2 DENV life cycle
DENV enters the cell via receptor-mediated endocytosis followed by fusion from within acidic endosomes and release of the nucleocapsid. Viral replication takes place in endoplasmic reticulum (ER)
membrane invaginations. Virus assembly occurs by budding of a newly formed nucleocapsid into the
ER lumen. The newly assembled immature particles are transported through the Golgi and Trans Golgi
Network (TGN) to the surface of the cell. Within the TGN, enzymatic cleavage of the prM protein occurs by the host protease furin. The pr peptide remains associated with the virion till its release to the
extracellular milieu. Reprinted from [39] with permission from Elsevier.
21
Chapter 2
rearrangement allows proteolytic cleavage of prM by the host protease furin, which
separates the pr peptide from the M protein (figure 2). The pr peptide functions
to stabilize the E protein while transiting acid compartments during virus egress. It
stays associated with M at low pH, and is released after secretion of the particle in the
pH-neutral extracellular space [30-33]. DENV-infected cells have been described to
secrete virions with varying maturation degree [34]. Indeed, electron micrographs
revealed the presence of virions with a spiky surface (immature), smooth surface
(mature), and surfaces with both spiky and smooth characteristics in varying
distributions (partially immature). At least 40% of all particles are partially immature
and 3% is fully immature when produced in mosquito cells [35,36]. Comparable
levels of prM were seen in viruses produced in mammalian endothelial and human
embryonic kidney cells, whereas dendritic cells appear to produce more mature virus
[37]. Fully immature DENV virions were found to be essentially non-infectious, as
the pr peptide is capping the E protein thereby preventing virus-cell binding and
membrane fusion [32,38]. It is not known to which degree prM cleavage must have
taken place to enable viral infectivity, yet it is clear that not all prM proteins have to
be cleaved [39].
2
Figure 3 DENV envelope proteins on the immature virion
DENV virions are assembled as immature virions (left side). On these particles, the fusion loop of the
E protein is covered by the prM protein (purple). Color coding of EDI, DII, DIII is same as in figure 1.
Three E-prM heterodimers form one spike on the immature particles, with the prM protein at the tip of
the spike (right side). Adapted from [39] and reprinted with permission from Elsevier.
Primary dengue virus infection
Dengue is transmitted to humans via mosquito bites. The mosquito usually
probes the skin several times before a blood vessel is reached. Most DENV particles
are inoculated in the skin and a minor fraction of particles is directly delivered to the
blood. DENV particles can infect cells resident in the skin, including keratinoctyes,
Langerhans cells and dendritic cells (DCs) [40-43]. Infected Langerhans cells and
DCs migrate to the lymph nodes, where other DENV target cells are infected,
22
DENV Life Cycle and Pathogenesis
including monocytes (which can also be directly infected by virus injected in the
blood vessels) and macrophages [40,44,45]. Moreover, monocytes are recruited to
the dermis were they differentiate into DCs that can be infected as well [46]. The
virus then spreads to other parts of the body and becomes a systemic infection [2].
Although cells derived from the mononuclear lineage are the main target cells of
DENV, also a variety of other cell types is permissive to infection, like for example
cells of hepatic and endothelial origin or fibroblasts [47,48]. Such a wide cell tropism
suggests that DENV uses several receptors or attachment factors. Indeed, molecules
that were described to interact with DENV are (amongst others) heat shock protein
70, heat shock protein 90, GRP78/BiP, a 37/67kDa high-affinity laminin receptor,
heparin sulphate, CD14, and C-type lectin receptors [45,49-58]. C-type lectin
receptors are expressed on cells of the myeloid lineage. For example, Dendritic Cell
Specific Intercellular adhesion molecule-3-Grabbing Non-integrin (DC-SIGN/
CD209) is abundantly expressed on immature dendritic cells (imDC) and on certain
types of macrophages [59]; the mannose receptor is mainly expressed on imDCs
and macrophages, but also on dermal fibroblasts and keratinocytes [60]; and C-type
lectin domain family 5, member A (CLEC5A/MDL-1) is expressed on monocytes
and macrophages [58]. The infected cells produce large amounts of progeny virus,
and approximately three days after the mosquito bite, viremia can be detected in
the blood of infected patients. Peak titers are usually measured three days after the
appearance of fever. Viral load can reach 106-108 viral copies/ml, though in patients
with severe disease, higher titers are observed as well. DENV viremia is cleared on
average 5 days after onset of fever [61,62].
Early in DENV infection the innate immune response is activated, which
is important for restricting the infection and viral clearance [63,64]. The expression
of IFNs and other pro-inflammatory cytokines is triggered by activation of pattern
recognition receptors, including Toll-like receptor (TLR) 3, TLR 7 and TLR 8; but
also the C-type lectins DC-SIGN, CLEC-5, and MR [58,65-68]. Subsequent binding
of IFNs to infected and neighboring cells leads to activation of the JAK/STAT pathway
and induces an antiviral state of the cell. Moreover, activation of natural killer (NK)
cells leads to secretion of type-II IFNγ, which is likewise important in the antiviral
defense [63,69]. DENV can however counteract the antiviral effects of IFNα/β
binding by downregulating the JAK/STAT pathway [70]. Therefore, activation of the
adaptive immune system is essential for complete clearance of infection.
IFN production not only induces an antiviral state, it also stimulates the
adaptive immune system by inducing imDC maturation and activation of lymphocytes
[71]. Approximately 6 days after infection, DENV specific B and T cells appear.
The majority of antibodies against DENV are generated against E and prM, which
suggests that the secreted virus particles indeed consist of a mixture of virions with
varying maturation state [72-74]. Serotype-specific, strongly neutralizing antibodies
are often targeting the E protein and can efficiently prevent infection [75]. NS1
antibodies are produced as the NS1 protein is expressed on the surface of infected
2
23
Chapter 2
cells and secreted in the extracellular space [76,77]. NS1 antibodies were shown
to mediate complement-mediated lysis of DENV infected cells [78,79]. Activated
CD4+ and CD8+ T cells also induce lysis of DENV-infected target cells and produce
a range of antiviral cytokines. Thus, infection is cleared through natural killer cells
and macrophages, assisted by the presence of neutralizing antibodies and activated T
cells [8,80].
2
Re-infection: Neutralization versus enhancement of infection
Upon re-infection with DENV, the adaptive immune response is rapidly
activated and anti-DENV antibodies are secreted. In case of a homologous reinfection, the pre-existing and newly produced antibodies efficiently prevent
infection and together with a rapid activation of memory T-cells, the individual is
protected from disease [81]. If heterotypic re-infection occurs shortly after primary
infection (estimations range from six months up three years), cross-reactive
antibodies are present in high titers thereby protecting the individual from disease
[82,83]. However, if re-infection occurs after a longer time-span, the pre-existing
cross-reactive antibodies are no longer protective against heterologous infection
and may even enhance the severity of disease. Indeed, severe disease development
is strongly associated with a heterologous secondary infection [62,84]. In contrast,
third and tertiary DENV infections only rarely lead to disease as cross-immunity is
then sufficient to prevent infection [9].
Role of antibodies in homologous re-infection
During homologous re-infection, antibodies play an important role in
neutralization. NS1 antibodies have been described to induce complement-dependent
lysis of infected cells [76] and lysis of antibody-opsonized viruses [85]. Antibodies
directed against the viral spike proteins can directly neutralize viral infectivity. The
dogma is that antibody-bound virions are taken up by Fc-receptor expressing cells and
are delivered to the endocytic pathway for degradation. Importantly, Fc-receptors are
expressed on myeloid cells, which are the main natural target cells of DENV. Therefore,
in case of dengue, neutralization has to occur at a post-attachment step [86,87].
Indeed, multiple high-affinity antibodies were identified that neutralized infection
by prevention of membrane fusion [88-90]. Most neutralizing antibodies were found
to target the E protein. In murine studies, antibodies binding to the DIII domain were
most potent, whereas in humans, antibodies targeting quaternary structures and
cross-linking multiple E proteins have been described as strongly neutralizing. These
antibodies likely act by preventing the essential conformational changes required for
membrane fusion [37,91-95]. Neutralization of DENV can be described as a “multiple
hit” phenomenon, in which a certain amount of available epitopes must have bound to
prevent infection. For strongly neutralizing antibodies, only a fraction of epitopes has
24
DENV Life Cycle and Pathogenesis
to be covered to efficiently prevent infection, whereas weakly neutralizing antibodies
have to bind more available sites to mediate neutralization [96]. Thus, in homotypic
re-infection, type-specific antibodies displaying high affinity to the virus are present
in high titers and prevent infection.
Enhanced disease during heterologous secondary infection
2
Approximately 2.5% of the individuals experiencing a secondary heterologous
re-infection develop severe disease [2,3]. It is generally believed that antibodies play
an important role in the development of severe disease as infants with declining
levels of maternal antibodies were found to have an increased risk for severe disease
during primary infection [7,84,97]. During a secondary heterologous re-infection,
pre-existing cross-reactive antibodies can recognize newly infecting virus serotype.
Functional studies revealed that cross-reactive antibodies generally have weakly
neutralizing properties [72,73,98,99]. The presence of weakly-neutralizing crossreactive antibodies is further increased through original antigenic sin, a phenomenon
in which cross-reactive memory B-cells are expanded, but the number of immune
cells against the currently remaining serotype remains low [8,73,98,100].
The cross-reactive antibodies bind to the virions, which are then internalized
by Fc-receptor bearing DENV target cells. Intriguingly, these particles escape from
degradation and fuse from within acidic endosomes [87]. This phenomenon is
called antibody-dependent enhancement of infection (ADE). In vitro, all antibodies
that can neutralize flavivirus infectivity also show enhancement of infection when
antibody concentrations are low enough. Important factors in ADE are: antibody
affinity, epitope accessibility, and antibody concentration. High affinity antibodies
that bind to accessible epitopes are more prone to neutralization than low affinity
antibodies that bind to cryptic sites. ADE occurs when the occupancy of viral particle
by a neutralizing antibody is below the threshold of neutralization [39,74,96,101].
At ADE conditions, antibody-opsonized virions not only escape from
degradation, these particles also have a higher chance to induce membrane fusion
and productive infection [102]. This might be related to the alternative entry route by
which antibody-opsonized DENV is taken up into the cell [103]. In addition, we and
others showed that virtually non-infectious fully immature particles are rendered
infectious by antibodies [104-106]. Upon entry of antibody-opsonized immature
particles in Fc-receptor bearing target cells, furin cleavage and maturation occurs
within the endosome, followed by infection of the cell [105,107]. ADE of infection of
both mature and immature DENV particles leads to an enhanced infected cell mass
and viral load early in infection. Epidemiological studies showed that an increased
viral load is associated with a higher risk of developing severe dengue [61].
Furthermore, not only memory B cells but also memory T cells specific for
the primary infecting serotype are preferentially expanded. Original antigenic sin of
T cells leads to the generation of low avidity T cells that are less efficient in clearing
25
Chapter 2
the infection. This results in the secretion of a vast amount of pro-inflammatory
cytokine and modulators which can cause capillary leakage as seen in severe dengue
[2,8,73,98,108,109]. Though these and other aspects like virulence of the infecting
strain and host genetic factors are clearly associated with severe disease, the complete
picture is not understood given the small percentage of individuals with a heterologous
secondary infection that develop severe disease [2,3].
2
Concluding remarks
DENV has a tremendous clinical impact, yet no antiviral treatment or vaccine
has been approved. The development of an efficient and – most importantly – safe
vaccine is challenging, as it is of upmost importance to prevent vaccine enhanced
disease through ADE and original antigenic sin [110]. Therefore, an optimal vaccine
has to neutralize all four DENV serotypes and elicit a long-term antibody response.
The most promising candidate vaccine so far is from Sanofi Pasteur. Their tetravalent
chimeric DENV/Yellow Fever (YF) vaccine (prM and E proteins of DENV and the
replicative backbone of the YF vaccine 17D) reduced hospitalization by 67%-80%
and dengue hemorrhagic fever by 80%-90% [111,112]. The efficacy to protect against
natural DENV infection was surprisingly low (30%-60%, depending on the study
population) [111-114]. The low protection against disease was rather unexpected,
as antibody titers towards all DENV serotypes were found using the in vitro plaque
reduction neutralization test (PRNT) [114,115].
The “golden standard” PRNT involves cells of endothelial origin [116].
Infection of these cells by DENV can be prevented by neutralizing antibodies.
However, as endothelial cells do not express Fc-receptors, the enhancing properties
of antibodies by Fc-receptor mediated entry cannot be assessed. Therefore,
neutralization in DENV target cells might require higher titers than the ones found
in standard PRNTs. Indeed, several groups have shown that Fc-receptor bearing
target cells often require higher antibody titers for neutralization, especially when
cross-reactive antibodies are tested [117-119]. For this, assays employing Fc-receptor
bearing cells might represent a better predicting neutralization test for DENV, yet
this approach is challenging in an industrial setting. Alternatively, the definition of
the correlate of protection should be re-defined in retro perspective based on the
results of the recent clinical trials, since the current correlate of protection of DENV
is based on early vaccine-related studies on the flaviviruses JEV, YFV, and TBEV; but
does not take the genetic variability between and within the different serotypes of
DENV into account [110].
In summary, dengue is a complex disease and multiple factors are important
in controlling disease outcome. More fundamental research into the virus life cycle
and host immune response is imperative to fully understand disease pathogenesis
and should guide the rational design of antiviral therapies and vaccines.
26
DENV Life Cycle and Pathogenesis
References
1.
Bhatt S, Gething PW, Brady OJ, Messina
JP, Farlow AW, Moyes CL, et al. The global
distribution and burden of dengue. Nature.
2013.
2.
Martina BE, Koraka P, Osterhaus AD.
Dengue virus pathogenesis: an integrated
view. Clin Microbiol Rev. 2009;22: 564-581.
3.
World Health Organization. WHO Fact
Sheet: Dengue and severe dengue. 2015.
Available: http://www.who.int/mediacentre/
factsheets/fs117/en/
4.
Horstick O, Tozan Y, Wilder-Smith A.
Reviewing dengue: still a neglected tropical
disease? PLoS Negl Trop Dis. 2015;9:
e0003632.
5.
Guzman MG, Kouri G, Valdes L, Bravo J,
Vazquez S, Halstead SB. Enhanced severity
of secondary dengue-2 infections: death
rates in 1981 and 1997 Cuban outbreaks. Rev
Panam Salud Publica. 2002;11: 223-227.
6.
7.
Halstead SB. Neutralization and antibodydependent enhancement of dengue viruses.
Adv Virus Res. 2003;60: 421-467.
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: 411-419.
8.
Rothman AL. Immunity to dengue virus:
a tale of original antigenic sin and tropical
cytokine storms. Nat Rev Immunol. 2011.
9.
Gibbons RV, Kalanarooj S, Jarman RG,
Nisalak A, Vaughn DW, Endy TP, et al.
Analysis of repeat hospital admissions for
dengue to estimate the frequency of third
or fourth dengue infections resulting in
admissions and dengue hemorrhagic fever,
and serotype sequences. Am J Trop Med
Hyg. 2007;77: 910-913.
10. Musso D, Cao-Lormeau VM, Gubler DJ.
Zika virus: following the path of dengue and
chikungunya? Lancet. 2015;386: 243-244.
11. Kuno G, Chang GJ, Tsuchiya KR, Karabatsos
N, Cropp CB. Phylogeny of the genus
Flavivirus. J Virol. 1998;72: 73-83.
12. Mukhopadhyay S, Kuhn RJ, Rossmann MG.
A structural perspective of the flavivirus life
cycle. Nat Rev Microbiol. 2005;3: 13-22.
13. Heinz FX, Stiasny K. Flaviviruses and
flavivirus vaccines. Vaccine. 2012;30: 43014306.
14. Zhang Y, Zhang W, Ogata S, Clements D,
Strauss JH, Baker TS, et al. Conformational
changes of the flavivirus E glycoprotein.
Structure. 2004;12: 1607-1618.
2
15. Kuhn RJ, Zhang W, Rossmann MG, Pletnev
SV, Corver J, Lenches E, et al. Structure of
dengue virus: implications for flavivirus
organization, maturation, and fusion. Cell.
2002;108: 717-725.
16. Hung JJ, Hsieh MT, Young MJ, Kao CL, King
CC, Chang W. An external loop region of
domain III of dengue virus type 2 envelope
protein is involved in serotype-specific
binding to mosquito but not mammalian
cells. J Virol. 2004;78: 378-388.
17. Hung SL, Lee PL, Chen HW, Chen LK, Kao
CL, King CC. Analysis of the steps involved
in Dengue virus entry into host cells.
Virology. 1999;257: 156-167.
18. Yu S, Wuu A, Basu R, Holbrook MR, Barrett
AD, Lee JC. Solution structure and structural
dynamics of envelope protein domain III
of mosquito- and tick-borne flaviviruses.
Biochemistry. 2004;43: 9168-9176.
19. Crill WD, Roehrig JT. Monoclonal antibodies
that bind to domain III of dengue virus E
glycoprotein are the most efficient blockers
of virus adsorption to Vero cells. J Virol.
2001;75: 7769-7773.
20. van der Schaar HM, Rust MJ, Chen C, van
der Ende-Metselaar H, Wilschut J, Zhuang
X, et al. Dissecting the cell entry pathway of
dengue virus by single-particle tracking in
living cells. PLoS Pathog. 2008;4: e1000244.
21. Acosta EG, Castilla V, Damonte EB.
Functional entry of dengue virus into Aedes
albopictus mosquito cells is dependent on
clathrin-mediated endocytosis. J Gen Virol.
2008;89: 474-484.
22. Heinz FX, Allison SL. The machinery for
flavivirus fusion with host cell membranes.
Curr Opin Microbiol. 2001;4: 450-455.
23. Kielian M, Rey FA. Virus membrane-fusion
proteins: more than one way to make a
hairpin. Nat Rev Microbiol. 2006;4: 67-76.
24. Modis Y, Ogata S, Clements D, Harrison
27
Chapter 2
SC. Structure of the dengue virus envelope
protein after membrane fusion. Nature.
2004;427: 313-319.
25. Krishnan MN, Sukumaran B, Pal U, Agaisse
H, Murray JL, Hodge TW, et al. Rab 5 is
required for the cellular entry of dengue and
West Nile viruses. J Virol. 2007;81: 48814885.
2
26. Zaitseva E, Yang ST, Melikov K, Pourmal
S, Chernomordik LV. Dengue virus
ensures its fusion in late endosomes using
compartment-specific lipids. PLoS Pathog.
2010;6: e1001131.
27. Welsch S, Miller S, Romero-Brey I, Merz
A, Bleck CK, Walther P, et al. Composition
and three-dimensional architecture of the
dengue virus replication and assembly sites.
Cell Host Microbe. 2009;5: 365-375.
28. Clyde K, Kyle JL, Harris E. Recent advances
in deciphering viral and host determinants of
dengue virus replication and pathogenesis J
Virol. 2006;80: 11418-11431.
29. Apte-Sengupta S, Sirohi D, Kuhn RJ.
Coupling of replication and assembly in
flaviviruses. Curr Opin Virol. 2014;9: 134142.
30. Li L, Lok SM, Yu IM, Zhang Y, Kuhn RJ, Chen
J, et al. The flavivirus precursor membraneenvelope protein complex: structure and
maturation. Science. 2008;319: 1830-1834.
31. Yu IM, Zhang W, Holdaway HA, Li L,
Kostyuchenko VA, Chipman PR, et al.
Structure of the immature dengue virus
at low pH primes proteolytic maturation.
Science. 2008;319: 1834-1837.
32. Yu IM, Holdaway HA, Chipman PR, Kuhn
RJ, Rossmann MG, Chen J. Association of the
pr peptides with dengue virus at acidic pH
blocks membrane fusion. J Virol. 2009;83:
12101-12107.
33. Perera R, Kuhn RJ. Structural proteomics of
dengue virus. Curr Opin Microbiol. 2008;11:
369-377.
34. Junjhon J, Lausumpao M, Supasa S,
Noisakran S, Songjaeng A, Saraithong P, et
al. Differential modulation of prM cleavage,
extracellular particle distribution, and virus
infectivity by conserved residues at nonfurin
consensus positions of the dengue virus pr-M
junction. J Virol. 2008;82: 10776-10791.
35. Plevka P, Battisti AJ, Junjhon J, Winkler
28
DC, Holdaway HA, Keelapang P, et al.
Maturation of flaviviruses starts from one or
more icosahedrally independent nucleation
centres. EMBO Rep. 2011;12: 602-606.
36. Junjhon J, Edwards TJ, Utaipat U, Bowman
VD, Holdaway HA, Zhang W, et al. Influence
of pr-M cleavage on the heterogeneity of
extracellular dengue virus particles J Virol.
2010;84: 8353-8358.
37. Dejnirattisai W, Wongwiwat W, Supasa S,
Zhang X, Dai X, Rouvinski A, et al. A new
class of highly potent, broadly neutralizing
antibodies isolated from viremic patients
infected with dengue virus. Nat Immunol.
2015;16: 170-177.
38. 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: 3047-3051.
39. Pierson TC, Diamond MS. Degrees of
maturity: the complex structure and biology
of flaviviruses. Curr Opin Virol. 2012;2: 168175.
40. 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: 14111418.
41. Limon-Flores AY, Perez-Tapia M, EstradaGarcia I, Vaughan G, Escobar-Gutierrez
A, Calderon-Amador J, et al. Dengue virus
inoculation to human skin explants: an
effective approach to assess in situ the early
infection and the effects on cutaneous
dendritic cells. Int J Exp Pathol. 2005;86:
323-334.
42. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR,
Brachtel E, Putvatana R, et al. Human skin
Langerhans cells are targets of dengue virus
infection. Nat Med. 2000;6: 816-820.
43. Marovich M, Grouard-Vogel G, Louder
M, Eller M, Sun W, Wu SJ, et al. Human
dendritic cells as targets of dengue virus
infection. J Investig Dermatol Symp Proc.
2001;6: 219-224.
44. Durbin AP, Vargas MJ, Wanionek K,
Hammond SN, Gordon A, Rocha C,
et al. Phenotyping of peripheral blood
mononuclear cells during acute dengue
DENV Life Cycle and Pathogenesis
illness demonstrates infection and increased
activation of monocytes in severe cases
compared to classic dengue fever. Virology.
2008;376: 429-435.
45. Miller JL, de Wet BJ, Martinez-Pomares L,
Radcliffe CM, Dwek RA, Rudd PM, et al. The
mannose receptor mediates dengue virus
infection of macrophages. PLoS Pathog.
2008;4: e17.
46. Schmid MA, Harris E. Monocyte recruitment
to the dermis and differentiation to dendritic
cells increases the targets for dengue virus
replication. PLoS Pathog. 2014;10: e1004541.
47. Suksanpaisan L, Cabrera-Hernandez A,
Smith DR. Infection of human primary
hepatocytes with dengue virus serotype 2. J
Med Virol. 2007;79: 300-307.
48. Avirutnan P, Malasit P, Seliger B, Bhakdi
S, Husmann M. Dengue virus infection of
human endothelial cells leads to chemokine
production, complement activation, and
apoptosis. J Immunol. 1998;161: 6338-6346.
49. Reyes-Del Valle J, Chavez-Salinas S, Medina
F, Del Angel RM. Heat shock protein 90 and
heat shock protein 70 are components of
dengue virus receptor complex in human
cells. J Virol. 2005;79: 4557-4567.
50. Chen Y, Maguire T, Hileman RE, Fromm
JR, Esko JD, Linhardt RJ, et al. Dengue virus
infectivity depends on envelope protein
binding to target cell heparan sulfate. Nat
Med. 1997;3: 866-871.
51. Germi R, Crance JM, Garin D, Guimet J,
Lortat-Jacob H, Ruigrok RW, et al. Heparan
sulfate-mediated binding of infectious
dengue virus type 2 and yellow fever virus.
Virology. 2002;292: 162-168.
52. Chen YC, Wang SY, King CC. Bacterial
lipopolysaccharide inhibits dengue virus
infection of primary human monocytes/
macrophages by blockade of virus entry
via a CD14-dependent mechanism. J Virol.
1999;73: 2650-2657.
53. Jindadamrongwech S, Thepparit C, Smith
DR. Identification of GRP 78 (BiP) as a liver
cell expressed receptor element for dengue
virus serotype 2. Arch Virol. 2004;149: 915927.
54. Thepparit C, Smith DR. Serotype-specific
entry of dengue virus into liver cells:
identification of the 37-kilodalton/67-
kilodalton high-affinity laminin receptor as
a dengue virus serotype 1 receptor. J Virol.
2004;78: 12647-12656.
55. Lozach PY, Burleigh L, Staropoli I, NavarroSanchez E, Harriague J, Virelizier JL, et al.
Dendritic cell-specific intercellular adhesion
molecule 3-grabbing non-integrin (DCSIGN)-mediated enhancement of dengue
virus infection is independent of DCSIGN internalization signals. J Biol Chem.
2005;280: 23698-23708.
2
56. Dejnirattisai W, Webb AI, Chan V,
Jumnainsong A, Davidson A, Mongkolsapaya
J, et al. Lectin switching during dengue virus
infection. J Infect Dis. 2011;203: 1775-1783.
57. Navarro-Sanchez E, Altmeyer R, Amara A,
Schwartz O, Fieschi F, Virelizier JL, et al.
Dendritic-cell-specific
ICAM3-grabbing
non-integrin is essential for the productive
infection of human dendritic cells by
mosquito-cell-derived dengue viruses.
EMBO Rep. 2003;4: 723-728.
58. Chen ST, Lin YL, Huang MT, Wu MF, Cheng
SC, Lei HY, et al. CLEC5A is critical for
dengue-virus-induced lethal disease. Nature.
2008;453: 672-676.
59. Geijtenbeek TB, Gringhuis SI. Signalling
through C-type lectin receptors: shaping
immune responses. Nat Rev Immunol.
2009;9: 465-479.
60. Sheikh H, Yarwood H, Ashworth A, Isacke
CM. Endo180, an endocytic recycling
glycoprotein related to the macrophage
mannose receptor is expressed on fibroblasts,
endothelial cells and macrophages and
functions as a lectin receptor. J Cell Sci.
2000;113 ( Pt 6): 1021-1032.
61. Vaughn DW, Green S, Kalayanarooj S, Innis
BL, Nimmannitya S, Suntayakorn S, et al.
Dengue viremia titer, antibody response
pattern, and virus serotype correlate with
disease severity. J Infect Dis. 2000;181: 2-9.
62. Halstead SB (2007) Dengue. The Lancet 370:
1644–1652.
63. Shresta S, Kyle JL, Snider HM, Basavapatna
M, Beatty PR, Harris E. Interferon-dependent
immunity is essential for resistance to
primary dengue virus infection in mice,
whereas T- and B-cell-dependent immunity
are less critical. J Virol. 2004;78: 2701-2710.
64. Diamond MS, Roberts TG, Edgil D, Lu B,
29
Chapter 2
Ernst J, Harris E. Modulation of Dengue
virus infection in human cells by alpha, beta,
and gamma interferons. J Virol. 2000;74:
4957-4966.
65. Fernandez-Garcia MD, Mazzon M, Jacobs
M, Amara A. Pathogenesis of flavivirus
infections: using and abusing the host cell
Cell Host Microbe. 2009;5: 318-328.
2
66. de Kruif MD, Setiati TE, Mairuhu AT, Koraka
P, Aberson HA, Spek CA, et al. Differential
gene expression changes in children with
severe dengue virus infections. PLoS Negl
Trop Dis. 2008;2: e215.
67. Tsai YT, Chang SY, Lee CN, Kao CL.
Human TLR3 recognizes dengue virus and
modulates viral replication in vitro. Cell
Microbiol. 2009;11: 604-615.
68. Akira S. TLR signaling. Curr Top Microbiol
Immunol. 2006;311: 1-16.
69. Azeredo EL, De Oliveira-Pinto LM, Zagne
SM, Cerqueira DI, Nogueira RM, Kubelka
CF. NK cells, displaying early activation,
cytotoxicity and adhesion molecules, are
associated with mild dengue disease. Clin
Exp Immunol. 2006;143: 345-356.
70. Munoz-Jordan JL, Fredericksen BL. How
flaviviruses activate and suppress the
interferon response. Viruses. 2010;2: 676691.
71. Erickson AK, Gale M,Jr. Regulation of
interferon production and innate antiviral
immunity through translational control of
IRF-7. Cell Res. 2008;18: 433-435.
72. Beltramello M, Williams KL, Simmons CP,
Macagno A, Simonelli L, Quyen NT, et al.
The human immune response to Dengue
virus is dominated by highly cross-reactive
antibodies endowed with neutralizing and
enhancing activity. Cell Host Microbe.
2010;8: 271-283.
73. Mathew A, West K, Kalayanarooj S, Gibbons
RV, Srikiatkhachorn A, Green S, et al. B-cell
responses during primary and secondary
dengue virus infections in humans. J Infect
Dis. 2011;204: 1514-1522.
30
2011;3: 2374-2395.
76. Muller DA, Young PR. The flavivirus NS1
protein: molecular and structural biology,
immunology, role in pathogenesis and
application as a diagnostic biomarker.
Antiviral Res. 2013;98: 192-208.
77. Amorim JH, Alves RP, Boscardin SB,
Ferreira LC. The dengue virus non-structural
1 protein: risks and benefits. Virus Res.
2014;181: 53-60.
78. Henchal EA, Henchal LS, Schlesinger
JJ. Synergistic interactions of anti-NS1
monoclonal antibodies protect passively
immunized mice from lethal challenge with
dengue 2 virus. J Gen Virol. 1988;69 ( Pt 8):
2101-2107.
79. Kurosu T, Chaichana P, Yamate M,
Anantapreecha S, Ikuta K. Secreted
complement regulatory protein clusterin
interacts with dengue virus nonstructural
protein 1. Biochem Biophys Res Commun.
2007;362: 1051-1056.
80. St John AL, Rathore AP, Yap H, Ng ML,
Metcalfe DD, Vasudevan SG, et al. Immune
surveillance by mast cells during dengue
infection promotes natural killer (NK) and
NKT-cell recruitment and viral clearance.
Proc Natl Acad Sci U S A. 2011;108: 91909195.
81. Rothman AL, Medin CL, Friberg H, Currier
JR. Immunopathogenesis Versus Protection
in Dengue Virus Infections. Curr Trop Med
Rep. 2014;1: 13-20.
82. Reich NG, Shrestha S, King AA, Rohani P,
Lessler J, Kalayanarooj S, et al. Interactions
between serotypes of dengue highlight
epidemiological impact of cross-immunity. J
R Soc Interface. 2013;10: 20130414.
83. SABIN AB. Research on dengue during
World War II. Am J Trop Med Hyg. 1952;1:
30-50.
84. Halstead SB, O’Rourke EJ. Dengue viruses
and mononuclear phagocytes. I. Infection
enhancement by non-neutralizing antibody.
J Exp Med. 1977;146: 201-217.
74. Rodenhuis-Zybert IA, Wilschut J, Smit JM.
Dengue virus life cycle: viral and host factors
modulating infectivity Cell Mol Life Sci.
2010;67: 2773-2786.
85. Shresta S. Role of complement in dengue
virus infection: protection or pathogenesis?
MBio. 2012;3: 10.1128/mBio.00003-12. Print
2012.
75. Wahala WM, Silva AM. The human antibody
response to dengue virus infection. Viruses.
86. Fischl W, Bartenschlager R. Exploitation
of cellular pathways by Dengue virus. Curr
DENV Life Cycle and Pathogenesis
Opin Microbiol. 2011;14: 470-475.
Nile virus infection. Cell Host Microbe.
2007;1: 135-145.
87. van der Schaar HM, Wilschut JC, Smit JM.
Role of antibodies in controlling dengue
virus infection. Immunobiology. 2009;214:
613-629.
97. Halstead SB. Antibodies determine virulence
in dengue Ann N Y Acad Sci. 2009;1171
Suppl 1: E48-56.
88. Cockburn JJ, Navarro Sanchez ME, Goncalvez
AP, Zaitseva E, Stura EA, Kikuti CM, et al.
Structural insights into the neutralization
mechanism of a higher primate antibody
against dengue virus. EMBO J. 2011.
98. Zompi S, Montoya M, Pohl MO, Balmaseda
A, Harris E. Dominant Cross-Reactive B Cell
Response during Secondary Acute Dengue
Virus Infection in Humans. PLoS Negl Trop
Dis. 2012;6: e1568.
89. Lok SM. Human antibodies stop dengue
virus by jamming its mechanics. Proc Natl
Acad Sci U S A. 2014;111: 1670-1671.
99. de Alwis R, Beltramello M, Messer WB,
Sukupolvi-Petty S, Wahala WM, Kraus A, et
al. In-depth analysis of the antibody response
of individuals exposed to primary dengue
virus infection. PLoS Negl Trop Dis. 2011;5:
e1188.
90. Costin JM, Zaitseva E, Kahle KM, Nicholson
CO, Rowe DK, Graham AS, et al. Mechanistic
study of broadly neutralizing human
monoclonal antibodies against dengue virus
that target the fusion loop. J Virol. 2013;87:
52-66.
91. de Alwis R, Smith SA, Olivarez NP,
Messer WB, Huynh JP, Wahala WM, et
al. Identification of human neutralizing
antibodies that bind to complex epitopes on
dengue virions. Proc Natl Acad Sci U S A.
2012;109: 7439-7444.
92. Crill WD, Chang GJ. Localization and
characterization of flavivirus envelope
glycoprotein cross-reactive epitopes. J Virol.
2004;78: 13975-13986.
93. Smith SA, de Alwis AR, Kose N, Jadi RS, de
Silva AM, Crowe JE,Jr. Isolation of dengue
virus-specific memory B cells with live virus
antigen from human subjects following
natural infection reveals the presence of
diverse novel functional groups of antibody
clones. J Virol. 2014.
94. Fibriansah G, Tan JL, Smith SA, de Alwis
AR, Ng TS, Kostyuchenko VA, et al. A potent
anti-dengue human antibody preferentially
recognizes the conformation of E protein
monomers assembled on the virus surface.
EMBO Mol Med. 2014;6: 358-371.
95. Fibriansah G, Ibarra KD, Ng TS, Smith SA,
Tan JL, Lim XN, et al. DENGUE VIRUS.
Cryo-EM structure of an antibody that
neutralizes dengue virus type 2 by locking E
protein dimers. Science. 2015;349: 88-91.
96. Pierson TC, Xu Q, Nelson S, Oliphant T,
Nybakken GE, Fremont DH, et al. The
stoichiometry
of
antibody-mediated
neutralization and enhancement of West
2
100. Halstead SB, Rojanasuphot S, Sangkawibha
N. Original antigenic sin in dengue. Am J
Trop Med Hyg. 1983;32: 154-156.
101. Dowd KA, Jost CA, Durbin AP, Whitehead
SS, Pierson TC. A dynamic landscape for
antibody binding modulates antibodymediated neutralization of West Nile virus.
PLoS Pathog. 2011;7: e1002111.
102. Flipse J, Diosa-Toro MA, Hoornweg TE,
Ayala-Núñez NV, van de Pol DPI, UrcuquiInchima S, et al. Antibody-dependent
enhancement of dengue virus infection in
primary human macrophages; balancing
higher fusion against antiviral responses.
In: Molecular Mechanisms of Dengue
Virus Infection - Cell Tropism, AntibodyDependent Enhancement, and Cytokines.
2015.
103. Ayala-Nunez NV, Hoornwega TE, van de
Pol DPI, Sjollema KA, van der Schaar H,
Smit JM. Seeing is believing: how antibodies
alter the cell entry pathway of dengue virus
particles in living macrophages. In: Dengue
virus cell entry - unraveling the role of
antibodies, maturation status, and antiviral
drugs. 2014.
104. Dejnirattisai W, Jumnainsong A, Onsirisakul
N, Fitton P, Vasanawathana S, Limpitikul
W, et al. Cross-reacting antibodies enhance
dengue virus infection in humans. Science.
2010;328: 745-748.
105. Rodenhuis-Zybert IA, van der Schaar
HM, da Silva Voorham JM, van der EndeMetselaar H, Lei HY, Wilschut J, et al.
Immature dengue virus: a veiled pathogen?
31
Chapter 2
PLoS Pathog. 2010;6: e1000718.
106. da Silva Voorham JM, Rodenhuis-Zybert
IA, Ayala Nunez NV, Colpitts TM, van der
Ende-Metselaar H, Fikrig E, et al. Antibodies
against the envelope glycoprotein promote
infectivity of immature dengue virus
serotype 2. PLoS One. 2012;7: e29957.
2
107. Zheng A, Umashankar M, Kielian M. In
vitro and in vivo studies identify important
features of dengue virus pr-E protein
interactions. PLoS Pathog. 2010;6: e1001157.
108. Friberg H, Bashyam H, Toyosaki-Maeda T,
Potts JA, Greenough T, Kalayanarooj S, et al.
Cross-reactivity and expansion of denguespecific T cells during acute primary and
secondary infections in humans. Sci Rep.
2011;1: 51.
109. Cardier JE, Marino E, Romano E, Taylor P,
Liprandi F, Bosch N, et al. Proinflammatory
factors present in sera from patients with
acute dengue infection induce activation
and apoptosis of human microvascular
endothelial cells: possible role of TNFalpha in endothelial cell damage in dengue.
Cytokine. 2005;30: 359-365.
110. Flipse J, Smit JM. The Complexity of a
Dengue Vaccine: A Review of the Human
Antibody Response. PLoS Negl Trop Dis.
2015;9: e0003749.
111. Villar L, Dayan GH, Arredondo-Garcia
JL, Rivera DM, Cunha R, Deseda C, et al.
Efficacy of a tetravalent dengue vaccine in
children in Latin America. N Engl J Med.
2015;372: 113-123.
112. Villar LA, Rivera-Medina DM, ArredondoGarcia JL, Boaz M, Starr-Spires L, Thakur
M, et al. Safety and immunogenicity of a
recombinant tetravalent dengue vaccine in
9-16 year olds: a randomized, controlled,
32
phase II trial in Latin America. Pediatr Infect
Dis J. 2013;32: 1102-1109.
113. Capeding MR, Tran NH, Hadinegoro SR,
Ismail HI, Chotpitayasunondh T, Chua
MN, et al. Clinical efficacy and safety of a
novel tetravalent dengue vaccine in healthy
children in Asia: a phase 3, randomised,
observer-masked, placebo-controlled trial.
Lancet. 2014;384: 1358-1365.
114. Sabchareon A, Wallace D, Sirivichayakul
C, Limkittikul K, Chanthavanich P,
Suvannadabba S, et al. Protective efficacy
of the recombinant, live-attenuated,
CYD tetravalent dengue vaccine in Thai
schoolchildren: a randomised, controlled
phase 2b trial. Lancet. 2012.
115. Wilder-Smith A. Dengue vaccines: dawning
at last? Lancet. 2014;384: 1327-1329.
116. Roehrig JT, Hombach J, Barrett AD.
Guidelines
for
Plaque-Reduction
Neutralization Testing of Human Antibodies
to Dengue Viruses. Viral Immunol. 2008;21:
123-132.
117. Wu RS, Chan KR, Tan HC, Chow A, Allen
JC,Jr, Ooi EE. Neutralization of dengue virus
in the presence of Fc receptor-mediated
phagocytosis distinguishes serotype-specific
from cross-neutralizing antibodies. Antiviral
Res. 2012;96: 340-343.
118. Moi ML, Lim CK, Chua KB, Takasaki T,
Kurane I. Dengue virus infection-enhancing
activity in serum samples with neutralizing
activity as determined by using FcgammaRexpressing cells. PLoS Negl Trop Dis. 2012;6:
e1536.
119. Chawla T, Chan KR, Zhang SL, Tan HC,
Lim AP, Hanson BJ, et al. Dengue virus
neutralization in cells expressing Fc gamma
receptors. PLoS One. 2013;8: e65231.
DENV Life Cycle and Pathogenesis
2
33