You are on page 1of 7

PERSPECTIVES

DENV molecular pathogenesis that have


V E C TO R - B O R N E D I S E A S E S — O P I N I O N
been particularly difficult to understand
using the current experimental methodolo-
Barriers to preclinical investigations gies. We also discuss the paradox that fur-
ther substantial advances in the field might
of anti-dengue immunity and dengue be possible only through controlled animal
experimentation, which has inherent limita-
pathogenesis tions because animal models do not pre-
cisely replicate human disease. We propose
that this contradiction can be reconciled
Ashley L. St. John, Soman N. Abraham and Duane J. Gubler with the choice of appropriate animal mod-
els to answer specific experimental questions
Abstract | Dengue virus (DENV) is a human pathogen that causes severe and and with cautious interpretation of results,
potentially fatal disease in millions of individuals each year. Immune-mediated keeping in mind the multifaceted complexity
pathology is thought to underlie many of the complications of DENV infection in of the interactions between DENV and the
humans, but the notable limitations of the available animal models have impeded human host.
our knowledge of the interactions between DENV and the immune system. In this Dengue virus infection in humans
Opinion article, we discuss some of the controversies in the field of dengue DENV is a member of the family Flaviviridae
research relating to the interaction between DENV and the mammalian host. We (TABLE 1), which contains several other
highlight key barriers hindering our understanding of the molecular pathogenesis notable human pathogens. There are four
of DENV and offer suggestions for the most effective ways in which the role of the antigenically distinct DENV serotypes
(DENV1–DENV4), which have a distribu-
immune system in the protection from, and pathology of, DENV infection can be
tion throughout tropical regions of the world
addressed experimentally. similar to that of their principal mosquito
vector, Aedes aegypti 2. The key pathophysio­
Dengue fever has emerged as a major global analyse the mechanisms underlying DENV- logical host response to DENV infection
public-health problem over the past four induced pathologies. Early key studies that manifests in the form of increased vascular
decades. Closely tied to unprecedented elucidated the course of infection, the inci- permeability, plasma leakage, microvascular
human population growth, urbanization, dence of pathological complications and the bleeding and reduced functioning of the
globalization and the lack of effective vector characteristic symptoms were carried out in coagulation cascade. Current estimates sug-
control, both the causative viruses and the experimentally infected human subjects4,5. gest that between 1% and 70% of individuals
mosquito vectors that transmit them have Today, there are limitations on human who experience infection have mild haem-
spread globally in the tropics, resulting in experimentation because of ethical and reg- orrhagic manifestations such as petechiae,
an increased frequency and magnitude of ulatory concerns, and because of the expense purpura, ecchymoses and epistaxis (nose-
epidemics and the emergence of the severe of undertaking large studies in genetically bleed)6,7 (FIG. 1). Severe dengue, known as
form of disease, dengue haemorrhagic fever diverse populations. Furthermore, human DHF and dengue shock syndrome (DSS),
(DHF)1,2. In 2013, an estimated 3.6 billion studies using patient populations (as occurs in <1% of infections2 and can be life-
people, more than half of the world’s popula- opposed to experimental infections) may threatening. During DHF–DSS, bleeding
tion, live in areas that are at risk for dengue show a correlation, but do not show une- involves multiple organs, frequently includ-
virus (DENV) infection3. There are neither quivocal causation, as these studies lack the ing the gastrointestinal tract, and fluid may
vaccines nor specific antiviral therapies ability to measure the responses of patients pool within body cavities (FIG. 1); lesions in
available for this disease. in comparison to exact controls. For dengue, the blood vessels or signs of necrosis have not
The research that has defined our additional variables could include unidenti- been observed, even in fatal cases8. DSS, the
understanding of the pathogenesis and fied factors that might influence the host’s most severe form of DHF, is characterized by
immunology of DENV infection can be susceptibility to disease in addition to influ- a rapid, weak pulse and sudden drop in blood
predominantly segregated into three types encing the disease course. Although animal pressure, which is the result of collapse of
of studies: those examining patients with models have supplemented our knowledge, the vascular system owing to hypovolaemia
dengue, to describe the clinical presentation; unfortunately a single animal model has not caused by vascular leakage.
in vitro studies to define the viral life cycle been able to faithfully reproduce all aspects It is not fully understood why most indi-
or investigate direct interactions between of human DENV infection. viduals resolve DENV infections quickly
DENV and cultured cells; and controlled In this Opinion article, we highlight the and without complications, whereas others
hypothesis-driven animal experiments to key aspects of anti-dengue immunity and experience a potentially fatal vascular leak

420 | JUNE 2013 | VOLUME 11 www.nature.com/reviews/micro

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Table 1 | Important flavivirus human pathogens particularly when these cells are activated
by pro-inflammatory factors derived from
Flavivirus species Prominent pathologies during human infection neighbouring immune cells23. Although
Dengue virus (DENV1–DENV4) Fever–arthralgia–rash syndrome, plasma leakage, haemorrhage most immune cells release pro-inflammatory
and severe disease associated with organ failure mediators when they are infected with
All four serotypes can cause a spectrum of illness ranging from DENV, some, such as mast cells16, can
asymptomatic to severe and fatal disease, as determined by directly detect and respond to the virus with-
a complex interaction of the virus with the innate immune
response of the individual host; the strain of virus, the
out being infected. This emphasizes the fact
previous infection history and age of the host, and host that there are likely to be additional mecha-
genetics all influence disease severity. nisms of DENV structural recognition by the
Yellow fever virus Fever, nausea, abdominal pain, jaundice and haemorrhage immune system beyond antibody detection.
For cells that are infected, pro-inflammatory
West Nile virus Fever, myalgia, encephalitis and long-term neurological defects
responses to DENV replication are initiated
Japanese encephalitis virus Fever, encephalitis and long-term neurological defects by intracellular pathogen recognition recep-
Tick-borne encephalitis virus Meningitis, encephalitis, mild fever and long-term neurological tors, including vesicle-associated Toll-like
defects receptor 3 (TLR3) and the cytosolic sensors
retinoic acid-inducible gene I protein (RIG‑I)
and melanoma differentiation-associated
syndrome or even severe frank haemorrhag- human skin explants15, a detailed analysis of protein 5 (MDA5; also known as IFIH1)24,25.
ing. Risk factors for severe disease include the DC and monocyte subsets that sustain These pathways lead to the production
the presence of heterologous antibodies infection within the skin has not been car- of pro-inflammatory cytokines, includ-
from a previous infection9, certain strains ried out. Most efforts to define the human ing tumour necrosis factor (TNF), type‑I
or subtypes of infecting virus10,11, and the cell types infected by DENV have sought interferons (IFNs), IFNγ and interleukin‑6
age and genetic background of the human to identify viral replication at a much later (IL‑6)24,25.
host 2,12–14. stage of infection: in the blood during peak
viraemia. In monkeys and mice, antigen Systemic dengue virus infection. As DENV
Key questions for dengue virus immunity has been detected in cells that are morpho­ disseminates in a host, it is detected first in
Dengue remains a disease with many logically consistent with mononuclear draining lymph nodes and then in remote
unanswered questions. There are multiple phagocytes19,20 (which are thought to be one lymph nodes19. In humans and non-human
levels of DENV–host interaction from the of the prime targets of DENV infection), primates, this results in viraemia (which
time virus particles are injected by mos­ but this is not well documented in human can be detectable 24–48 hours before the
quitoes, to the establishment of systemic infection. Phenotypic differences potentially onset of clinical symptoms4), and DENV
infection, to its resolution. Each stage exist among the target cells for infection in can be isolated from the blood during the
requires specialized considerations for the skin (early infection), in the lymphoid acute phase of disease. At this stage, sub-
effective experimental design to investigate organs (during viral dissemination) and in stantial changes occur to the cellularity of
the mechanisms underlying disease. various tissues when systemic infection has the haematopoietic system, as would be
been established. expected during systemic viral infection;
The course of host–virus interactions these changes include leukopenia, neutro-
Initial target cells for infection. Little is Clearance of dengue virus from the injec- penia, thrombocytopenia and, occasionally,
understood regarding the early events fol- tion site. Early in DENV infection, immune eosinophilia26 (FIG. 2). DENV infection is also
lowing DENV inoculation into the skin, cells are targets of infection, but also pro- characterized by altered bone marrow cellu-
including immune responses at the site of mote DENV clearance from the skin. Much larity, but the mechanisms or consequences
inoculation and the impact of the infecting emphasis is placed on the role of DENV- of these changes are unknown26.
viral strain or subtype, both of which could specific neutralizing antibodies in viral clear- Vascular pathology, including microvas-
potentially determine the host’s success in ance21. However, during primary infection, cular permeability of endothelial cells, occurs
viral clearance. The role of the infecting viral substantial levels of neutralizing antibodies as viraemia declines27. Although infected
strain is discussed below. Subcutaneously should not be present until the later stages endothelial cells have been observed in human
injected virus can potentially encounter of disease, as the adaptive immune response infection28, it is uncertain how frequently this
several cells of the immune system, includ- requires time to develop. This suggests that happens, because of the paucity of autopsies
ing tissue-resident dendritic cells (DCs), in the early days of infection additional, on human patients with dengue. Human
macrophages and mast cells15,16 (FIG. 2). The antibody-independent mechanisms of viral endothelial cells can be infected in cell culture,
cell types that are first infected in the skin clearance from the injection site are required. and infection of liver sinusoidal cells occurs
have not been extensively studied, but it It is possible that, as occurs in West Nile in immunocompromised animals29. Although
is thought that macrophages and DCs are virus infection, γδ T cells play a part in viral controversial, it has been suggested that
initial targets, and receptors have been iden- clearance from the skin22. In mice, natural endothelial cells are not commonly infected
tified for DENV uptake by both cell types: killer (NK) cells and NK T cells (recruitment during human infection with DENV30,31 and
the mannose receptor on macrophages and of which is promoted by activated skin- that the breakdown in the endothelial cell
DC‑SIGN (DC-specific ICAM‑3‑grabbing resident mast cells) have been implicated in barrier is consistent with other mechanisms
non-integrin 1) on DCs17,18 (FIG. 2). Aside viral clearance at the site of skin injection of vascular permeability26. Owing to both
from the demonstration that Langerhans and in draining lymph nodes16. Stromal and the lack of evidence that endothelial cells are
cells are permissive to DENV infection in endothelial cells can also produce cytokines, directly targeted by DENV and the massive

NATURE REVIEWS | MICROBIOLOGY VOLUME 11 | JUNE 2013 | 421

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

not yet explained. Unfortunately, the com-


Headache, fever plexity of the interactions between DENV
and the host cannot be adequately modelled
Bleeding gums,
nose and eyes by in vitro manipulation of human cells and
Vascular symptoms:
Hypovolaemia
biological products outside the organs and
Low blood pressure systems that govern their responses to infec-
Vascular symptoms:
Leukopenia Shock tion. Not only are the specific target cells of
Thrombocytopenia DENV not clearly known, but also in vitro
Neutropenia Hepatic injury analysis does not capture the responses
Late eosinophilia Fluid pooling in
Reduced coagulation of cells that do not become infected but
body cavities
Gall bladder might nonetheless respond to DENV and
thickening to the inflammatory products in their
Skin symptoms: Haemorrhaging
Rash microenviron­ment in vivo. As a further
within organs
Bruising complication, interactions between DENV
Petechiae and host cells can change over the course of
Purpura Vomiting infection, which begins as a localized cuta-
Intestinal bleeding
neous infection with innate immune activa-
tion and develops into a systemic infection
characterized by viral replication in target
organs (including the lymphoid system) and
high viral titres in the blood19. The most
Infrequent
complications: severe pathologies associated with DENV
Joint pain Encephalitis infection frequently manifest in patients
Acute pancreatitis as viraemia resolves and fever begins to
Renal failure
Myocarditis subside27, but it is unclear at what stage of
Altered haematopoiesis
Splenic rupture infection these pathological processes are
Pulmonary haemorrhage initiated.

The role of immunological memory


Primary DENV infection results in long-
lasting immunity to the infecting serotype
and, potentially, partial immunity to subse-
Figure 1 | Dengue virus pathogenesis in humans.  Systemic infection with dengue virus (DENV)
Nature Reviews | Microbiology quent infection with other serotypes. Many
affects multiple organ systems. This diagram depicts the clinical symptoms and pathogenesis of
dengue in humans, across the spectrum of mild to severe disease. Clinical manifestations associated individuals in dengue-endemic countries
with dengue fever are listed in blue boxes, those associated with dengue haemorrhagic fever are listed have experienced DENV infection previ-
in purple boxes, and more rare complications of DENV infection are listed in the green box. ously. Re‑infection with the same serotype
as that which caused the primary infection
has not been documented, demonstrating
pro-inflammatory response that is detectable versus DHF–DSS in humans. Some evidence that immunity to a homologous DENV
in the serum of patients with dengue32, it has has been obtained for there being both lower strain can be highly protective and lifelong 5.
been postulated that immune factors act as expression of TNF receptors on granulo- Pre-existing neutralizing antibodies must
intermediaries in the response of endothe- cytes during DHF–DSS35 and an association have a major role in preventing subsequent
lial cells to DENV. In particular, cytokine between DHF–DSS and certain TNF poly- infection with the same DENV serotype.
storm (elevated levels of many cytokines in morphisms12. In one study, serum TNF Secondary infection with a heterologous
the serum) has been identified as a poten- was elevated during both DHF–DSS and serotype can cause a broad spectrum of
tial underlying mechanism of vascular dengue fever compared with levels in healthy illnesses, ranging from asymptomatic infec-
pathology21. controls, but a significant difference in TNF tion to severe haemorrhagic disease. For
During DENV infection, copious amounts levels between DHF–DSS and dengue fever example, some epidemiological studies have
of certain pro-inflammatory and vaso­active groups was not detected36. Thus, it is possible shown that patients with a secondary DENV
cytokines, including TNF and vascular that TNF has a role in DENV infection, but infection and babies born to mothers who
endothelial growth factor A (VEGFA), are whether it is (or can be, in some instances) have previously been infected with DENV
produced32. However, although these fac- a significant cause of DHF–DSS in humans are more likely to develop severe disease37,38.
tors are frequently elevated in the serum is still unclear. Multiple pro-inflammatory One theory for why this occurs was first
of patients with dengue, some studies have factors might act in concert to promote described in reference to other flaviviruses
raised questions about their direct role in vasculopathy. (Murray Valley encephalitis virus, West Nile
dengue pathology. For example, in spite of virus and Japanese encephalitis viruses)39
the well-established role of TNF in promot- Drawbacks of in vitro analysis. Ultimately, and is termed antibody-dependent enhance-
ing vascular leakage in other diseases and the mechanisms of many of the immune ment of infection (ADE). ADE involves
experimental contexts33,34, there has been no changes that occur in response to DENV the binding of immune complexes of non-
clear correlation shown between levels of infection, and the roles of these immune neutralizing antibody and infectious virus
TNF and the manifestation of dengue fever changes in protection versus pathology, are to the Fc receptors of immune cells29,40,41

422 | JUNE 2013 | VOLUME 11 www.nature.com/reviews/micro

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

and has been shown to enhance DENV


infectivity in vitro in human cells42. Studies
of DENV in vitro and in primates infused
with enhancing antibody have shown that
ADE can enhance infection of monocytes
by promoting viral uptake and intracellular
replication40,43. However, it should be noted Epidermis
that this enhancement occurs in only a small
fraction of secondary infections and with Mannose
only certain strains of virus2,44. DENV DC-SIGN
receptor
Another immunological concept that
Viral RNA
was first identified in the context of another
virus — in this case, influenza virus45 — and Infected DC
later shown to be relevant to DENV infec- Cellular
Infected cytotoxicity
tion46,47 is original antigenic sin. This theory Activated macrophage
describes how amplification of pathogen- mast cell
specific lymphocytes during secondary T cell
heterologous challenge can constitute a Dermis
misallocation of resources for the immune
system if those lymphocytes are not as effec- NK cell
Lymphatic
tive in promoting clearance of the second- vessel Trafficking
ary challenge strain as they were for the to lymph node Cellular
primary strain. It has been reported that recruitment
T cell responses in children experiencing a
secondary DENV infection have a low strain
specificity, resulting in T cell anergy and
Blood
apoptosis, and possibly higher viral titres vessel
owing to a lack of optimal viral clearance
by T cells46. In other experimental models Figure 2 | Host responses to cutaneous dengue virus injection.  Most infections by dengue virus
resulting in original antigenic sin, T cell Nature Reviews | Microbiology
(DENV) occur after subcutaneous injection of the virus into the skin. Released viral particles may infect
responses are usually highly specific for the nearby cells (thought to be predominantly monocytes or dendritic cells (DCs)) or activate resident
primary challenge strain, and this might also immune cells such as mast cells. A local inflammatory response to DENV in the skin prompts the recruit-
be the case for DENV48. ment of leukocytes from the vasculature, including natural killer (NK) cells and T cells, which promote
ADE and original antigenic sin have both the killing of virus-infected cells at the site of injection. DENV is thought to then travel to draining
been offered as explanations for the asso- lymph nodes via lymphatic vessels to establish systemic infection. These localized inflammatory
ciation of secondary DENV infection with responses occur many days before there are any signs of severe infection.
an increased incidence of DHF–DSS21,46,
as both theories provide an understanding
of how immunological memory could be not be a requirement for experimental whether unidentified factors released along
detrimental to the host when a secondary design to understand dengue pathogenesis. a similar time course could be more con-
challenge is highly similar to, but antigeni- Understanding the differential processes that sequential to pathology. Recent data show
cally distinct from, the primary infecting lead to dengue pathogenesis during primary that mast cell proteases such as chymase
virus. Each theory provides a potential versus secondary infection is an important are released early during acute infection,
mechanism for achieving high viral titres aim for the field. promoting DENV-induced vascular leakage
in vivo. Although viraemia levels have been in mice, and serum levels of chymase have
associated with disease severity 49,50, people The role of immune pathology been correlated with the severity of disease
with secondary infections frequently do Some of the emphasis on the role of anti- in human patients20. This is one example
not have higher levels of viraemia or severe body in dengue pathogenesis comes from of an immunomodulatory product that is
disease. Also, contrary to the widely held the experimental demonstration that anti- released along with cytokines early during
view, ADE (as a specific mechanism to body alone or post-immune serum is suf- acute DENV infection. Another group of
explain secondary infection as a risk factor ficient to enhance the pathology of DENV vasoactive lipid products, the leukotrienes,
for DHF–DSS) has not been demonstrated infection in animal models29,53. Similar also seem to enhance DENV-induced vascu-
to occur in vivo in humans, although it can studies demonstrating causation are still lar leakage in the immunocompetent mouse
be achieved with human products in an needed for host responses that, on the basis model20. Blockade of TNF can reduce vascu-
ex vivo experimental setting 51. DHF–DSS of human studies, are associated with vascu- lar pathology in immunocompromised ani-
is often diagnosed in patients with primary lar leak syndrome, such as cytokine storm. mals during terminal disease54, but its role
infection, and capillary leakage has been Factors that might be correlated with dengue in the development of DHF–DSS in humans
reported to more frequently accompany pri- severity include TNF and VEGF, but animal remains unclear 36. Cytokine storm can also
mary infection than secondary infection in studies have not yet clearly established the occur as a result of infections of entirely
certain studies52. Secondary infection is thus role of these host factors in disease progres- different aetiology without achieving the
not a requirement for DHF–DSS and should sion, the cellular source of these factors, or highly specific symptoms that are attributed

NATURE REVIEWS | MICROBIOLOGY VOLUME 11 | JUNE 2013 | 423

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

to DENV infection, suggesting that more spectrum of clinical outcomes for patients viral and immune outcomes might also be
information is required if we are to fully infected with DENV, from asymptomatic useful. Examples that are currently being
understand the mechanisms underlying infection to classical dengue fever and, occa- applied in some studies include quantita-
DENV-induced vascular pathology. sionally, lethal complications of haemor- tive real-time reverse transcription PCR
rhage and/or shock. These complex factors (qRT-PCR), which can be used as a sensi-
Virus-intrinsic or host-associated risk factors and seemingly contradictory observations tive substitute for plaque assays (although
Virus-intrinsic factors are likely to influ- will probably be reconciled only with con- this has the caveat that qRT-PCR does not
ence the ability of certain strains to replicate trolled experimental investigation, which accurately measure infectious DENV),
robustly or induce severe pathology. For would require preclinical studies using ani- and flow cytometry, which can be used in
example, mutations in the DENV polymerase mal models and a range of unadapted viral place of immunohistochemistry to quantify
non-structural protein 3 (NS3) and envelope strains. rare infected populations. Therefore, it is
protein enhance viral fitness and promote worthwhile revisiting lesser-used models to
neurovirulence, potentially by altering viral Animal models of dengue virus infection determine whether additional information
binding to cell surface receptors or increasing Many animal models have been used in about dengue pathogenesis and the host
the efficiency of NS3 (REF. 55). There are many DENV research, including multiple strains immune response can be garnered using
examples of interactions between DENV of immunocompetent and immunocom- modern techniques and a broader range of
proteins and immune products (for example, promised mice, New World and Old World low-passage viral strains.
NS1 binds to complement component primates, and even humanized animals65. In response to the difficulty of assess-
C4 (REF. 56), and NS5 binds to signalling inter- In the pursuit of an ideal model, however, ing DENV virology in immunocompe-
mediary STAT2 (signal transducer and activa- self-imposed boundaries have emerged tent animals, and in an effort to achieve
tor or transcription 2) to antagonize the IFN with regards to acceptable in vivo experi- uniform severity of infection and clearly
pathway 57). Phylogenetic analyses have shown mentation using the tools that are currently interpretable results, most recent animal
that the emergence of a new DENV variant available. Non-human primates have been studies have gravitated towards the use of
can correlate with an increased likelihood underused, in part because results have been immunocompromised models (such as the
of DHF–DSS in humans11. Certain changes highly variable, depending on the primate AG129 IFN-deficient mouse model) and
within DENV structural proteins could alter species and infecting viral strain used. In ‘antibody-enhanced’ infection (often using
antibody binding to viral epitopes58, but some studies, primates have shown mild monoclonal antibodies to DENV, along with
some regions that have shown variation do symptoms of DENV infection or vascular mouse-adapted DENV). These systems have
not correspond to surface epitopes of mature leakage66–68. Primates have also been used to clear advantages for answering certain ques-
DENV virions11. Evolution of non-structural test the safety, immunogenicity and protec- tions. For example, investigations with the
genes has also been associated with increased tive capacity of vaccine candidates before aim of identifying antiviral drugs might be
epidemic potential59 and reduced disease these candidates are tested in human clinical most definitive in immunocompromised
severity 10, supporting the existence of a trials69. This stage of preclinical testing in mice, in which almost unrestricted DENV
DENV-intrinsic component to virulence primates is virtually inevitable for dengue replication can occur and there is uniform
that requires further evaluation. vaccine candidates and itself warrants fur- disease severity 73. However, we must be
Conversely, some host genetic factors ther study of the pathophysiological changes careful not to overextend the conclusions
are either associated with or protective caused by DENV in the primate model. from such experiments. In many aspects,
against the development of DHF–DSS. Immunocompetent animals (including particularly relating to host immunity and
In addition to TNF12, certain isoforms of primates, but especially wild-type mice) potentially also to immune-mediated path­
major histocompatibility complex (MHC) are considered a poor choice for DENV ology, there might be differences between
class I molecules, MHC class II molecules, research, even in studies addressing immune the mouse immune response and the
immuno­globulin G Fc region receptor II function directly, owing to their relative response that develops during human infec-
(FcγRII) molecules, cytotoxic T lymphocyte resistance to infection compared with tion. Moreover, we do not know whether the
protein 4 (CTLA4) and transforming growth humans70,71. It should be noted, however, that underlying mechanisms of key pathological
factor β1 (TGFβ1) have all been linked to each model has distinct benefits and limita- events such as vascular leakage would be
DHF–DSS13,60–63, whereas in some cases, tions. In addition, many of the accepted the same in mice as during human disease
particular isoforms seem to be protective61. limitations of certain models (such as the or whether the same cell types that become
Certain polymorphisms in TAP1 and TAP2, expectation that immunocompetent mice infected in humans are infected in immuno-
genes associated with antigen presentation would not support infections with replicat- compromised animals. The absence of key
and peptide loading on MHC, also have posi- ing viruses) are not adequately represented inflammatory pathways could allow other-
tive or negative correlations with DHF–DSS64. in the literature with data, but rather, are wise resistant mouse cells to be permissive
These studies have identified target host discussed anecdotally 70,71. This makes assess- for DENV replication. In our opinion, the
factors that might influence dengue patho- ing other potential causes of model ‘failure’, animal models that have lent themselves to
genesis; however, as many of these factors such as the choice of viral strain, difficult. studying the basic virology of DENV, such
have been identified in an individual popu- Some of these concepts regarding the resist- as immunocompromised mice, might not be
lation or outbreak, they require corrobora- ance of certain species to infection date the most appropriate for studying immunity
tion in independent studies and subsequent back to the earliest in vivo studies carried or immune pathogenesis. We do not advo-
experimental validation. out using DENV72, and scarce supporting cate the adoption of any one existing model
Virus–host interactions are not the data have been published in the intervening as the gold standard because they each have
same for all infected patients or strains of time. As new techniques have now become their pros, cons and caveats. Rather, as out-
virus. This fact is underscored by the broad available, more sensitive means of measuring lined below, researchers should select the

424 | JUNE 2013 | VOLUME 11 www.nature.com/reviews/micro

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

most appropriate existing model, with these models for dengue might have limitations, Ashley L. St. John and Soman N. Abraham
are also at the Department of Pathology,
considerations in mind, to address a unique but should not be dismissed as inherently
Duke University Medical Center.
experimental question. flawed. It should be noted that severe dengue
Soman N. Abraham is also at the Department of
Investigations that aim to understand does not manifest uniformly in humans, so
Immunology and the Department of Molecular Genetics
the development of effective anti-dengue lack of severity in a model should not pre- and Microbiology, Duke University Medical Center,
immunity and to use that understanding as vent the study of dengue in varied models. Durham, North Carolina 27710, USA.
a basis for rational vaccine design require an Conversely, as the limitations of animal Correspondence to D.J.G. 
intact immune system to fully encompass models for dengue are well known, the e‑mail: duane.gubler@duke-nus.edu.sg
the immune response. Immunocompetent results of these studies should be used to doi:10.1038/nrmicro3030
mice are thought to be relatively resistant inform our collective knowledge of DENV Published online 8 May 2013
to DENV compared with humans, but this virology, pathogenesis and immunity in 1. Gubler, D. J. Dengue, urbanization and globalization:
the unholy trinity of the 21st century. Trop. Med. Health
fact also affords us the opportunity to study order to introduce new theories and concepts 39, 3–11 (2011).
effective anti-dengue immunity in a host to the field, but not to limit the potential of 2. Gubler, D. J. Dengue and dengue hemorrhagic fever.
Clin. Microbiol. Rev. 11, 480–496 (1998).
that can respond to, and clear, the infection. similar investigations in humans. For exam- 3. Wilder-Smith, A. et al. DengueTools: innovative tools
Using selective knockout mice could provide ple, there seem to be differences in the ability and strategies for the surveillance and control of
dengue. Glob. Health Action 5, 17273 (2012).
a mechanistic understanding of how unique of humans and mice to develop neutralizing 4. Siler, J. F., Hall, M. W. & Hitchens, A. P. Results
cell types or inflammatory factors contrib- antibodies to certain epitopes of DENV obtained in the transmission of dengue fever. JAMA
84, 1163–1172 (1925).
ute to immunity and pathogenesis. There proteins77. For immunocompromised mice, 5. Sabin, A. B. Research on dengue during World War II.
is likely to be a complex interplay between drugs might reduce dengue pathology but Am. J. Trop. Med. Hyg. 1, 30–50 (1952).
6. Aggarwal, A., Chandra, J., Aneja, S., Patwari, A. K. &
the processes of anti-dengue immunity and never allow long-term survival of animals Dutta, A. K. An epidemic of dengue hemorrhagic fever
DENV-induced pathology, as supported by that cannot clear the infection. Thus, limita- and dengue shock syndrome in children in Delhi.
Indian Pediatr. 35, 727–732 (1998).
the association of secondary infection with tions in the degree to which we can extend 7. Vaughn, D. W. et al. Dengue viremia titer, antibody
variable disease severity during dengue out- conclusions from animal studies should not response pattern, and virus serotype correlate
with disease severity. J. Infect. Dis. 181, 2–9 (2000).
breaks27,74–76. Studying immune pathology inhibit our pursuit of understanding this 8. Bhamarapravati, N., Tuchinda, P. & Boonyapaknavik, V.
mechanistically has proved difficult because pathogen in a range of animal models as well Pathology of Thailand haemorrhagic fever: a study of
100 autopsy cases. Ann. Trop. Med. Parasitol. 61,
mice with functional immune systems rarely as in reasoned human trials. 500–510 (1967).
fully recapitulate the severity of human 9. Zompi, S., Montoya, M., Pohl, M. O., Balmaseda, A.
& Harris, E. Dominant cross-reactive B cell response
DHF. However, we challenge the premise Conclusions during secondary acute dengue virus infection in
that full recapitulation of the disease time The field of dengue research has made great humans. PLoS Negl. Trop. Dis. 6, e1568 (2012).
10. Steel, A., Gubler, D. J. & Bennett, S. N. Natural
course or pathological phenotype is a neces- progress over the past 50 years, particularly attenuation of dengue virus type‑2 after a series of
sity. Changes that occur in vivo in response in terms of defining dengue clinically, deter- island outbreaks: a retrospective phylogenetic study of
events in the South Pacific three decades ago. Virology
to DENV infection need not be equally mining how to identify and treat dengue 405, 505–512 (2010).
detrimental to the survival of animals and with supportive care, and understanding the 11. Messer, W. B., Gubler, D. J., Harris, E., Sivananthan, K.
& de Silva, A. M. Emergence and global spread of a
humans to reveal information regarding the epidemiology and basic biology of DENV, dengue serotype 3, subtype III virus. Emerg. Infect.
progression of disease, and such information including viral structure and replication. Dis. 9, 800–809 (2003).
12. Vejbaesya, S. et al. TNF and LTA gene, allele, and
could be crucial for identifying a therapeu- Animal models have been developed for extended HLA haplotype associations with severe
tic or prophylactic strategy. Using animal antiviral drug testing, and many groups dengue virus infection in ethnic Thais. J. Infect. Dis.
199, 1442–1448 (2009).
models that have fallen out of favour for have designed promising vaccine candidates. 13. Chen, R. F. et al. Combination of CTLA‑4 and TGFβ1
dengue research, such as wild-type immuno- Importantly, many of the remaining ques- gene polymorphisms associated with dengue
hemorrhagic fever and virus load in a dengue‑2
competent mice and non-human primates, tions about DENV as a pathogen centre on outbreak. Clin. Immunol. 131, 404–409 (2009).
to study those physiological changes will how the immune system, both innate and 14. de la, C. S. B., Kouri, G. & Guzman, M. G. Race: a risk
factor for dengue hemorrhagic fever. Arch. Virol. 152,
require careful experimental design, a real- acquired, perceives and responds to DENV 533–542 (2007).
istic assessment of the likelihood that con- infection, how genetic variation in both the 15. Wu, S. J. et al. Human skin Langerhans cells are targets
of dengue virus infection. Nature Med. 6, 816–820
clusions can be extended to human disease, host and the virus influence this perception, (2000).
the use of selected unpassaged viral strains and where the fine line lies for protection 16. St John, A. L. et al. Immune surveillance by mast
cells during dengue infection promotes natural killer
and subtypes, and the use of more sensitive versus pathology. These questions cannot (NK) and NKT-cell recruitment and viral clearance.
measures of pathology than the extreme be definitively answered in the context of Proc. Natl Acad. Sci. USA 108, 9190–9195 (2011).
17. Tassaneetrithep, B. et al. DC-SIGN (CD209) mediates
outcomes of haemorrhaging, prolonged an immunocompromised system alone, dengue virus infection of human dendritic cells. J. Exp.
viraemia or death. Some potential surrogates but must be directly addressed with bold Med. 197, 823–829 (2003).
18. Miller, J. L. et al. The mannose receptor mediates
for end-stage pathology include more sensi- experimental design of preclinical studies, dengue virus infection of macrophages. PLoS Pathog.
tive methods to objectively measure vascular including in immunocompetent animals, 4, e17 (2008).
19. Marchette, N. J. et al. Studies on the pathogenesis of
permeability or immune cell signalling, sta- to fully define anti-dengue immunity. We dengue infection in monkeys. 3. Sequential
tistically significant changes in the number believe that meeting this challenge will be distribution of virus in primary and heterologous
infections. J. Infect. Dis. 128, 23–30 (1973).
of haematopoietic cells (for example, loss of a prerequisite to identifying the most effec- 20. St John, A. L., Rathore, A. P., Raghavan, B., Ng, M. L.
platelets or proliferation of lymphocytes), tive targeted therapies and vaccine strategies & Abraham, S. N. Contributions of mast cells and
vasoactive products, leukotrienes and chymase, to
the levels of immune factors upstream of against dengue. dengue virus-induced vascular leakage. eLife (in the
vascular leakage (such as heightened levels press).
Ashley L. St. John, Soman N. Abraham and Duane 21. Rothman, A. L. Immunity to dengue virus: a tale of
of cytokines or other vasoactive factors), original antigenic sin and tropical cytokine storms.
J. Gubler are at the Program in Emerging Infectious
and alternate time points of assessment to Diseases, Graduate Medical School, Duke-National
Nature Rev. Immunol. 11, 532–543 (2011).
22. Wang, T. et al. IFN-γ-producing γδ T cells help control
those thought to be analogous to the human University of Singapore, 8 College Road, murine West Nile virus infection. J. Immunol. 171,
course of infection. Thus, individual animal 169857, Singapore. 2524–2531 (2003).

NATURE REVIEWS | MICROBIOLOGY VOLUME 11 | JUNE 2013 | 425

© 2013 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

23. Starnes, T. et al. Cutting edge: IL‑17F, a novel cytokine complex receptors. Infect. Immun. 32, 469–473 63. Fernandez-Mestre, M. T., Gendzekhadze, K., Rivas-
selectively expressed in activated T cells and (1981). Vetencourt, P. & Layrisse, Z. TNF-α‑308A allele, a
monocytes, regulates angiogenesis and endothelial cell 42. Halstead, S. B., Chow, J. S. & Marchette, N. J. possible severity risk factor of hemorrhagic
cytokine production. J. Immunol. 167, 4137–4140 Immunological enhancement of dengue virus manifestation in dengue fever patients. Tissue
(2001). replication. Nature New Biol. 243, 24–26 (1973). Antigens 64, 469–472 (2004).
24. Tsai, Y. T., Chang, S. Y., Lee, C. N. & Kao, C. L. Human 43. Halstead, S. B. & O’Rourke, E. J. Antibody-enhanced 64. Soundravally, R. & Hoti, S. L. Polymorphisms of the
TLR3 recognizes dengue virus and modulates viral dengue virus infection in primate leukocytes. Nature TAP 1 and 2 gene may influence clinical outcome of
replication in vitro. Cell. Microbiol. 11, 604–615 265, 739–741 (1977). primary dengue viral infection. Scand. J. Immunol. 67,
(2009). 44. Halstead, S. B., Venkateshan, C. N., Gentry, M. K. & 618–625 (2008).
25. Loo, Y. M. et al. Distinct RIG‑I and MDA5 signaling by Larsen, L. K. Heterogeneity of infection enhancement 65. Cassetti, M. C. et al. Report of an NIAID workshop on
RNA viruses in innate immunity. J. Virol. 82, 335–345 of dengue 2 strains by monoclonal antibodies. dengue animal models. Vaccine 28, 4229–4234
(2008). J. Immunol. 132, 1529–1532 (1984). (2010).
26. Halstead, S. B. Antibody, macrophages, dengue virus 45. Webster, R. G. Original antigenic sin in ferrets: the 66. Onlamoon, N. et al. Dengue virus-induced
infection, shock, and hemorrhage: a pathogenetic response to sequential infections with influenza hemorrhage in a nonhuman primate model. Blood
cascade. Rev. Infect. Dis. 11 (Suppl. 4), S830–S839 viruses. J. Immunol. 97, 177–183 (1966). 115, 1823–1834 (2010).
(1989). 46. Mongkolsapaya, J. et al. Original antigenic sin and 67. Halstead, S. B., Shotwell, H. & Casals, J. Studies on
27. Halstead, S. B. Dengue. Lancet 370, 1644–1652 apoptosis in the pathogenesis of dengue hemorrhagic the pathogenesis of dengue infection in monkeys. I.
(2007). fever. Nature Med. 9, 921–927 (2003). Clinical laboratory responses to primary infection.
28. Gubler, D. J. & Zaki, S. in Pathology of Emerging 47. Halstead, S. B., Rojanasuphot, S. & Sangkawibha, N. J. Infect. Dis. 128, 7–14 (1973).
Infections 2 Ch. 3 (eds Nelson, A. M. & Original antigenic sin in dengue. Am. J. Trop. Med. 68. Omatsu, T. et al. Common marmoset (Callithrix
Horsburgh, C. R.) 43–71 (American Society for Hyg. 32, 154–156 (1983). jacchus) as a primate model of dengue virus infection:
Microbiology Press, 1998). 48. Singh, R. A., Rodgers, J. R. & Barry, M. A. The role of development of high levels of viraemia and
29. Zellweger, R. M., Prestwood, T. R. & Shresta, S. T cell antagonism and original antigenic sin in genetic demonstration of protective immunity. J. Gen. Virol.
Enhanced infection of liver sinusoidal endothelial cells immunization. J. Immunol. 169, 6779–6786 (2002). 92, 2272–2280 (2011).
in a mouse model of antibody-induced severe dengue 49. Gubler, D. J., Reed, D., Rosen, L. & Hitchcock, J. R. Jr. 69. Guirakhoo, F. et al. A single amino acid substitution in
disease. Cell Host Microbe 7, 128–139 (2010). Epidemiologic, clinical, and virologic observations on the envelope protein of chimeric yellow fever-dengue 1
30. Jessie, K., Fong, M. Y., Devi, S., Lam, S. K. & dengue in the Kingdom of Tonga. Am. J. Trop. Med. vaccine virus reduces neurovirulence for suckling mice
Wong, K. T. Localization of dengue virus in naturally Hyg. 27, 581–589 (1978). and viremia/viscerotropism for monkeys. J. Virol. 78,
infected human tissues, by immunohistochemistry and 50. Vaughn, D. W. et al. Dengue in the early febrile phase: 9998–10008 (2004).
in situ hybridization. J. Infect. Dis. 189, 1411–1418 viremia and antibody responses. J. Infect. Dis. 176, 70. Bente, D. A. & Rico-Hesse, R. Models of dengue virus
(2004). 322–330 (1997). infection. Drug Discov. Today Dis. Models 3, 97–103
31. Halstead, S. B. Controversies in dengue pathogenesis. 51. Kliks, S. C., Nisalak, A., Brandt, W. E., Wahl, L. & (2006).
Paediatr. Int. Child Health 32 (Suppl. 1), 5–9 (2012). Burke, D. S. Antibody-dependent enhancement of 71. Yauch, L. E. & Shresta, S. Mouse models of dengue
32. Hober, D. et al. Serum levels of tumor necrosis factor- dengue virus growth in human monocytes as a risk virus infection and disease. Antiviral Res. 80, 87–93
alpha (TNF-α), interleukin‑6 (IL‑6), and interleukin‑1β factor for dengue hemorrhagic fever. Am. J. Trop. Med. (2008).
(IL‑1β) in dengue-infected patients. Am. J. Trop. Med. Hyg. 40, 444–451 (1989). 72. Simmons, J. S., St. John, J. H., & Reynolds, H. K.
Hyg. 48, 324–331 (1993). 52. Meltzer, E., Heyman, Z., Bin, H. & Schwartz, E. Experimental Studies of Dengue (Bureau of Printing,
33. Edwards, M. J., Heniford, B. T. & Miller, F. N. Tumor Capillary leakage in travelers with dengue infection: Manila, 1931).
necrosis factor mediates disseminated intravascular implications for pathogenesis. Am. J. Trop. Med. Hyg. 73. Rathore, A. P. et al. Celgosivir treatment misfolds
inflammation (DII) in the genesis of multiple organ 86, 536–539 (2012). dengue virus NS1 protein, induces cellular pro-survival
edema. J. Surg. Res. 54, 140–144 (1993). 53. Balsitis, S. J. et al. Lethal antibody enhancement of genes and protects against lethal challenge mouse
34. Kunder, C. A., St John, A. L. & Abraham, S. N. dengue disease in mice is prevented by Fc modification. model. Antiviral Res. 92, 453–460 (2011).
Mast cell modulation of the vascular and lymphatic PLoS Pathog. 6, e1000790 (2010). 74. Watts, D. M. et al. Failure of secondary infection with
endothelium. Blood 118, 5383–5393 (2011). 54. Shresta, S., Sharar, K. L., Prigozhin, D. M., American genotype dengue 2 to cause dengue
35. Wang, L. et al. Implications of dynamic changes Beatty, P. R. & Harris, E. Murine model for dengue haemorrhagic fever. Lancet 354, 1431–1434
among tumor necrosis factor-α (TNF-α), membrane virus-induced lethal disease with increased vascular (1999).
TNF receptor, and soluble TNF receptor levels in permeability. J. Virol. 80, 10208–10217 (2006). 75. Thein, S. et al. Risk factors in dengue shock syndrome.
regard to the severity of dengue infection. Am. J. Trop. 55. de Borba, L., Strottmann, D. M., de Noronha, L., Am. J. Trop. Med. Hyg. 56, 566–572 (1997).
Med. Hyg. 77, 297–302 (2007). Mason, P. W. & Dos Santos, C. N. Synergistic 76. Vitarana, T., de Silva, H., Withana, N. &
36. Chakravarti, A. & Kumaria, R. Circulating levels of interactions between the NS3hel and E proteins Gunasekera, C. Elevated tumour necrosis factor in
tumour necrosis factor-α and interferon-γ in patients contribute to the virulence of dengue virus type 1. dengue fever and dengue haemorrhagic fever. Ceylon
with dengue and dengue haemorrhagic fever during PLoS Negl Trop. Dis. 6, e1624 (2012). Med. J. 36, 63–65 (1991).
an outbreak. Indian J. Med. Res. 123, 25–30 (2006). 56. Avirutnan, P. et al. Antagonism of the complement 77. Wahala, W. M., Huang, C., Butrapet, S., White, L. J. &
37. Halstead, S. B. et al. Dengue hemorrhagic fever in component C4 by flavivirus nonstructural protein NS1. de Silva, A. M. Recombinant dengue type 2 viruses
infants: research opportunities ignored. Emerg. Infect. J. Exp. Med. 207, 793–806 (2010). with altered e protein domain III epitopes are
Dis. 8, 1474–1479 (2002). 57. Ashour, J. et al. Mouse STAT2 restricts early dengue efficiently neutralized by human immune sera. J. Virol.
38. Kliks, S. C., Nimmanitya, S., Nisalak, A. & Burke, D. S. virus replication. Cell Host Microbe 8, 410–421 86, 4019–4023 (2012).
Evidence that maternal dengue antibodies are (2010).
important in the development of dengue hemorrhagic 58. Kochel, T. J. et al. Effect of dengue‑1 antibodies on Acknowledgements
fever in infants. Am. J. Trop. Med. Hyg. 38, 411–419 American dengue‑2 viral infection and dengue The authors thank A. P. S. Rathore for critical reading of the
(1988). haemorrhagic fever. Lancet 360, 310–312 (2002). manuscript. This work was supported by the National Medical
39. Hawkes, R. A. Enhancement of the infectivity of 59. Bennett, S. N. et al. Selection-driven evolution of Research Council of Singapore (grant NIG/1053/2011) and by
Arboviruses by specific antisera produced in domestic emergent dengue virus. Mol. Biol. Evol. 20, the Duke-National University of Singapore Signature Research
fowls. Aust. J. Exp. Biol. Med. Sci. 42, 465–482 1650–1658 (2003). Program, funded by the Ministry of Health, Singapore.
(1964). 60. Zivna, I. et al. T cell responses to an HLA‑B*07‑
40. Goncalvez, A. P., Engle, R. E., St Claire, M., restricted epitope on the dengue NS3 protein correlate Competing interests statement
Purcell, R. H. & Lai, C. J. Monoclonal antibody- with disease severity. J. Immunol. 168, 5959–5965 The authors declare no competing financial interests.
mediated enhancement of dengue virus infection (2002).
in vitro and in vivo and strategies for prevention. Proc. 61. Chiewsilp, P., Scott, R. M. & Bhamarapravati, N.
Natl Acad. Sci. USA 104, 9422–9427 (2007). Histocompatibility antigens and dengue hemorrhagic FURTHER INFORMATION
41. Daughaday, C. C., Brandt, W. E., McCown, J. M. & fever. Am. J. Trop. Med. Hyg. 30, 1100–1105 (1981). Duane J. Gubler’s homepage:
Russell, P. K. Evidence for two mechanisms of dengue 62. Loke, H. et al. Strong HLA class I–restricted T cell https://www.duke-nus.edu.sg/content/gubler-duane-j
virus infection of adherent human monocytes: trypsin- responses in dengue hemorrhagic fever: a double- ALL LINKS ARE ACTIVE IN THE ONLINE PDF
sensitive virus receptors and trypsin-resistant immune edged sword? J. Infect. Dis. 184, 1369–1373 (2001).

426 | JUNE 2013 | VOLUME 11 www.nature.com/reviews/micro

© 2013 Macmillan Publishers Limited. All rights reserved

You might also like