Вы находитесь на странице: 1из 25

Dengue

Marı́a G Guzman, Omar Fuentes, Eric Martinez, and Ana B Perez, PAHO/WHO Collaborating Center for the Study of Dengue and
Its Vector, ‘Pedro Kouri’ Tropical Medicine Institute, Havana, Cuba
Ó 2017 Elsevier Inc. All rights reserved.
This article is an updated version of the previous edition article by María G. Guzmán, Ana B. Perez, Omar Clemente Fuentes González, Gustavo Kouri,
volume 2, pp. 98–119, Ó 2008, Elsevier Inc.

Introduction During epidemics in Australia, 1897, Greece, 1928, and


Taiwan, 1931, a severe syndrome characterized by shock,
Vector-borne diseases are some of the most common and devas- hemorrhagic manifestations, and death was reported.
tating maladies. Currently, vectors that spread disease are an However, the association of hemorrhagic illness with dengue
important public health concern; however, they have been
neglected, with major consequences for health and socioeco- Table 1 Major dengue epidemics and pandemics up to World War II
nomic development. During the 1960s, most vector-borne
Years
disease control programs deteriorated. By the 1980s, many
important diseases such as dengue emerged in new areas or ree- Pre-nineteenth China (992)a; French West Indies (1635); Panama
merged in locations they had once called home. Dengue hemor- century (1699); Egypt, Indonesia, Philadelphia, India, Arabia,
rhagic fever and dengue shock syndrome (DHF/DSS) extended and Persia (1779–80)
from Southeast Asia and the Western Pacific to the American 1818 Lima, Peru
region. The 1998, 2002, and more recently the 2013 dengue 1824–28 Suez, Egypt; north of Calcutta, India; Curacao; Virgin
epidemics were unprecedented in terms of number of reported Islands; Jamaica; Lesser Antilles; Cuba; northern
Colombia; Vera Cruz, Mexico; Bermuda; southern
cases and the large geographic affected areas.
United States
Today dengue is the most rapidly spreading mosquito-borne
1845–68 St. Louis, United States; Senegal; Cairo, Egypt; Rio
disease in the world. In the past 50 years, dengue incidence has Janeiro, Brazil; Calcutta and Kanpur, India; Hawaii;
increased 30-fold with increasing expansion to new countries southern United States; Tahiti; Lima, Peru; Cadiz,
including extension from urban to rural areas (WHO, 2010). Spain; Canary Islands; Port Said, Egypt; Tanzania;
Kenya
1870–77 East African coast; Arabian peninsula; Bombay, Pune,
The Concept and Calcutta, India; Singapore; Java, Indonesia;
Dengue is an acute mosquito-transmitted disease of humans Taiwan; Mauritius Islands; southern United States;
caused by any of four serotypes of the dengue virus, with Aedes Lima, Peru
1883–90 Gibraltar; Greek Islands; southern Turkey; Syria;
aegypti (Ae. aegypti) the most important vector.
Palestine; the Nile delta of Egypt; Istanbul on the
Bosporus; Varna and Trabzon on the Nlack Sea;
Historical Summary Israel; Zanzibar; Tanzania; Fiji
1894–99 Townsville and Charters Towers, Brisbane, Australia;
According to some, the origin of the word ‘dengue’ has its roots Indochina and Hong Kong; India; Texas and Florida,
in the Swahili term ‘ki denga pepo,’ or a disease characterized by United States; Havana, Cuba; San Juan, Puerto Rico;
the sudden cramp-like seizure caused by an evil spirit, applied to Eritrea; Somalia
an outbreak in Zanzibar, 1870. The term ‘denga’ or ‘dyenga’ was 1901–07 Hong Kong; Bangkok, Thailand; Rangoon, Burma;
used to name the disease on the east coast of Africa in 1823. The India; Singapore and Penang, Malaysia; Hawaii; Java,
term ‘dengue’ spread with the slave trade from East Africa to the Indonesia; the Philippines; Taiwan; Brisbane,
Australia; Galveston and Houston, United States;
Caribbean islands and particularly from Cuba during the
Havana, Cuba; Natal, South Africa; Egypt
outbreak of 1828. Dengue means ‘affectation.’
1912–18 Panama; Meerut, northern India; Iquique, Chile;
Epidemics of classical dengue have been recognized for more northern Argentina; Queensland, Australia; San Juan,
than 200 years and doubtless occurred long before that. The first Puerto Rico; Taiwan; Rio Grande do Sul, Brazil;
accurate clinical description of dengue during the Philadelphia Sudan
epidemic (1780) was published by Benjamin Rush in 1789. 1920–28 Australia; India; Manila, Philippines; Taiwan; Ghana;
Outbreaks of possible dengue illness were reported during Niteroi, Brazil; from Texas to Florida and Georgia,
the eighteenth and nineteenth centuries along the Atlantic United States; Venezuela; Yemen; Senegal; South
coast, Caribbean islands, and in the Mississippi basin. Also in Africa; Athens, Greece; Egypt; Vietnam; Durban,
tropical Asia and Australia, dengue pandemics occurred at South Africa
1930–36 Pacific Islands; Taiwan; Japan; Java and Sumatra,
periods of 15–20 years during the nineteenth century, evolving
Indonesia; Malaysia; Burma; India; Florida, Georgia
to shorter intervals among pandemics during the first half of
and Alabama, United States; India; Ghana; Egypt
the twentieth century. Table 1 summarizes the major dengue 1940–45 Australia; Hawaii; Caribbean and South-East Asian
pandemics and epidemics up until World War II. countries; Japan; India; Philippines
Bancroft in 1906 first identified Ae. aegypti mosquitoes as
a
dengue vectors. This observation as well as the viral etiology Chinese Encyclopedia of Disease and Remedies (AD 265–420) was edited in AD 610
and again in 992.
of the disease was confirmed later by studies in human volun- Adapted from Gubler, D.J., Kuno, G. (Eds.), 1997. Dengue and Dengue Hemorrhagic
teers (Halstead, 2012). Fever. CAB International, Wallington, UK, with permission.

International Encyclopedia of Public Health, 2nd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803678-5.00103-X 233


234 Dengue

Figure 1 Annual number of dengue cases and countries with dengue report to PAHO/WHO, 1980–2013.

viruses was recognized during outbreaks of viral hemorrhagic Today severe dengue is observed in all countries with dengue
fever in Manila and Bangkok during the 1950s. Indeed, during transmission.
the 1950s, DHF/DSS was recognized as a syndrome associated Figure 1 shows the reported dengue cases and countries
with dengue viruses throughout Southeast Asia. with transmission by year in the Americas.
According to Gubler and Kuno (1997) and Gubler (1997), Table 2 presents the most important features reported in the
the ecologic disruption and demographic changes occurring Americas during the past 50 years of the twentieth century.
during and after World War II were conducive to increased Dengue surveillance in Africa has been very poor. At the end
transmission of dengue. The need for stored water contributed of the twentieth century, some epidemics were reported;
to the increased number of Ae. aegypti. Moreover, war facilitated however, currently the four serotypes have been isolated from
the transportation of the mosquitoes and their eggs to new Africa, and available data suggest that DENV is endemic in at
geographic areas through the transport of supplies and other least 34 countries (Were, 2012; Figure 2).
war materials. Conditions were thus created for the emergence
of DHF/DSS in Southeast Asia.
Current Epidemiological Situation
The second half of the twentieth century was characterized
by the geographic expansion of the disease, having been The beginning of the new millennium was characterized by an
preceded by the extension of the mosquito vector and the expanding distribution of Ae. aegypti and Ae. albopictus to most
dengue viruses. DHF/DSS extended first throughout Southeast tropical and subtropical areas of the world, co-circulation of
Asia and later to the Western Pacific to finally reach the Amer-
ican region. Countries evolved from a no endemic or Table 2 Main features of dengue epidemiology in the past 60 years
hypo-endemic situation to that of high endemicity. During in the American region
the 1980s and the 1990s, DHF/DSS became a major pediatric
Year Features
problem in Southeast Asia and the Western Pacific. By 2000,
dengue epidemics reached African and Eastern Mediterranean 1953 First dengue virus isolation (DENV-2 isolation in Trinidad
countries (Guzman and Kouri, 2003). During the 1950s, an and Tobago) in no epidemic situation
Ae. aegypti control program targeting urban yellow fever preven- 1963–64 DENV-2 and DENV-3 epidemic
tion in the American region had eliminated the vector from 1968–69 DENV-2 and DENV-3 epidemic
many countries, and consequently there was a decrease or elim- 1977 DENV-1 introduction in Jamaica
ination of dengue epidemics. However, due to the deteriora- 1981 First DHF/DSS epidemic, Cuba
tion of the mosquito control programs, the reinvasion by Ae. 1981 DENV-4 introduction in Caribbean countries
1985 Introduction of Ae. albopictus mosquito
aegypti started in some countries by the 1970s, continuing
1989 Second DHF/DSS epidemic, Venezuela
during the 1980s and the 1990s (Guzman and Kouri, 2003; 1994 Reintroduction of DENV-3 in Nicaragua, Panama, and
San Martin et al., 2010; Brathwaite et al., 2012). Costa Rica
The first DHF/DSS epidemic in the Americas was reported in 2013 More than 2 million cases reported
1981 in Cuba. More than 344 000 patients with more than Current Expansion of Ae. aegypti mosquito, dengue
10 000 severe and very severe cases and 158 fatalities (101 chil- situation hyperendemicity with co-circulation of multiple
dren) were reported (Kouri et al., 1989). After the second DHF serotypes, endemicity of DHF
epidemic occurred 8 years later in Venezuela, an increase in the
Adapted from Guzman, M.G., Kouri, G., 2003. Dengue and dengue hemorrhagic
number of DHF cases and epidemics and in the number of fever in the Americas: lessons and challenges. J. Clin. Virol. 27, 1–13 with
countries reporting the severe syndrome has been observed. permission.
Dengue 235

Known endemic

Suspected endemic

Figure 2 Known or suspected dengue endemic in Africa. Reproduced from Were, F., 2012. The dengue situation in Africa. Paediatr. Int. Child
Health 32, 18–21.

multiple serotypes of dengue virus, development of endemicity (2009–10; 2013), and southeastern France (2010), with an
of severe dengue in many countries of the tropical world, and epidemic of more than 2000 cases in Islas Madeiras, Portu-
increased frequency of dengue activity (Simmons et al.,2012; gal. Coinfections of dengue with malaria, HIV, leptospirosis,
Figure 3). influenza, chikungunya, and Zika virus have been increas-
According to WHO, dengue has been recognized in over ingly reported.
100 countries and causes an estimated 50–100 million infec- In this last decade, travelers have played an important role
tions annually with more than 2.5 billion people at risk. There in global dengue epidemiology, carrying viruses from one
are about 500 000 DHF cases and an average case fatality rate of region to another (Wilder-Smith et al., 2014). Figure 4 exem-
5% (around 20 000 deaths). Recent estimates report plifies travellers from dengue endemic countries that Islas
390 million dengue infections with 96 million cases annually Madeiras received in 2012.
worldwide, more than three times the dengue burden esti- Finally, a very interesting observation is that while malaria
mated by WHO (Bhatt et al., 2013). Although the distribution is declining, the number of dengue cases is rising. This fact
and burden of dengue is still not well characterized, the disease supports the serious epidemiological global dengue situation
is endemic in Africa, the Americas, the Eastern Mediterranean, (Table 3; Brady et al., 2012)
Southeast Asia, and the Western Pacific. Circulation of the Dengue burden determination is still a research priority. As
four serotypes has been reported in these regions. not all infections are symptomatic, asymptomatic infections
Particularly, an increase in dengue transmission in Africa, should be considered as well as those infected individuals
India, The Americas, and Eastern Mediterranean countries with a mild illness not detected by the surveillance systems.
such as Pakistan, Saudi Arabia, Sudan, and Yemen character- Figure 5 shows the estimated number of symptomatic and
ized the last decade. An example of the dengue situation is inapparent dengue infections by geographic region (Bhatt
the recent report in 2013 of more than 2 million cases in et al., 2013). The real impact of asymptomatic infections in
the American region, the highest figure in the history of dengue transmission is not well known, although a very recent
dengue in this geographic area. Dengue has resurged in study has supported the contribution of individuals with an
Hawaii, Galapagos Islands, Easter Island, and Buenos Aires. asymptomatic DENV infection to the dengue transmission
Autochthonous dengue transmission has been recently dynamic by efficiently infecting mosquito vector (Duong
confirmed in Hawaii (2015), Japan (2014), Florida et al., 2015). This issue constitutes a research priority.
236 Dengue

Figure 3 Dengue risk areas. Simmons, Cameron P., Farrar, Jeremy J., Van Vinh Chau, Nguyen, Wills, Bridget, 2012. Dengue. N. Engl. J. Med. 366,
1423–1432.

Figure 4 Map of air travel volume from endemic countries and cities to islas Madeiras, 2012. Reproduced from Wilder-Smith, A., Quam, M.,
Sessions, O., Rocklov, J., Liu-Helmersson, J., Franco, L., Khan, K., 2014. The 2012 dengue outbreak in Madeira: exploring the origins. Euro Surveill.
9 (8), 19. Available online: http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId¼20718.
Dengue 237

Table 3 Comparison of dengue and malaria burden and possible recombination. The major drivers of geographic
dengue spread and increased incidence are (Gubler, 2011):
Dengue
l lack of effective mosquito control,
Brady et al. WHO malaria l changing life styles,
WHO (2012) report, 2013
l unplanned urbanization,
Population at risk 2.5 billion 3.97 billion 3.4 billion l globalization.
Number of endemic >100 128 97
countries
Number of infections 50 million 70–500 million 219 million The Agent
per year
Hotta and Kimura in 1943 were the first to isolate dengue
viruses through inoculating the brains of suckling mice with
the serum of clinically ill dengue patients. Almost simulta-
neously (1944), Sabin isolated viruses from US soldiers in
Global
India, New Guinea, and Hawaii. These first isolations were later
classified as serotypes 1 and 2 (DENV-1 and DENV-2). Sero-
Oceania types 3 and 4 (DENV-3 and DENV-4) were isolated by Ham-
Symptomac
Americas mond during the epidemic of Manila, Philippines in 1956.
Inapparent
These closely related virus serotypes belong to the genus fla-
Asia
vivirus, family Flaviviridae. This family of enveloped RNA
Africa
viruses causes significant disease both in humans and animals.
0 100 200 300 400 Flavivirus, the largest of the three genera of the family,
comprises over 70 viruses, mostly arthropod-borne viruses,
Figure 5 Estimates of symptomatic and inapparent dengue infections with yellow fever virus the prototype.
by geographic regions. Adapted from Bhatt, S., Gething, P.W., Brady, Mature particles of dengue viruses are spherical with a diam-
O.J., Messina, J.P., Farlow, A.W., Moyes, C.L., Drake, J.M.,
eter of 50 nm, containing multiple copies of the three structural
Brownstein, J.S., Hoen, A.G., Sankoh, O., Myers, M.F., George, D.B.,
virus proteins, a host-derived membrane bilayer, and a single
Jaenisch, T., Wint, G.R., Simmons, C.P., Scott, T.W., Farrar, J.J.,
Hay, S.I., 2013. The global distribution and burden of dengue. Nature, copy of a positive-sense, single-stranded RNA genome. The
496, 504–507. genome of the four viruses contains a single open reading frame
of 10 233 nucleotides encoding for a polyprotein ordered
as 50 -C-prM-E-NS1-NS2A-NS2B-NS3-NS4A-NS4B-NS5–30 . The
The current pandemic and the increase in dengue epidemics full-length polypeptide is processed by viral and host proteases
are the direct result of changes in demographic and economic into the capside (C), membrane (M), and envelope
trends, such as (E) structural proteins that make up the virus and seven
l the global population explosion, with an increased pop- nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B,
ulation density; and NS5). The C protein binds strongly with RNA to form the
l unplanned urbanization (resulting in substandard housing nucleocapsid. The pre-membrane (prM) glycoprotein
and inadequate water, sewer, and waste management represents the precursor of the mature M protein. It has
systems); a crucial role, as it prevents the E glycoprotein from
l rapid movement of individuals potentially infected with undergoing an irreversible acid-catalyzed conformational
dengue, both within and between countries, favoring the change during its transportation through host cell acidic
introduction of dengue viruses; compartments. The major structural protein (E) represents the
l deterioration and breakdown of the public health infra- target for neutralizing antibodies, protective immunity, and the
structure that through surveillance monitors for diseases antibody-dependent enhancement (ADE) phenomenon. It also
such as dengue; mediates receptor binding and pH-dependent membrane
l deterioration of vector control programs that had controlled fusion activity. E glycoprotein is divided into three functional
populations of Aedes and other mosquitoes: domains, termed domain I, II, and III. Domain I, the hinge
region, is linked to the two other regional functional domains
B climate change and particularly temperature and
(this region is responsible for the changes in E structure due to
humidity changes are factors that favor increased
variation of external pH). Domain II has a hydrophobic-rich
dengue transmission;
peptide sequence featuring the membrane fusion activity.
B social and economic factors (e.g., poverty, illiteracy,
Domain III is probably involved in the binding to receptor
and low-risk perception).
molecules on the host cell membrane. NS1 glycoprotein can
Finally, new dengue strains can enter into a specific popula- be found on the cell surface, inside the cell, and as a secreted
tion via migration (gene flow), as dengue viruses are often form. It may have a role in RNA replication and in the
transported large distances by hosts and vectors. This fact, in pathogenesis of clinical disease.
conjunction with the increase in the size and density of the The NS3 protein has three enzymatic activities, a trypsin-like
human host population, provides more opportunities for viral serine protease, an RNA helicase, and an RNA triphosphatase
transmission with an extension of serotypes and genotypes activity. Together with NS2B, it forms the protease domain of
with a wider geographic distribution, greater mixing of strains, the virus.
238 Dengue

Protein MW Function
E (kDa)

E 55-60 Cell receptor binding, envelope fusion.


M
prM 18-19 Proper E folding and assembly. Avoid E
premature fusion during secretion.

C C 9-12 Binds the RNA to form the nucleocapsid

NS1 42-50 Viral replication??

NS2a 22 RNA synthesis and virus assembly??


RNA
NS2b 14 Cofactor of serine protease activity of NS3.

NS3 67-70 Serine protease (polyprotein processing),


NTPase and helicase (synthesis of viral RNA),
5´ Positive single-stranded RNA (11 kb) 3´ and triphosphatase (capping pathway).

Cap ORF NS4a 16 RNA synthesis and virus assembly??

Structural proteins Non structural proteins NS4b 27 RNA synthesis and virus assembly??

NS5 104- RNA dependent- RNA polymerase (synthesis


106 of viral RNA), and methyltransferase
C prM E NS1 NS2 NS3 NS4 NS5 (capping pathway).

Cytoplasm
Host signalase
Endoplasmic Viral serine protease (NS2b-NS3)
reticulum Host furin
Unknown

Intralumen space

Figure 6 Dengue virus genome, viral proteins and their functions and polyprotein processing in cell endoplasmic reticulum.

Functions of NS2A, NS4A, and NS4B are not well known humans. Heparan sulfate is thought to be a co-receptor,
but are believed to play a role in RNA replication. which associates with other molecules to form functional
NS5 is the largest and most conserved viral protein, and it is complexes and enhances the efficiency of virus infection into
considered the putative RNA-dependent RNA polymerase host cells. DC-SIGN serves as an important attachment factor
responsible for viral RNA replication. The methyltransferase for dengue virus on dendritic cells. Apparently carbohydrate
activity of this protein is probably involved in the molecules in the extracellular matrix are strongly related to
methylation of the 50 cap structure (Figure 6). DENV receptors (Hidari and Suzuki, 2011).
Dendritic cells (skin Langerhans cells), monocytes, and After attachment of E protein to surface host receptors,
macrophages seem to be the primary target cells in vivo for virus virions enter cells via receptor-mediated endocytosis. Later,
replication. In vitro studies suggest that fibroblasts, keratinocytes, virions are found in uncoated prelysosomal vesicles, where
endothelial and hepatic cells, and neurons can also be infected. an acid-catalyzed membrane fusion releases the nucleocapsid
Mastocytes are also targets for DENV infection (Figure 7). into the cytoplasm. The viral genome is thus released into the
Several putative receptors have been identified on human cytoplasm and is used as a template for a large precursor poly-
and mosquito cell types. Studies suggest that heparin sulfate protein. In the process of virus attachment to cell surface and
and DC-SIGN are indispensable for dengue virus infection in membrane fusion, domains I, II, and III of the E glycoprotein

DC-SIGN FcγR Y IgG antibody Virus

Figure 7 Major target cells of dengue virus infection.


Dengue 239

1. Binding
2. Receptor-mediated
endocytosis
3. Low ph-mediated
membrane fusion
4. Uncoating
5. Translation
6. Polyprotein processing
and RNA replication
7. Virion assembly
8. Virion transportation and
maturation
9. Virion release

Figure 8 Replication cycle of dengue virus.

play an important role. After synthesis of RNA strands of nega- and Bartenschlager, 2011). Therefore, partially immature parti-
tive polarity, the plus-stranded RNA molecules are generated. cles expand the pool of infectious virus and likely contribute to
These genomic plus-stranded RNAs (RNSs) serve as templates the pathogenesis of the illness as they probably are involved in
for further negative strand production, or they are packaged ADE and the neutralization phenomena (Voorham et al., 2012;
into the new virus particles. Rodenhuis-Zybert et al., 2011a,b).
Cleavage at the amino-terminal portion of the polyprotein by Dengue viruses show substantial genetic diversity (Holmes
host cell enzymes produces the structural proteins that form and Twiddy, 2003; Holmes and Burch, 2000). In addition to
superstructure for the enveloped virus. The remaining viral poly- the four serotypes, clusters of variants (known as genotypes)
protein is cleaved by the virus protease, producing the NS have been identified within each serotype. Molecular epidemi-
proteins that are required for producing more virus copies. ologic approaches have demonstrated the extensive genetic
Translocation of prM, E, and NS1 into the lumen of the variability of these viruses (Table 4). DENV-1, DENV-2, and
endoplasmic reticulum occurs co-translationally and is initi- DENV-3 classify into five genotypes, while DENV-4 into 4
ated by a signal sequence at the C terminus of the C protein. genotypes. In addition to genotypes, also intra-genotypic
Host signalized cleavages at the prM–E and E–NS1 junctions. groups within each genotype have been suggested (Amarilla
Soon after translation, the prM and E proteins associate as a het- et al., 2009). There is some evidence that recombination in
erodimeric complex. The prM acts as a ‘chaperone’ to aid the natural populations of DENV serotypes could contribute to
folding and maturation of E protein. These heterodimers drive the genetic variation of DENV. The circulation of different sero-
the assembly and budding of immature particles at the rough types and genotypes of DENV in a particular geographic area
endoplasmic reticulum membrane. Prior to or during release has been documented as well as the coexistence of two different
of the infectious virus from cells, the host enzyme furin cleaves serotypes in a given mosquito or patient, making feasible the
the prM protein, probably dissociating the prM/E heterodimer, recombination phenomena.
resulting in rearrangement of the E proteins into dimers on the At the population level, the replacement of one viral geno-
surface of the mature particle. The pr protein is secreted and the type for another of the frequent lineage replacement within one
M protein remains anchored in the virus membrane. The genotype and mutations of nonstructural proteins have been
matured virions are transported through an exocytic way into associated with a higher virus transmissibility and epidemic
the plasma membrane and released to extracellular space via severity, respectively. Studies in Mexico support that, for each
secretion (Figure 8). Both mature and immature virions are
produced during in vitro DENV propagation; however, the Table 4 Genotypes within dengue serotypesa
maturation state of dengue virions in humans is unknown.
Differences in the conformation of E and prM/M proteins on Serotype Genotypes
the surface of mature and immature virions may have impor-
DENV-1 Sylvatic/Malaysia, Americas/Africa, South Pacific, Asia,
tant implications in dengue pathogenesis and vaccine develop- and Thailand
ment. Recent studies have supported that infectivity of DENV-2 American, American/Asian, Asian 1, Asian 2, Cosmopolitan,
immature particles is restored by prM-specific antibodies, and Sylvatic
highly cross-reactive among the dengue serotypes, permitting DENV-3 Southeast Asia/South Pacific, Thailand, Indian subcontinent,
virus entry via the Fc receptor expressed on the surface of and Americas
immune cells and presumably targeting the virus sites where DENV-4 Sylvatic/Malaysia, South-East Asia, and Indonesia
furin-mediated cleavage of prM occurs. As these antibodies a
Classification in genotypes uses nucleotides from the entire E gene region.
are not present during a primary DENV infection, enhanced Rico-Hesse, R., 2003. Microevolution and virulence of dengue viruses. Adv. Virus
secondary infection might be due to these antibodies (Fischil Res. 59, 315–341.
240 Dengue

serotype, multiple introductions of viral lineages have occurred Older populations, having taken many blood meals, have
with little co-circulation (Carrillo-Valenzo et al., 2010). Virus a greater potential for virus transmission.
evolution in Mexico is typified by frequent lineage replace- Females usually fly no more than 50 m, but they can easily
ment, with only a single lineage dominating a specific serotype fly 100–200 m, and can travel 3 km in search for a site to
at a specific time point. A different situation was reported in oviposit in. They can be transported by cars, trucks, aircraft,
a highly endemic area such as Vietnam where multiple lineages and even hurricanes for still longer distances. Ae. aegypti
of DENV-1 were observed during the period of 2003–08 sug- mosquitoes are peridomestic; that is, they prefer to rest inside
gesting equivalent viral fitness (Raghwani et al., 2011). In the the house, rather than in the garden. Most resting is on walls.
same area, previous studies suggested that a DENV-2 genotype These mosquitoes can live for months; yet most usually
replacement occurred (Asian-American genotype by Asian I survive only a few weeks. Half of them die in the first week
DENV-2 genotype). The higher viremia produced during the and 95% in the first month of life.
Asian I DENV-2 infections would likely increase the probability The mosquito’s preferred breeding sites are in areas of stag-
of human to mosquito transmission and in consequence facil- nant water, such as flower vases, uncovered barrels, buckets,
itate greater population diffusion and possibly an increased and discarded tires. The most dangerous areas are wet shower
incidence of more severe disease (Hang et al., 2010). Recently, floors and toilet bowls, as they allow the mosquitoes to breed
Rodriguez-Roche et al. (2012) studying isolates from Vene- right in the residence. Universally, automobile and truck tires
zuela found that changes at nonstructural proteins such as are the main breeding sites for these mosquitoes.
NS1, NS2A, and NS4B may play an important role in virus Other mosquitoes such as Ae. albopictus, Ae. polynesiensis,
evolution. In the same study they have demonstrated the and some forms of Ae. scutellaris can also transmit the disease.
co-circulation of several lineages of DENVs and an association Generally Ae. aegypti is the main vector in urban transmission,
of DENV-2 with severe cases (Rodriguez-Roche et al., 2012). but Ae. albopictus is shown to be more competent and more
susceptible to experimental infection. Ae. albopictus may be
a less selective feeder so it is of less epidemiological signifi-
Dengue Vectors cance, as it may bite nonhuman hosts as well as humans,
diluting its capacity to acquire and transmit dengue.
Dengue viruses are transmitted to humans by mosquitoes from The DENVs originated and are maintained in a forest trans-
the Aedes genus and mainly by Ae. aegypti. They are widely mission cycle involving forest-dwelling Aedes mosquitoes and
distributed generally within the limits of 40 North or South lower primates from Asia and Africa. Sylvatic cycles in Asia
latitude, being most frequently found in the tropics (Figure 9). are associated with Macaca and Presbytus sp. monkeys and
Females feed on any vertebrate host, but prefer humans mosquitoes of the genus Ochlerotatus, and in Africa with Eryth-
(Christophers, 1960). They fly upwind, following chemoattrac- rocebus patas monkeys and several sylvatic mosquitoes such as
tant odors. The first step can be to enter a house. Blood feeding Ae. taylori, Ae. furcifer, Ae. luteocephalus, and Ae. opok. In Asia,
and oviposition occur mostly in the morning and late in the circulation of sylvatic DENV-1, DENV-2, and DENV-4 has
afternoon. Only the female bites for blood, which she needs been detected in sylvatic mosquitoes and/or sentinel monkeys.
to mature her eggs. In Africa, sylvatic DENV-2 has been associated to sylvatic
She takes a complete blood meal of 2–3 ml of blood, and mosquitoes (Franco et al., 2011).
will produce a batch of about 100 eggs in about 3 days. The urban cycle, mosquito to human to mosquito, is the most
Stomach distention triggers ovarian development. Thus, important in terms of human transmission and disease outcome;
smaller blood meals produce fewer eggs, and refeeding with however, some sylvatic DENV isolates from patients have been re-
repeated biting by the same female occurs when the volume ported including one from a DHF case (Franco et al., 2011).
of ingested blood is too small for efficient egg production.
The Disease
Dengue infection may be clinically unapparent or cause an
illness with varied intensity, including patients with fever
and body pains to the severe picture of shock and large
hemorrhages with or without organ impairment. Plasma
leakage rather than bleeding is a major determinant of most
cases of dengue shock, associated with significant hematocrit
elevations and accumulation of fluid in serous cavities, such
as pleural effusions, ascites, and pericardial leakage. Dengue
severity can also be associated with encephalopathy, cardio-
myopathy, or hepatopathy, as well as kidney dysfunction
(Simmons et al., 2012).
Dengue is a very dynamic illness, in spite of its short dura-
tion (no more than 1 week in nearly 90% of the cases). Its clin-
ical expression can change as the days go by and can also
worsen suddenly. Dengue illness can evolve into three phases:
the febrile phase – observed in most of the patients – and the
Figure 9 Ae. aegypti mosquito. critical and the recovery phases (WHO, 2010).
Dengue 241

scope of this review; however, a very good description appears


at the WHO guidelines for dengue prevention and control
(WHO, 2010).

New Dengue Clinical Classification

For three decades the WHO recommended the classification of


dengue as dengue fever (DF) and DHF with or without DSS. Clin-
ical manifestations of DF should include fever and two of the
following symptoms: headache, retroocular pain, osteomyoartic-
ular pains, rash, leukopenia, and bleeding (WHO, 1997).
Figure 10 Petechiae in a DHF patient. Kindly supplied by S. Zagne, Four criteria characterize DHF: (1) fever or history of fever in
Niteroi, RJ, Brasil. the previous week; (2) spontaneous bleeding – usually pete-
chiae, or other – or, at least, a positive tourniquet test; (3)
thrombocytopenia less than 100 000 per mm3; and (4) plasma
The febrile phase is variable in duration and is associated leakage, evidenced by a 20% elevation of the hematocrit level,
with the level of viremia (virus in blood). Fever is generally or by a 20% decrease of the hematocrit level after the critical
the first clinical manifestation of illness with a variable inten- phase of illness, or by the presence of pleural effusion, ascites,
sity. It is associated with headache and vomiting, as well as or pericardial leakage determined by imaging studies, usually
body pains. In children, fever is frequently the only clinical sonography (WHO, 1997).
manifestation or is associated with rash and/or unspecific In recent years several authors questioned the usefulness of
digestive symptoms. The pharynx may become reddened, the former WHO dengue clinical classification in DF and DHF
but other symptoms and signs of the respiratory system are (Bandyopadhyay et al., 2006; Deen et al., 2006; Rigau-Perez,
not frequent or important (Capeding et al., 2010). A slight 2006). According to them, this classification fails as it is too
abdominal pain and diarrhea may occur. Diarrhea is more dependent on laboratory results, and also it does not include
frequently reported in patients younger than 2 years of age severe dengue patients with organ impairment such as patients
and in adults. The leukocyte count is usually decreased. Pete- with encephalitis, myocarditis, and severe hepatitis. Addition-
chiae or ecchymosis can be present, with or without thrombo- ally, the classification is not useful for patient clinical manage-
cytopenia (Figure 10). ment, either. Taking into account these arguments the Special
Most cases recover after defervescence, however in some, the Programme for Research and Training in Tropical Diseases
patient’s state worsens when the fever drops. Therefore, the (TDR/WHO) conducted an international study as part of the
period during which the fever subsides indicates the beginning DENCO (Dengue Control) projects, one of whose components
of the critical phase. was clinical, with the main purpose to obtain information from
Dengue is a dynamic disease (Balasubramanian et al., 2012). a high number of patients with a laboratory confirmed dengue
The critical phase coincides with the leakage of plasma that infection and to find a better way to clinically classify them, as
can lead to shock, with coldness in the teguments, weak pulse, well as to identify warning signs of severity useful to improve
delayed capillary filling, tachycardia, and hypotension. At the patient management.
beginning, not all clinical signs of shock are observed being Clinical information was obtained from nearly 2000 patients
enough to detect a narrowing of the differential arterial tension (from seven countries in two continents) with confirmed
(AT) or pulse pressure (difference of 20 mmHg or less between dengue. The study concluded that 18–40% of the cases could
the maximum or systolic AT and the minimum or diastolic AT). not be classified by the WHO Classification as DF or DHF,
At this stage, the hematocrit level is increased and the platelets and over 15% of cases with shock could not be classified as
– which were decreasing already – reach their lowest figure. severe cases of dengue either, since they did not fill the criteria
With an adequate and appropriate fluid administration, of DHF/DSS. Another consistent result was the binary classifica-
dengue shock is rapidly reversible, although it may recur during tion of the disease as dengue and severe dengue (Alexander et al.,
the next 12–24 h, and sometimes more. The prognosis depends 2011; Figure 11), considering that a spectrum of disease exists
primarily on the early recognition and treatment of shock, rather than two distinct entities (Farrar et al., 2013).
which depends on careful monitoring and prompt action, for Criteria for severe dengue were (1) severe plasma leakage,
preventing other complications such as metabolic acidosis, expressed by hypovolemic shock, and/or difficulty breathing
multiorgan failure, major hemorrhages, and disseminated due to the fluid accumulated within the lungs; (2) severe
intravascular coagulation (DIC) (Khan et al., 2008). At the hemorrhage, according to the criterion of the assisting physi-
recovery phase, the patient’s improvement is usually evident, cian; and (3) organ damage: severe hepatitis (transaminases
but on occasions a state of fluid overload occurs, as well as higher than 1000 units), encephalitis, or a serious damage of
secondary bacterial infections. other organs, such as myocarditis. These severity criteria had
The differential diagnosis of dengue includes influenza, 95% of sensitivity and 97% specificity (Alexander et al.,
exanthematous infections (measles or rubella), severe bacterial 2011). The new WHO Dengue Classification was validated in
infections (meningococcemia, bacterial sepsis, typhoid fever, 18 countries (Barniol et al., 2011). Further studies have sup-
or leptospirosis), and other viral hemorrhagic fevers. Details ported the usefulness of the new classification (Basuki et al.,
of the treatment of dengue and severe dengue are out of the 2010; Narvaez et al., 2011; Lima et al., 2013).
242 Dengue

Dengue ± Warning signs Severe dengue

1.Severe plasma leakage


With 2.Severe hemorrhage
Without 3.Severe organ impairment
warning
signs

Presumptive diagnosis 1. Severe plasma leakage leading to


•Fever, and two or Warning signs* •Shock (DSS)
more> •Abdominal pain or tenderness •Fluid accumulation with
•Vomiting, nausea •Persistent vomiting
•Rash respiratory distress
•Clinical fluid accumulation 2. Severe bleeding
•Body pains •Mucosal bleed
•Leucopenia as evaluated by clinician
•Lethargy; restlessness 3. Severe organ involvement
•Petechiae or Tourniquet
•Liver enlargement >2cm Liver: AST or ALT>=1000
test
•Laboratory: Increase in HCT CNS: Impaired consciousness
Neighbourhood concurrent with rapid decrease heart and other organs
dengue/history of travel in platelet count
to dengue endemic area * Requiring strict observation and medical intervention

Figure 11 WHO dengue classification, 2010. Reproduced from WHO, 2010. Working to Overcome the Global Impact of Neglected Tropical
Diseases. First WHO report on neglected tropical diseases, Geneva.

‘Warning signs’ such as intense and continuous abdominal outbreaks. The intensity of dengue transmission and the
pain, frequent vomiting, sleepiness, and/or irritability, as well simultaneous circulation of several serotypes are also of
as a sudden decrease in temperature leading to hypothermia importance. An eco-biosocial conceptual framework useful
sometimes associated with fainting due to orthostatic hypoten- for research investigation has been proposed. Figure 12 inte-
sion are indicators of the presence of shock. These signs appear grates both the integral hypothesis proposed for the develop-
more frequently at defervescence, and allow identification of ment of DHF/DSS and the eco-biosocial conceptual
the loss of fluid to the extracellular space, which the patient framework (Arunachalam et al., 2014).
can hardly compensate or cannot compensate at all (Gibson
et al., 2013; de Andrade et al., 2014). The revised WHO/TDR Environmental Factors
(2009) classification shows a very high sensitivity for identi- In general, the epidemiological and ecological factors are
fying severe dengue and is easy to apply (Prasad et al., 2009). important determinants for the development of a dengue
The better performance of the revised classification in the detec- outbreak. Figure 12 shows the macrodeterminants of dengue
tion of severe clinical manifestations allows for improved transmission. Vector factors such as abundance and types of
detection of patients with severe dengue and may reduce deaths mosquito production sites, density of adult females, age of
(Pamplona et al., 2014). females, frequency of feeding, host preference, host availability,
and innate susceptibility to infection are among the microde-
terminants of dengue transmission.
Risk Factors for Dengue Transmission and DHF/DSS
Transmission dynamics of dengue virus are determined by the Host Factors
interaction of the environment, the agent, the host population, Individual risk factors determine the expression of the disease
and the vector. These components can be divided into macro- in a particular person in a given population. The presence or
determinants and microdeterminants. The new WHO dengue absence of these individual risk factors in the matrix of epide-
clinical classification was introduced in 2010, hence most path- miological and viral factors not only defines whether or not
ogenesis studies, including risk factor studies for severe disease, the persons with a secondary dengue infection develop a clin-
have been done using the old WHO clinical classification, ical picture of DHF, but also determines the infection outcome.
namely, DF and DHF/DSS.
In particular, DHF occurs as a consequence of a very Individual Risk Factors
complex mechanism where the virus, host, and environment Infection with one serotype gives long-lasting immunity to this
interact in such a way that this severe form of the disease serotype and short-lived protection against the others. Homo-
occurs in 2–4% of individuals with a secondary infection. typic immunity is probably based on neutralizing antibodies.
An integral hypothesis for the development of DHF The transient nature of heterotypic immunity is probably due
epidemics was early proposed and later documented to cross-reactive antibodies to E protein. Over time, these anti-
(Kouri et al., 1989). The intersection of the three groups of bodies decline and an individual is susceptible to infection
factors determines the occurrence of the severest dengue with a different DENV serotype.
Dengue 243

Social context
Vector exposition
Risk perception
Health system: case
Age. Sex management, early detection,
chronic diseases quality of care, availability of
Nutritional status medical supplies, equipment
Ethnicity
Viral factors
Circulating serotypes/
Immunity genotypes
Previous infection Viral load: viraemia titres
ADE Viral tropism
Original antigenic sin Virulence
Host Virus evolution
Transient autoimmunity Virus
Unbalanced T-cell Sequence of serotypes
responses

Genetic factors
Vector control
Genes involved in cell
Policies: Legislation related to
entry, replication, Vector control activities
immune response
Control actions: surveillance,
type/coverage of actions,
costs, human resources,
Vector ecology eficiency/sustainability.
Climate variability Social context Stakeholders
Availability/quality of Population density,
breeding sites urbanization, migration
Feeding opportunities Housing, habits, traditions
Vector capacity Water supply
Vector species Poverty

Figure 12 Eco-biosocial conceptual framework for the development of dengue. Adapted from Kouri, G.P., Guzman, M.G., Bravo, J.R., 1987.
Why dengue haemorrhagic fever in Cuba? 2. An integral analysis. Trans. R. Soc. Trop. Med. Hyg. 81, 821–823 and Arunachalam, N., Tana, S.,
Espino, F., Kittayapong, P., Abeyewickreme, W., Thet Wai, K., 2010. Eco-bio-social determinants of dengue vector breeding: a multicountry study in
urban and periurban Asia. Bull. World Health Organ. 88, 173–184. http://dx.doi.org/10.2471/BLT.09.067892..

A secondary heterotypic infection (e.g., a second infection of virus load in blood and the fast dissemination of the infection
caused by a different serotype of dengue virus) is associated (Figure 13). The outcome of a secondary DENV infection is
with the development of DHF/DSS, and is considered the main modulated by the individual’s age, infection with a different
individual risk factor for severe disease. Maternally transferred (second) serotype of DENV, sequence of infection, and interval
or naturally acquired antibodies against one serotype do not between infections.
prevent against infection with another serotype. In fact, previous Longer intervals between sequential infections, when
exposure to one serotype may exacerbate disease caused by expo- heterologous sequential infections occur, generally do not
sure to a second serotype. The ADE phenomenon is mainly medi- protect against clinical disease. Guzman et al. (2002b) have
ated by IgG cross-reacting but not cross-neutralizing antibodies reported a higher disease severity after more than 20 years
specific to E and prM dengue proteins. Uptake of the virus by of the primary infection in individuals secondarily infected
FcgR expressing cells (monocytes/macrophage) increases by DENV-2 and DENV-3. Recently, this observation has
through ADE, and severe disease may result from higher levels been extended to symptomatic dengue infection as a long

Binding of virus/Ab Enhancement of Higher viral load


Aiding of virus
complexes to FcR virus replication and number of
entry
infected cells
Y
Y

Monocyte
Fc γ R Y Non-neutralizing IgG antibody Virus

Figure 13 Antibody-dependent enhancement.


244 Dengue

DSS Fatal cases


14 14

12 12

10 10

8 8

6 6

4 4

2 2

0 0
<1 2 4 6 8 10 12 14 <1 2 4 6 8 10 12 14
Age Age

Figure 14 Age distribution in DSS and fatal children cases, 1981 DHF/DSS epidemic. Adapted from Bravo et al., 1987. Trans. R. Soc. Trop. Med.
Hyg. 81, 816–820 with permission.

interval between sequential DENV infections has been asso- a secondary infection. Figure 14 shows the age distribution
ciated with symptomatic as opposed to inapparent infection of DSS and fatal children cases during the Cuban 1981
(Montoya et al., 2013). In children, primary infection with epidemic. No severe or fatal cases were observed in children
any DENVs varies from inapparent to mild disease while in aged 1–2 years who developed a primary dengue infection.
adults the disease is often overt mostly as dengue. Both age This observation supports the role of secondary infections as
and DENV serotypes are important determinants of infection major risk factors for DHF/DSS.
outcome. Primary DENV-2 and DENV-4 infections in Age was demonstrated to be an important variable in the
children are usually silent. In adult, primary DENV-2 infec- outcome of secondary DENV-2 infections. During the 1981
tions are also silent (Guzman et al., 2000, 2002a; Halstead, DHF Cuban epidemic, DHF/DSS case fatality and hospitalization
2012). rates were highest in young infants and the elderly. Indeed, the
DHF presents primarily as a childhood disease in South- risk that a child would die during a secondary DENV-2 infection
east Asian countries, while in the tropical Americas all age was nearly 15-fold higher than adults aged 15–39 years
groups are involved. Epidemic adult DHF/DSS was observed (Figure 15). Similar observations have been reported in Vietnam
for the first time during the Cuban DHF epidemic of 1981. and Singapore (Whitehorn and Simmons, 2011). Differences in
Subsequently, reports of DHF in adults have progressively baseline microvascular permeability between children and adults
increased, initially in the Americas and now in some South- could contribute to this phenomenon (Gamble et al., 2000).
east Asian countries. In children, DHF can present in infants A higher frequency of DHF/DSS in females has been re-
born to dengue-immune mothers and in children with ported by some (Anders et al., 2011).

30

25

20 Deaths/havana
Deaths/10 000

Deaths/cuba

15

10

0
3−4 5−9 10−14 15−19 20−24 25−29 30−34 35−39 40−44 45−49 50−54 55−59 60−64 65+
Ages

Figure 15 Age-specific DHF/DSS death rates per 10 000 secondary DENV-2 infections in Havana, and Cuba, 1981. Adapted from Guzman, M.G.,
Kouri, G., Bravo, J., Valdes, L., Vazquez, S., Halstead, S.B., 2002a. Effect of age on outcome of secondary dengue 2 infections. Int. J. Infect. Dis. 6,
118–124 with permission.
Dengue 245

Table 5 Chronic diseases as risk factors for DHF/DSS Viral Factors


The pathogenic potential of distinct dengue virus serotypes and
DHF DHF genotypes have been investigated. In secondary infections,
epidemic, epidemic DHF epidemic
a gradient of severity has been described for the four serotypes
1981a 1997 2001–02
Fatal cases DSS Fatal cases DSS
(from the most to least severe): DENV-2, DENV-3, DENV-1,
Children Adults Children Adults Adults and DENV-4. The sequence of the infecting viruses is also asso-
ciated with severity. Severity during a secondary infection has
Bronchial 23% 13% 25% 8.3% 22.2% been related to antecedent primary DENV-1 infection.
asthma Secondary DENV-2 infection has been associated with a greater
Sickle cell 6% 17% – 8.3% 11.1% pleural effusion index and more severe hemorrhagic manifesta-
anemia tions. A higher risk of DHF/DSS has been linked to sequential
a
Significantly different compared to general population. DENV-1 then DENV-3, as opposed to DENV-2 then DENV-3
Data from Bravo et al., 1987. Trans. R. Soc. Trop. Med. Hyg. 81, 816–820; Valdes infections. In addition, Asian genotypes of both DENV-2 and
et al., 1999. Pan Am. J. Public Health 6, 16–24; Gonzalez, D., Casdtro, O.E., Kouri, DENV-3 seem to be more virulent than those found in the
G., et al., 2005. Classical dengue hemorrhagic fever resulting from two dengue
infections spaced 20 years or more apart. Havana, Dengue 3 epidemic, 2001–2002. Americas, having been associated with DHF outbreaks.
Int. J. Infect. Dis. 9, 280–285. Full-length sequencing of the DENV-2 Asian and American
genotypes expose several nucleotide differences, specifically of
Some studies suggest that moderate to severe malnutrition the E gene and at the 50 and 30 UTRs. Amino acid differences in
reduces the risk of DHF/DSS while others suggest that under- the prM and E proteins have also been determined and corre-
and overnutrition DHF patients have either a greater risk of lated with virulence and particularly with the higher replicative
shock or unusual presentations and complications (Thisyakorn ability of some genotypes in human dendritic cells (DC) and
and Nimmannitya, 1993; Nguyen et al., 2005; Kalayanarooj macrophages, and in the efficiency to infect and disseminate
and Nimmannitya, 2005). in Ae. aegypti. These data support the hypothesis that differ-
Asian individuals are highly susceptible to DHF. During ences among genotypes exist, with possible implications for
the DHF epidemics of 1981, 1997 and 2001–02 in Cuba, pathogenesis and transmission.
hospitalization rates for Blacks were consistently lower than Furthermore, mutant variants of dengue viruses can escape
that for Whites; however, dengue infection rates were the antibody neutralization, and also seem able to contribute to
same. In agreement with these epidemiological observations, the complexity of the DHF phenomenon.
Sierra et al. (2006) found a reduced risk of DHF in Blacks The association of genetic variation and disease severity in an
compared to Whites. These differences in susceptibility to individual is not well documented. Recent studies have been
DHF/DSS among racial groups coincide with epidemiolog- directed to determining the viral population structure in both
ical observations in African and Black Caribbean mosquitoes and patients. Although the sequences of the major
populations. variants are the same, the extent of sequence variation in
Bronchial asthma, sickle cell anemia, and diabetes melli- mosquitoes is lower than in patients. In addition, intra-host vari-
tus have been associated with a higher risk of DHF/DSS ation in patients is apparently lower than expected (Lourenco
(Table 5). A chronic medical condition has been observed and Recker, 2010). Further studies on intra-host virus popula-
in 70% of fatal adult cases in Puerto Rico (Rigau-Perez and tion and its implication on disease severity are needed.
Laufer, 2006). A case–control study in adult patients showed
that diabetes mellitus was associated with increased risk of
DHF. In addition, individuals with diabetes mellitus and Kinetics of Dengue Infection
hypertension had a higher risk of developing DHF compared
with individuals with no diabetes and no hypertension Dengue disease begins 5–7 days following the bite from an
(Pang et al., 2012). infected mosquito. After subcutaneous injection of the virus
Genetic factors have been associated with protection against into the skin, released viral particles infect neighboring cells,
or susceptibility to DHF/DSS. These factors include allelic vari- mainly resident skin immature dendritic cells (DCs) and Lang-
ants of genes that encode cellular receptors (DC-SIGN, FcgRIIA, erhans cells, which act as sentinels, sensing the antigenic micro-
vitamin D receptor, mannose-binding lectin, G6PD deficiency, environment and capturing antigens. Infected DCs in the dermis
TNFa, IL-10, TGF-b1, DC-SIGN (CD209) promoter), migrate to the paracortex of regional draining lymph nodes,
molecules involved in antigen recognition (HLA I and II where the virus localizes and replicates primarily likewise in cells
molecules), and cytokines (TNFa) among others (Lan and such as monocytes and macrophages. Later, replication also
Hirayama, 2011; Perez et al., 2010; Garcia et al., 2010). Also occurs in other target organs, like the liver, spleen, or brain.
alleles of MHC class I polypeptide-related sequence A The virus is then released from those tissues and spreads through
(MICA) and MHC class I polypeptide-related sequence B the blood to infect white blood cells and other lymphatic tissues.
(MICB) associated with symptomatic versus asymptomatic DENV genome and protein antigens are found mainly in phago-
infection as well as MICB and PLCE1 alleles associated with cytic cells of lymph node, spleen, lung, in perivascular cells of
susceptibility to DSS have been identified (Garcia et al., brain, in hepatocytes, and in endothelial cells in spleen. Viremia
2011; Khor et al., 2011). How many genes contribute to in peripheral blood can be detectable 24–48 h before the onset
dengue susceptibility, how they interact to cause severe of clinical symptoms, peaks during the early days (first 2–3 days)
illness, and how the pathogenesis mechanisms might be of acute illness and then falls suddenly. Symptoms of illness can
genetically predicted remain unknown. last 2–7 days. Higher levels of viremia have been detected in
246 Dengue

Dengue virus skin Spreading of Immune response Disease outcome


inoculaon infecon to ssues
IgM / IgG Viral
antibodies clearance and
recovery

Complement Bening evoluon

Complicated evoluon

CD4+ T cells
CD8+ T cells Immune-
1st 2nd pathogenesis
viremia viremia Y

Y
Effects on vascular endothelium
Cytokines and
chemokines
releasing

Plasma leakage/hemorrage

Incubation period Acute phase Recovery

Figure 16 Kinetic of dengue infection.

patients with severe disease compared to uncomplicated Table 6 Pathological findings observed in severe dengue illness
patients. Immune response seems to be required for the resolu-
Tissue organ Pathology
tion of a DENV infection; however, as severity is associated with
secondary infections and the critical phase of illness appears Skin Swelling endothelial cells, perivascular edema,
when viremia has declined, immune mechanisms have been infiltration of mononuclear cells
involved in the pathogenesis of the severe clinical picture Lymphoid organs Proliferation of reticulo-endothelial cells in the
(Figure 16). spleen and lymph nodes
Table 6 shows some of the pathological findings observed Liver Degeneration with focal necrosis of hepatic
in distinct tissues and organs observed in fatal DHF cases. cells and hyaline necrosis in Kupffer cells
In addition to the particular serotype, genotype, and dengue Kidney A mild immunocomplex-type
strain, specific viral proteins play an important role in dengue glomerulonephritis
Myocardium Left ventricle thick and hard, and the chamber
pathogenesis (Ye et al., 2013; Table 7).
is not filled to the full volume
Lungs Thickening of interstitial septa
Immune Response against DENV Infection Brain and spinal cord Perivascular edema
Bone marrow Depression of hematopoietic cells is observed
Innate Immunity during febrile phase
Vessel walls Deposition of serum complement,
The mechanisms of innate immunity provide early defense
immunoglobulin, and fibrinogen
against DENV infection. The principal components of innate Mucosa of Hemorrhage occurs in the subcutaneous
immunity are cells such as DCs, phagocytic cells (monocytes gastrointestinal tract tissues
and macrophages) and natural killer (NK) cells, and soluble Pleural and abdominal Serous effusion with high protein content
mediators as blood proteins, comprising members of the cavities (mostly albumin)
complement system and other pro-inflammatory mediators Adrenal glands Depletion of lipid in the cortex
and cytokines that regulate and coordinate most of the func-
tions of the cells of innate immunity.
DCs are target cells in the skin following a bite by a dengue-
infected mosquito. They may also facilitate the spread of the IRF transcription factor. This factor stimulates type I interferon
virus within the host. These cells express receptors which ensure (IFN I) gene expression, and NF-kB transcription factor that
that the innate immune system can respond to the presence of stimulate the expression of genes encoding molecules required
genome or proteins of the dengue virus. In a dengue infection, for inflammatory responses including some mediators like
the TLR3 in the endosomal membrane and RIG-I and MDA5 inflammatory cytokines and chemokines. IFN I functions to
in the cytoplasm are sensors of double-stranded viral RNA that inhibit viral replication in both infected and uninfected cells
activate signal transduction events which, in turn, activate the by inducing an ‘antiviral state’ (Costa et al., 2013).
Dengue 247

Table 7 Role of viral proteins in the pathogenesis of dengue MHC molecules, whereas helper CD4þ T lymphocytes
infection recognize exogenously processed viral peptides presented by
class II MHC molecules (Weiskopf and Sette, 2014).
Viral protein Role in the pathogenesis
Once CD4þ T cells are activated, they proliferate and differen-
E Target of ADE-mediating antibodies tiate to a Th1 pattern, releasing important cytokines for the
Mimicry with plasminogen control of the infection such as IFNg, IL-2, and TNFa. Higher
prM Target of ADE-mediating antibodies frequencies of DENV-specific IFNg-producing T cells are
Apoptosis induction in infected cells observed in children who subsequently develop a subclinical
Generates cross-reacting antibodies with endothelial cells infection, compared with those who develop a symptomatic
NS1 Massive complement activation and vascular leakage; secondary DENV infection. Full differentiation of CD8þ CTLs
apoptosis of endothelial cells
often requires cytokines produced by CD4þ helper cells. They
Share common antibodies epitopes with human blood
undergo massive proliferation during infection recognizing
clotting, integrin/adhesin proteins expressed on
platelets and endothelial cells a few viral peptides. Effectors’ CTLs kill infected cells by necrosis
Binds to C4b and modulates complement activation or apoptosis. They can activate nucleases within infected cells
Apoptosis induction in hepatocytes and endothelial cells that degrade viral genomes and also produce cytokines, having
NS2A Blockade IFN-mediated signal transduction a pivotal role in the control of the viral infection.
Autophagia induction Virus or virus-infected cells stimulate B lymphocytes to
NS2B Apoptosis induction in infected cells produce antibodies. B cells from lymph nodes and spleen are
NS3 Apoptosis induction in infected cells activated and differentiated to plasmablasts and plasma cells
NS4A Blockade IFN-mediated signal transduction to secrete antibodies. The plasmablast numbers peak consis-
NS4B Potent inhibitor of IFN signaling by decreasing STAT1
tently at day 6 or 7 and drop to baseline level within 2–3 weeks
phosphorylation
after the onset of disease. The first isotype of antibodies
NS5 Induces transcription and translation of IL-8
Blockade IFN-mediated signal transduction detected during a dengue infection is IgM, 5 days after the onset
of clinical signs of illness. A role for IgM antibodies in the
control of a DENV infection through complement fixation
and/or favoring virus uptake by phagocytic cells has been sug-
gested. Thus, the viremia reduction most likely is due to virion
Local inflammatory response in the skin stimulates the clearance by IgM antibodies.
recruitment of leukocytes from the vasculature, including NK The neutralization capacity of antibodies in the serum is
cells. Due to its ability to rapidly limit viral dissemination, broadly cross-reactive against the four DENV serotypes, both
NK cells kill infected cells and seem to be a key early mecha- during the acute phase of illness and at 3 months post-onset
nism of immunity against DENV infection, before an adaptive of symptoms.
immune response has developed (Beltrán and López-Vergès, Antibodies protect the host through the neutralization of
2014). Activated DCs and monocytes produce IFNa, IL-12, viruses, lysing infected cells, and opsonizating viral particles,
IL-15, and IL-18, the major cytokines that stimulate NK func- which facilitate phagocytosis by monocytes and macrophages
tion enhancing their cytotoxic activity. DCs are also considered and antigen clearance. Most DENV-specific antibodies
the most important antigen-presenting cells (APCs) after they generated in humans are serotype-cross-reactive, weakly
migrate to the draining lymph nodes where they present viral neutralizing, and directed against the immature prM, and E
antigens to naive T cells. and NS1 proteins.
Neutralizing antibodies seems to be the main correlate of
protection. Antibody-mediated neutralization of viral infec-
Adaptive Immunity
tivity may occur at two levels: at the level of cell binding,
DENV-specific B and T cells are generated about 6 days through the inhibition of the interaction between the virus
postinfection, and they help to completely control the with cell-surface receptors and at the level of viral fusion,
infection. DCs in the draining lymph nodes present viral through binding of antibodies to the E protein fusion loop,
antigens to naive T cells, which results in the proliferation or blocking the conformational changes of the E protein that
and in the differentiation of specific T cells into effector cells, are required for membrane fusion. However, after a primary
triggering the adaptive cellular immune response. T cells infection, an individual is vulnerable to new infections with
recognize a complex of a particular viral epitope presented by other serotypes. Cross-immune protection is observed after
a specific MHC molecule. MHC molecules are extremely a sequential infection, with a different serotype occurring
polymorphic, with several 1000 variants known in humans, shortly after the primary infection (some weeks to few months)
implying a wide diversity of peptides that could be presented (Friberg et al., 2012).
to T cells. More than 350 antigenic regions on DENV proteins
identified in humans have been described in the literature,
Sequential Infection: Secondary Immune Response
most of them restricted by HLA MHC class I alleles. CD4þ
T cells recognize epitopes from C, E, and NS3 proteins, Upon a secondary, heterologous, DENV infection, preexisting B
whereas the NS3, NS4B, and NS5 proteins are the dominant memory cells are activated to rapidly produce antibodies, but
targets for CD8þ T cells. Cytotoxic T lymphocytes (CTLs) are these are predominantly directed against the viral serotype of
predominantly CD8þ T cells and recognize cytosolic, usually the primary infection, a phenomenon known as ‘original anti-
endogenously synthesized, viral peptides presented by class I genic sin’ (Figure 17). Massive expansion of memory B cells
248 Dengue

Figure 17 Original antigenic sin phenomenon.

IgG

IgG

Viremia
NS1 Viremia
NS1
IgM
IgM
IgA IgE IgA IgE

Primary infection Secondary infection

Figure 18 Kinetic of the antibody response against dengue virus sequential infection.

occurs during days 4–6 of symptoms and the magnitude of the containing the fusion loop, including epitopes present only
plasmablast response is higher during a secondary infection as on the intact fully assembled viral particle.
well as in severe dengue cases (Smith et al., 2014). Additionally, antibodies recognize viral antigens on the surface
The titer of DENV-specific antibodies in serum rapidly of infected cells, killing them through complement-mediated
increases within days of the onset of fever and will bind to the cytotoxicity (CMC) or by using effector cells as NK cells and
heterologous serotype; they have less neutralizing capacity macrophages, via antibody-dependent cell cytotoxicity (ADCC).
against the new infecting serotype than to the serotype of the Both mechanisms could be of importance in the elimination of
primary infection. Although there is detectable IgM production, the infected cells in a secondary infection. The antibodies that
IgG1 and IgG3 serum antibodies are the predominant mediate CMC and ADCC recognize viral proteins expressed on
immunoglobulins throughout the course of illness. the plasma membrane of infected cells, presumably E, prM, and
Neutralization of virion infectivity by antibodies binding NS1 proteins could be their main targets. Moreover, IgM and
to the extracellular virion and blocking its interaction with IgG bind viral-soluble antigens forming immune complexes
the host target cells represents the major antibody function and act as opsonins favoring antigen removal by the phagocytic
of protection during a secondary infection. Serotype-specific cells. The kinetic of the different classes of antiviral-specific anti-
neutralizing antibodies target diverse regions of the E protein, bodies after a primary or a secondary dengue infection are shown
but mainly recognize a region in the E protein domain I/II in Figure 18.
Dengue 249

Homologous Heterologous Y Homologous Ab


secondary infection secondary infection Y Heterologous Ab
FcγR
Y Y Y Virus nucleocapsid

Y
Y Y Y

Y
Y Y Virus
Y

Y
Y Y Viral proteins
Y

Y
Cell proteolitic enzyme
Y

Y Y Y

Y Y

Y
Y

Monocyte/macrophage

Neutralization
Y Antibody-dependent
Y Y Y Y enhancement

Y
Y Y
Y

Y
Y

Protection Increase of viral load and spread of infection

Figure 19 Role of antibody during homologous and heterologous secondary infection.

It has been proposed that cross-reactive T cells raised against Table 8 Immune effector mechanisms against dengue infection
the original infecting serotype (primary infection) also domi-
Classes or
nate during a secondary heterologous infection, being impli-
Effector subpopulation Mechanism Target protein
cated in the ‘original antigenic sin’ phenomenon (Figure 19).
The expansion of preexisting lower avidity cross-reactive Antibodies IgM, IgG Virus neutralization E, prM
memory T cells dominates the responses over the naive IgG Antibody-dependent ?? E, prM, NS1
T cells that show a higher avidity for the new DENV serotype. cell cytotoxicity
It is further hypothesized that peptide variants derived from IgM, IgG Complement-mediated ?? E, prM, NS1
the secondary infecting serotype can induce a response that is cytotoxicity
qualitatively different from the response induced by the orig- T cells CD4þ Cytotoxicity NS3, E, prM, C
CD8þ Cytotoxicity NS3, C, NS1,
inal antigen (primary infecting serotype), such as inducing
NS4b and NS5
a different pattern of cytokine production. However, high titers
of cross-reacting neutralizing antibodies could protect those
individuals who are exposed to a heterotypic secondary infec-
tion. Very early blockade of the inoculated virus and control interaction of the virus–antibody complex with Fc receptors.
of the viral infection may serve to prevent development of This phenomenon, called ADE, was originally proposed by Hal-
the disease or modulate or progression to severe outcomes. stead as an important pathogenic mechanism in DHF/
The most important effector mechanisms and their viral targets DSS (Figure 19).
are shown in Table 8. ADE increases the efficiency of the virus infection and favors
the early spread of the infection, thus increasing the number of
DENV-infected cells. Cell infection via ADE appears to remodel
Immunopathogenesis of Dengue
and to suppress the innate immune response with increasing
Severe disease has been associated with a heterologous secondary virus particle production in infected cells. This phenomenon
infection as the most important human risk factor. Preexisting called intrinsic ADE can occur along both TLR-dependent and
antibodies from a previous DENV infection play an important -independent pathways. It seems to suppress IFN I-mediated
role in this predisposition. When non-neutralizing properties antiviral responses. The suppression of the innate immune
against the new serotype predominate in these antibodies, system during ADE likely facilitates longer survival of infected
dengue virus infection of macrophages may be enhanced. The cells with the possible increase of virus output. In addition,
enhancement of the infection is facilitated through an efficient other molecules produced by infected cells may induce
250 Dengue

a pathological host response. For example, C-type lectin a disruption of the intestinal barrier inducing microbial trans-
domain family 5 member A (CLEC5A), which directly location. This has been associated with extensive immune acti-
interacts with the dengue virion, results in the stimulation of vation that could play a role in dengue pathogenesis. On the
the release of pro-inflammatory cytokines (Costa et al., 2013). other hand, apoptosis is observed with impairment of CD8þ
Higher viremia titers in severe illness compared to that in T-cell cytokine production, decreased circulating of CD4þ
mild disease have been observed. In general, immune and CD8þ T cell counts, impairment of T cell proliferation,
responses are essential for the resolution of a DENV infection; and increased T cell apoptosis.
however, as severe dengue is associated with secondary infec- NS1 protein–antibody complexes and several cytokines also
tion and the complications emerge at the time when viremia is activate the release of complement activation products such as
declining, severity has been explained as a consequence of C3a and C5a that have a direct effect on the vascular perme-
immunopathology. The activation of different immune func- ability. Other soluble mediators such as PAF, prostaglandins,
tions and the duration and magnitude of the immune and stress oxidative metabolites may also be involved in severe
response depend on how the virus interacts with the host disease. Molecular mimicry has also been associated with
target cells and on how much the virus spreads. The more viral platelet and endothelial injury. Antibodies elicited by NS1,
antigens are present in different parts of the body, depending prM, or E proteins recognized in a cross-reactive fashion host
on the virus spreading in the early phases of the infection, the ‘self’ epitopes contained in blood-clotting elements, plasmin-
more mechanisms of the host immune response will be ogen, or proteins expressed on platelets and endothelial cells.
involved in stopping and clearing the infection. These mech- These antibodies could mediate the decrease of the circulating
anisms depend on the kind of infection (primary, secondary, levels of these coagulation factors, as well as cytotoxicity or
tertiary, or quaternary), the infecting serotype and the apoptosis of cells (Wan et al., 2013). Table 9 shows the main
sequence of infection. mechanisms associated with vascular leakage, liver damage,
During a secondary infection, the expansion of preexisting leukopenia, and hemorrhagic manifestations during a severe
lower avidity cross-reactive memory T cells dominates the dengue infection.
responses over the naive T cells, and peptide variants derived In summary, the presence of enhancing antibodies
from the second serotype seems to induce a response qualita- combined with a misbalanced T cell response and deficient
tively different from the response induced by the original control of the infection contribute to the immunopathogenesis
antigen, such as inducing a different pattern of cytokine of dengue and development of severe disease in an individual
production. These altered T cell responses may contribute to with a susceptible genetic background.
the ‘cytokine storm’ observed during a heterologous secondary
infection, and thus to the immunopathogenesis of DHF/DSS.
Dengue Diagnosis
Different HLA alleles are associated with differential magnitude
of anti-DENV responses; some of them are associated with an Dengue diagnosis is important for clinical care, surveillance
increased risk of severe illness and a weaker CD8þ response support, pathogenesis studies, vaccine and drug development,
(St. John et al., 2013). and clinical trials. Dengue laboratory diagnosis can be per-
The absence of structural damage in the vasculature found formed through virus isolation, genome and antigen detection,
in autopsy studies suggests that circulating factors are primarily and serology, with the last the most widely applied in routine
responsible for the existence of microvascular permeability diagnosis (Figure 20). Assay choice depends on the timing of
during a DENV infection. Therefore, soluble mediators sample collection and the purpose of testing (Guzman et al.,
produced by leukocytes in response to the infection represent 2014; Guzman et al., 2010a).
crucial mediators to enhance plasma extravasation seen in
severe cases. Consequently, cytokines and other inflammatory Virus Isolation
mediators act on the endothelium, potentially altering normal Viremia is present 2–3 days before, and then for 3–4 days after
functions of endothelial cells. Although elevated levels of the onset of fever. The decrease in viremia coincides with the
pro-inflammatory cytokines and chemokines occur in patients appearance of specific IgM antibodies and the remission of
with DF, most of them are found at higher levels in patients fever. Dengue viruses can be isolated in serum collected in
experiencing severe disease. Key inflammatory cytokines the acute phase of illness and preferably before fever disap-
involved in the massive increase of vascular permeability pears. Viruses can also be isolated from tissues (liver, spleen,
include TNFa, IL-6, IFNg, IL-8, and ligand (CCL)-2, CCL-3, lymph nodes, lung, and thymus) obtained at necropsy.
CXCL-8, CXCL-10, which activate macrophages or nearby Mosquito cell lines (Ae. albopictus C6/36 and Ae. pseudoscu-
vascular endothelium to increase the expression of adhesion tellaris AP61) are the cell culture systems of choice for dengue
molecules that promote leukocyte and plasma extravasations. virus isolation. Mammalian cell cultures such as Vero cells,
TNFa and IL-10 could also be involved in the hepatic dysfunc- LLCMK2 cells, and others have been employed with less
tion, hypotension, thrombocytopenia, and hemorrhagic mani- efficiency.
festations. Besides the effects of cytokines and chemokines, Direct mosquito inoculation improves the sensitivity of
leading to increased vascular permeability, it was also demon- virus detection; however, insectaria facilities and technical skill
strated that overproduction of soluble matrix metalloprotei- are required. The intracerebral inoculation of suckling mice is
nase (MMP) 9 and 2 and HMGB1 produced by infected cells the oldest and least sensitive method for isolating virus, and
increases endothelial permeability (Sun and Kochel, 2013). is only used when no other method is available. Inoculated
Alternatively, elevated LPS levels detected during a DENV infec- mice develop encephalitis. Figure 21 shows the biological
tion in correlation with disease severity may suggest systems for dengue virus isolation.
Dengue 251

Table 9 Associated mechanisms to the main signs of severe disease

Phenomenon Associated mechanisms

Plasma leakage Endothelial cell apoptosis via production of nitric oxide or inflammatory activation
induced by antibodies anti-NS1
Immune recognition of infected endothelial cells
Dysfunction of endothelial cells caused by high levels of TNFa and IL-8
Complement activation and systemic generation of anaphylatoxins and SC5b-9 that
increase vascular permeability
Direct infection of endothelial cells modulates differently the cell surface expression of
molecules critically involved in the interactions between endothelial and inflammatory
cells
Changes in surface and soluble VEGFR2 expression induced by DENV, which regulates
vascular permeability via binding VEGF
Liver damage Hepatocyte infection
Apoptosis of hepatocytes induced by DENV infection
Immune cytotoxicity
Leukopenia Bone marrow suppression
Apoptosis of infected cells
Apoptosis of leukocytes by immune mechanisms
Hemorrhagic manifestations Thrombocytopenia Infection of platelets
Consumption of platelets
Cross-reactive anti-NS1 and anti-prM antibodies
Platelet dysfunction
High levels of IL-10 and soluble TNF receptor-II
Deficiency in coagulation Cross-reactive anti-E antibodies that recognize
plasminogen
Cross-reactive anti-NS1 antibodies recognize fibrinogen
Fibrinolysis activation induced by DENV infection, which
degrades fibrinogen directly and causes secondary
activation of pro-coagulant homeostatic mechanisms
High levels of IL-6 associated with elevated levels of tPA
and a deficiency in coagulation
Marked hepatic dysfunction

Opportunity

Direct methods Indirect methods

Virus isolaon Genome Angen Serology Serology


detecon detecon IgM IgG

Confidence

Figure 20 Laboratory diagnostic methods employed for diagnosis of a dengue infection. Guzman et al., 2011. Nat. Rev.
252 Dengue

Cytopathic effect in mosquito cells Mosquito inoculation Sick newborn mice


inoculation

Figure 21 Main systems for viral isolation.

The immunofluorescence assay with serotype-specific Genome Detection


monoclonal anti-dengue antibodies on squashed mosquito Dengue virus RNA can be detected by PCR directly from clinical
heads, infected cells, or brain tissues from inoculated mice is samples such as serum, plasma, and autopsy-fresh and
the choice method for virus identification; however, PCR, paraffin-embedded tissues, and from the supernatant of
ELISA, and immunohistochemical techniques can also be dengue virus–infected mosquito cell cultures and infected
employed. adult mosquito and mosquito larvae. DNA amplification is
preceded by a reverse transcription (RT) reaction, producing
Antigen Detection cDNA from the target RNA.
Secreted NS1 glycoprotein is a diagnostic marker during the Because of its high sensitivity, PCR allows the detection of
early phase of infection. Its detection in sera may be a valuable concurrent infections by multiple serotypes, detection of
surrogate marker of viremia and may also serve as a prognostic dengue in stored samples over long periods, and detection in
marker of disease progression. NS1 detection is influenced by mosquito pools (groups of captured mosquitoes tested
the day of illness, kind of infection (primary or secondary together), favoring its application in entomological surveil-
infection), and infecting virus. At present, several commercial lance (Figure 23). PCR in conjunction with nucleotide
kits for NS1 detection, both as ELISA and rapid test formats, sequencing or restriction enzyme analysis has allowed the
are available which have different sensitivity and specificity study of genetic strain variability in order to identify the origin
profiles. NS1 glycoprotein can also be detected in urine of epidemics and reveal markers of virulence.
collected from febrile patients, tissues collected from fatal The introduction of real-time PCR is supplanting conven-
cases and in infected mosquitoes (Guzman et al., 2010b; tional PCR, allowing the rapid detection (in less than 2 h)
Duong et al., 2011; Hunsperger et al., 2014). Sensitivity of and the quantification of minimum copies of the RNA.
diagnosis increases when both markers, NS1 and IgM, are Currently, many RT-PCR and real-time RT-PCR protocols are
studied during the first 4 days of acute illness (Guzman available. A recent CDC RT-PCR assay has been produced
et al., 2010b; Figure 22). Immunohistochemical techniques that enables dengue diagnosis in the first 7 days of illness
have been shown to be useful for dengue antigen detection (Santiago et al., 2013). Protocols for RT-PCR and real-time
in formalin-fixed paraffin-embedded tissue samples from RT-PCR for multiplex detection of several arboviruses and
fatal cases. hemorrhagic fever viruses are also available.

D2 D3 D4 D1 MW

Figure 22 NS1 and IgM dengue detection by day of illness. Figure 23 Standard RT-PCR and nested PCR for dengue RNA
Increasing in diagnostic sensitivity. Data collected from Guzman et al., detection as described. Lanciotti et al., 1992. J. Clin. Microbiol. 30,
2010. PLoS NTD. 545–551; Photo kindly supplied by D. Rosario, IPK, Habana.
Dengue 253

Serological Diagnosis Table 10 Laboratory criteria of dengue infection


During primary dengue infection, anti-dengue IgM is detected
5–6 days after onset of fever, and then persists for 30– Criteria
60 days. In some individuals, IgM may persist for more than Confirmed dengue infection Isolation of dengue virus from serum or
170 days. IgG levels slowly rise, are detectable by the 9th to autopsy samples
10th day after symptom onset, and then persist for life. IgG or IgM seroconversion or
Unlike a primary infection, a secondary infection with demonstration of a fourfold or greater
a heterologous serotype is characterized by a rapid (1 or change in reciprocal IgG titers in
2 days after fever onset), high, and cross-reactive IgG response. paired serum samples
In some cases, an IgM response is not detectable. Demonstration of dengue antigen in
Specific IgA and IgE antibodies also develop during both serum or autopsy tissue
Demonstration of positive PCR/real-time
primary and secondary infections. The IgA is broadly
PCR in serum or autopsy tissue
cross-reactive to the four serotypes, and the level of IgE has Highly suggestive or probable Positive IgM antibody test on a single
been related to disease severity. Anti-dengue IgA and IgE may dengue infection late acute- or convalescent-phase
complement dengue diagnosis as possible markers of recent serum to dengue antigen
infection. Reciprocal HI antibody titer 1280 or an
The detection of anti-dengue IgM is considered the best indi- equivalent IgG ELISA titer
cator of an active or recent infection. MAC-ELISA (IgM
antibody–capture ELISA) diagnosis represents one of the most
important advances in dengue diagnosis, being an invaluable
tool for routine diagnosis and surveillance. Serum, blood on Control and Prevention
filter paper, and saliva are useful for IgM detection if samples
Vector Control
are taken during the appropriate time period, around the 5th
day after the onset of fever. Different commercial kits for Vector surveillance provides important evidence for the presence
anti-dengue IgM and IgG detection are commercially available, and density of vector populations including vector resistance to
with variable sensitivity and specificity (Hunsperger et al., 2009). insecticides. It also targets the evaluation and analysis of the
The presence of anti-dengue IgG in serum is a criterion for the impact of control programs (Kay, 1999). Sampling surveys
diagnosis of a past infection. However the presence of high titers include larval, adult, standard and sticky ovitrap surveys.
of IgG in an acute serum sample, or seroconversion or fourfold Indoor fogging (intradomiciliary) or out fogging in the
increase in paired sera from a suspected dengue case are criteria streets by airplane, trucks, or other types of equipment
for a recent or confirmed dengue infection, respectively. ELISA (thermo-nebulization or ultra-low volume) using different
has gained acceptance as a faster and more convenient alternative insecticides especially pyrethroids, are the most important
to determine IgG antibodies, although hemagglutination inhibi- way up to now for the control of Ae. aegypti (Esu et al.,
tion (HI) assay is still considered the gold standard technique. 2010). However, the use of airplanes or other methods of
Due to the presence of cross-reactive antigens shared by flavi- out-fogging in the urban ecosystem is not efficient for species
viruses, specific diagnosis is not possible in most occasions. that live inside houses and its use must be analyzed according
When a serological specific diagnosis is required, neutralization to the conditions and the possible environmental damage. The
assay and particularly plaque reduction neutralization test targets are the infected mosquito females that maintain the
(PRNT) is employed. Neutralizing antibodies inhibit dengue virus in circulation. Biological control methods, such as Bacillus
virus infection and provide greater specificity; however, neutral- thuringiensis var. israelensis, are used to reduce vector larvae
ization assay is not recommended for routine diagnosis, as it is (Boyce et al., 2013). In recent years, the use of biological agents
labor intensive and time consuming. In addition, reproducibility such as Wolbachia (family Rickettsiaceae), a bacterium that
among laboratories is low, as several factors such as cell line, affects the metabolism and capacity of transmission of DENV
virus strain, and expression of receptors and attachment factors in Ae. aegypti mosquitoes has caught the attention of many
that complement virus propagation cell line can contribute to researchers. This bacterium was found in naturally infected
this heterogeneity. Nonetheless, this assay is recommended Ae. albopictus mosquitoes but the possibility of obtaining
when a different flavivirus is suspected (Guzman et al., 2014). strains of Wolbachia-infected Ae. aegypti and its propagation in
Table 10 shows the laboratory criteria of dengue infection. nature would be an alternative method (McMeniman et al.,
The identification of prognosis markers for DENV disease 2009). On the other hand, the use of transgenic mosquitoes
severity is currently a research priority. Besides clinical to compete with normal mosquitoes in nature is a new method
warning signs recommended by the WHO (WHO/TDR, and could be a successful strategy for the control of Ae. aegypti
2009), other studies are conducted to identify some populations in some settings. Some experiments have showed
biomarkers for disease severity. Higher levels of viremia, good results in a short time (Alphey, 2008).
greater consumption of von Willebrand factor and its cleaving The use of larvicides such as temephos, an organofosforade,
enzyme ADAMTS-13, elevated levels of cell-free circulating is changed by insect growth regulators as piryproxifen with very
DNA, and the increased expression of mitochondrial ribo- low toxicity (WHO, 2003).
somal proteins in DSS patients have been associated with However, the main problem of Ae. aegypti control is its
severe disease (Ha et al., 2011; Djamiatun et al., 2012). A ecology, linked to humans. If people refuse the use of any
dengue pathogenesis assay may become a reality in the near control method, they will fail and if the environmental condi-
future (Loke et al., 2010). tions and water distribution are inadequate, the mosquito can
254 Dengue

find breeding sites in houses and other dwellings because of the complete long-lasting protection against the four serotypes
existence of containers of water. in order to avoid any enhancement of dengue illness after
In summary, Ae. aegypti control has different components, subsequent infection. These requirements, in addition to the
where the use of insecticides must be accomplished with effi- lack of an animal model and our elementary knowledge of
cient public service of garbage, residual water, distribution of disease pathogenesis, have negatively influenced dengue
water, and citizen responsibility. vaccine development.
Conventional live-attenuated and inactivated vaccines,
subunit vaccines based on recombinant strategy, chimeric,
Education and Community Participation
and DNA vaccines are among the main dengue vaccine strate-
Sanitary public health education leading to low mosquito pop- gies (Thomas, 2014). Figure 24 shows the chimeric dengue
ulations through cleanup campaigns with significant public vaccine candidates.
involvement and action is a necessary step. The public deserves DENV themselves have been employed as backbones for
to know the risks and how to reduce them. chimeric vaccines. In addition to chimeric vaccine candidates,
The community must be involved in the prevention, surveil- dengue genes of structural and nonstructural proteins, particu-
lance, and control of the vector to ensure program sustain- larly the E gene including its domain III, have been expressed in
ability. Communities with effective vector control integration systems such as E. coli bacteria, baculovirus, yeast, and
are cleaner, and do not have the large mosquito populations Drosophila flies. Candidates have been evaluated both in mice
that communities with reproduction of the mosquito in and monkeys, showing variable grades of immunogenicity
containers do. and protection.
In each country, the maximum authority must organize Several candidates are or have been in stages of human
a control program with fully integrated activities into the estab- testing. The Sanofi Pasteur tetravalent live–attenuated
lished administrative structures, mass social organizations, chimeric dengue/yellow fever vaccine, which is based on
churches, schools, business, and factories in order to make a yellow fever vaccine 17D backbone expressing the prM
a significant contribution to vector control (Kroeger et al., and E genes of each of the four DENV serotypes, has been eval-
2006). The WHO and the Pan-American Health Organization uated in Phase III efficacy trials in humans. The CYD 23 trial
(PAHO) have developed the Integrated Management Strategy showed that vaccinated Thai children developed neutralizing
for Prevention and Control of Dengue to face the challenge antibodies to the four viruses after 2 or 3 doses. However the
of the control of dengue (WHO/TDR, 2009). protective efficacy (30%), which differed by serotype was
lowest for DENV-2 (Sabchareon et al., 2012). In 2015, results
of two Phase III studies developed in children from Latin
Vaccine Development
American and Southeast Asian countries were published
Dengue vaccines have been under study since the 1940s; with a vaccine efficacy of 56.5% (Asian study) and 60.8%
however, in recent years their development has accelerated (American study). Higher protective efficacy figures were
dramatically. Constructing a dengue vaccine has been a chal- observed against dengue hospitalization, disease severity,
lenge. A potential vaccine must provide a balance between the and in dengue seropositive individuals. DENV-2 showed the
immunogenicity it evokes and attenuation of pathogenicity of lowest serotype-specific efficacy (35–42.3%), while DENV-3
the virus. The vaccine should be safe and also provide and DENV-4 showed figures around 74–78%. These studies

ChimeriVaxTM
Chimeric Dengue -Yellow fever 17D genome cDNA

5’ C prM E C NS 3’
DENV YFV genome

Chimeric recombinant DENV attenuated

NIH: Den4/Den

5’ CC prM E NS 3’
DENV 1,2 or 3 DENV 4 genome

CDC: Den2/Den

5’ C prM E NS 3’
DENV 1,2 or 3 DENV 2 genome

Figure 24 Chimeric dengue virus vaccine candidates.


Dengue 255

Table 11 Current dengue vaccine candidates in clinical stages

Phase of
Strategy Candidates development

Live-attenuated chimeric DENV-1 to DENV-4, prM and E genes inserted into the Sanofi Pasteur Phase III
vaccine prM and E regions of the yellow fever 17D vaccine
DENV-1 to DENV-4, prM and E genes inserted into the Inviragen/Takeda Phase II
prM and E regions of attenuated DENV-2
(16681, PDK 53)
DENV-1 to DENV-3 prM and E genes inserted into the NIAID Phase II
prM and E regions of DENV-4 backbone (attenuated by
deleting 30 nucleotides in the 30 UTR)
Recombinant subunit vaccines E/NS1 protein subunit expressed in Drosophila Merck Phase I
Inactivated vaccine Whole purified inactivated virus GSK/WRAIR/Biomanguinhos Phase I
DNA vaccine prM and E protein genes US Naval Medical Research Center Phase I

represent the first Phase III dengue vaccine studies (Villar Anders, K.L., Nguyet, N.M., Van Vinh Chau, N., Hung, N.T., Thuy, T.T., Lien
et al., 2015; Capeding et al., 2014) contributing new informa- Le, B., Farrar, J., Wills, B., Hien, T.T., Simmons, C.P., 2011. Epidemiological
factors associated with dengue shock syndrome and mortality in hospitalized
tion on this important topic.
dengue patients in Ho Chi Minh City, Vietnam. Am. J. Trop. Med. Hyg. 84,
WHO, through the Initiative for Vaccine Research, and the 127–134.
Dengue Vaccine Initiative in conjunction with the TDR/WHO Arunachalam, N., Tana, S., Espino, F., Kittayapong, P., Abeyewickreme, W., Thet
made joint efforts to accelerate the development and introduction Wai, K., 2010. Eco-bio-social determinants of dengue vector breeding: a multi-
of a dengue vaccine (Hombach et al., 2005). Table 11 shows country study in urban and periurban Asia. Bull. World Health Organ. 88, 173–
184. http://dx.doi.org/10.2471/BLT.09.067892.
some of the vaccine candidates and their current clinical stage. Arunachalam, N., Tana, S., Espino, F., Kittayapong, P., Abeyewickreme, W., Wai, K.T.,
Tyagi, B.K., Kroeger, A., Sommerfeldg, J., Petzold, M., 2014. Eco-bio-social
determinants of dengue vector breeding: a multicountry study in urban and peri-
Conclusion urban Asia. Ann. N.Y. Acad. Sci. http://dx.doi.org/10.1111/nyas.12413 [Epub
ahead of print].
de Andrade, S.M.O., Moreira Herkert, C.M., Da Cunha, R.V., Rodrigues, M.D., Da
Today, dengue ranks as the most important mosquito-borne Silva, B.A.K., 2014. A new approach to reducing mortality from dengue. Open J.
viral disease in the world. To address this situation, WHO Clin. Diagn. 4, 12–16.
recently published a global strategy for dengue prevention Balasubramanian, S., Ramachandran, B., Amperayani, S., 2012. Dengue viral infection
and control in the period 2012 to 2020 with the main goal in children: a perspective. Arch. Dis. Child. http://dx.doi.org/10.1136/archdischild-
2012-301710.
to reduce the disease burden and with two principal objectives, Bandyopadhyay, S., Lum, L.C., Kroeger, A., 2006. Classifying dengue: a review of the
to reduce mortality and morbidity from dengue by 2020 by at difficulties in using the WHO case classification for dengue haemorrhagic fever.
least 50% and 25%, respectively (WHO, 2012). To achieve Trop. Med. Int. Health 11, 1238–1255.
these objectives, it is important to move on from a reactive Barniol, J., Gaczkowski, R., Vega Barbato, E., Da Cunha, R.V., Salgado, D.,
Martinez, E., Soria Segarra, C., Pleites Sandoval, E.B., Mishra, A., Safitri
response to an emergency situation to proactive risk assess-
Laksono, I., Lum, L.C., Martinez, J.G., Nunez, A., Balsameda, A., Allende, I.,
ment, to develop early warning systems, and preventive Ramirez, G., Dimaano, E., Thomacheck, K., Akbar, N.A., Ooi, E.E., Villegas, E.,
measures, guided by entomological as well as epidemiological Hien, T.T., Farrar, J., Horstick, O., Kroeger, A., Jaenisch, T., 2011. Usefulness and
surveillance. Research continues playing a crucial role. In this applicability of the revised dengue case classification by disease: multi-centre study
scenario, commitments and obligations from partners, organi- in 18 countries. BMC Infect. Dis. 11, 106.
Basuki, P.S., Budiyanto, Puspitasari, D., Husada, D., Darmowandowo, W.,
zations, and countries, as well as leadership by WHO are of Ismoedijanto, Soegijanto, S., Yamanaka, A., 2010. Application of revised dengue
utmost importance (WHO, 2012). classification criteria as a severity marker of dengue viral infection in Indonesia.
Southeast Asian J. Trop. Med. Public Health 41, 1088–1094.
Beltrán, D., López-Vergès, S., 2014. NK cells during dengue disease and their
See also: Arboviruses. recognition of dengue virus-infected cells. Front. Immunol. 5, 1–6.
Bhatt, S., Gething, P.W., Brady, O.J., Messina, J.P., Farlow, A.W., Moyes, C.L.,
Drake, J.M., Brownstein, J.S., Hoen, A.G., Sankoh, O., Myers, M.F.,
George, D.B., Jaenisch, T., Wint, G.R., Simmons, C.P., Scott, T.W., Farrar, J.J.,
Hay, S.I., 2013. The global distribution and burden of dengue. Nature 496,
References 504–507.
Boyce, R., Lenhart, A., Kroeger, A., Velayuhan, R., Roberts, B., Horstick, O., 2013.
Alexander, N., Balmaseda, A., Coelho, I.C., Dimaano, E., Hien, T.T., Hung, N.T., Bacillus thuringiensis israelensis (Bti) for the control of dengue vector: systematic
Janisch, T., Kroeger, A., Lum, L.C., Martinez, E., Siqueira, J.B., Thuy, T.T., literature review. Trop. Med. Int. Health 18, 564–577.
Villalobos, I., Villegas, E., Wills, B., 2011. Multicentre prospective study on dengue Brady, O.J., Gething, P.W., Bhatt, S., Messina, J.P., Brownstein, J.S.,
classification in four South-east Asian and three Latin American countries. Trop. Hoen, A.G., Moyes, C.L., Farlow, A.W., Scott, T.W., Hay, S.I., 2012. Refining
Med. Int. Health. http://dx.doi.org/10.1111/j.1365-3156.2011.02793.x. the global spatial limits of dengue virus transmission by evidence-based
Alphey, L.B.M., 2008. Aedes aegypti control: the concomitant role of competition, consensus. PLoS Negl. Trop. Dis. 6, e1760. http://dx.doi.org/10.1371/
space and transgenic technologies. J. Appl. Ecol. 45, 1258–1265. journal.pntd.0001760.
Amarilla, A.A., De Almeida, F.T., Jorge, D.M., Alfonso, H.L., De Castro-Jorge, L.A., Brathwaite, O., San Martín, J.L., Montoya, R., Diego, J., Zambrano, B., Dayan, G.H.,
Nogueira, N.A., Figueiredo, L.T., Aquino, V.H., 2009. Genetic diversity of the E 2012. Review: the history of dengue outbreaks in the Americas. Am. J. Trop. Med.
protein of dengue type 3 virus. Virol. J. 6, 113. Hyg. 87, 584–593.
256 Dengue

Capeding, M.R., Tran, N.H., Hadinegoro, S.R., Ismail, H.I., Chotpitayasunondh, T., Gubler, D.J., Kuno, G. (Eds.), 1997. Dengue and Dengue Hemorrhagic Fever. CAB
Chua, M.N., Luong, C.Q., Rusmil, K., Wirawan, D.N., Nallusamy, R., International, Wallington, UK.
Pitisuttithum, P., Thisyakorn, U., Yoon, I.K., Van Der Vliet, D., Langevin, E., Laot, T., Gubler, D.J., 2011. Emerging vector-borne flavivirus diseases: are vaccines the
Hutagalung, Y., Frago, C., Boaz, M., Wartel, T.A., Tornieporth, N.G., Saville, M., solution? Expert Rev. Vaccines 10, 563–565.
Bouckenooghe, A., 2014. Clinical efficacy and safety of a novel tetravalent dengue Guzman, M.G., Buchy, P., Enria, D., Vazquez, S., 2014. Laboratory diagnosis of
vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, dengue. In: Gubler, D.J., Ooi, E.E., Vasudevan, S., Farrar, J. (Eds.), Dengue and
placebo-controlled trial. Lancet 384, 1358–1365. Dengue Hemorrhagic Fever, second ed. CAB International, Wallingford, UK.
Capeding, R.Z., Brion, J.D., Caponpon, M.M., Gibbons, R.V., Jarman, R.G., Guzman, M.G., Halstead, S.B., Artsob, H., Buchy, P., Farrar, J., Gubler, D.J.,
Yoon, I.K., Libraty, D.H., 2010. The incidence, characteristics, and presen- Hunsperger, E., Kroeger, A., Margolis, H.S., Martinez, E., Nathan, M.B.,
tation of dengue virus infections during infancy. Am. J. Trop. Med. Hyg. 82, Pelegrino, J.L., Simmons, C., Yoksan, S., Peeling, R.W., 2010a. Dengue:
330–336. a continuing global threat. Nat. Rev. Microbiol. 8, S7–S16.
Carrillo-Valenzo, E., Danis-Lozano, R., Velasco-Hernandez, J.X., Sanchez-Burgos, G., Guzman, M.G., Jaenisch, T., Gaczkowski, R., Ty Hang, V.T., Sekaran, S.D.,
Alpuche, C., Lopez, I., Rosales, C., Baronti, C., De Lamballerie, X., Holmes, E.C., Kroeger, A., Vazquez, S., Ruiz, D., Martinez, E., Mercado, J.C., Balmaseda, A.,
Ramos-Castaneda, J., 2010. Evolution of dengue virus in Mexico is characterized Harris, E., Dimano, E., Leano, P.S., Yoksan, S., Villegas, E., Benduzu, H.,
by frequent lineage replacement. Arch. Virol. 155, 1401–1412. Villalobos, I., Farrar, J., Simmons, C.P., 2010b. Multi-country evaluation of the
Costa, V., Fagundes, C.T., Souza, D.G., Martins, M., 2013. Inflammatory and innate sensitivity and specificity of two commercially-available NS1 ELISA assays for
immune responses in dengue infection protection versus disease induction. Am. J. dengue diagnosis. PLoS Negl. Trop. Dis. 4.
Pathol. 182, 1950–1961. http://dx.doi.org/10.1016/j.ajpath.2013.02.027. Guzman, M.G., Kouri, G., 2003. Dengue and dengue hemorrhagic fever in the
Christophers, S.R., 1960. Aedes Aegypti L. The Yellow Fever Mosquito: Its Life History, Americas: lessons and challenges. J. Clin. Virol. 27, 1–13.
Bionomics and Structure. Cambridge University Press, Cambridge, UK. Guzman, M.G., Kouri, G., Bravo, J., Valdes, L., Vazquez, S., Halstead, S.B., 2002a. Effect
Deen, J.L., Harris, E., Wills, B., Balmaseda, A., Hammond, S.N., Rocha, C., of age on outcome of secondary dengue 2 infections. Int. J. Infect. Dis. 6, 118–124.
Dung, N.M., Hung, N.T., Hien, T.T., Farrar, J.J., 2006. The WHO dengue Guzman, M.G., Kouri, G., Valdes, L., Bravo, J., Alvarez, M., Vazques, S., Delgado, I.,
classification and case definitions: time for a reassessment. Lancet 368, Halstead, S.B., 2000. Epidemiologic studies on dengue in Santiago de Cuba,
170–173. 1997. Am. J. Epidemiol. 152, 793–799. Discussion 804.
Djamiatun, K., Van Der Ven, A.J.A.M., De Groot, P.G., Faradz, S.M.H., Hapsari, D., Guzman, M.G., Kouri, G., Valdes, L., Bravo, J., Vazquez, S., Halstead, S.B., 2002b.
Dolmans, W.M.V., et al., 2012. Severe dengue is associated with comsumption of Enhanced severity of secondary dengue-2 infections: death rates in 1981 and
von Willebrand factor and its cleaveing enzyme ADAMTS-13. PLoS NTD 6, e1628. 1997 Cuban outbreaks. Rev. Panam Salud Publica 11, 223–227.
Duong, V., Ly, S., Lorn Try, P., Tuiskunen, A., Ong, S., Chroeung, N., Lundkvist, A., Ha, T., Huy, N.T., Murao, L.A., Lan, N.T.P., Thuy, T.T., Tuan, H.M., et al., 2011.
Leparc-Goffart, I., Deubel, V., Vong, S., Buchy, P., 2011. Clinical and virological Elevated levels of cell free circulating DNA in patients with acute dengue virus
factors influencing the performance of a NS1 antigen-capture assay and infection. PLoS One 6, e25969.
potential use as a marker of dengue disease severity. PLoS Negl. Trop. Dis. 5, Halstead, S., 2012. Controversies in dengue pathogenesis. Paediatr. Int. Child Health
e1244. 32, 5–9.
Duong, V., Lambrechts, Louis, Paul, R.E., Lye, S., Laya, R.S., Long, K.C., Huyg, R., Hang, V., Holmes, D.C., Veasna, D., Quy, N.T., Hien, T.T., Quail, M., Churcher, C.,
Tarantola, A., Scott, T.W., Sakuntabhai, A., Buchya, P., 2015. Asymptomatic Parkhill, J., Cardosa, J., Farrar, J., Wills, B., Lennon, N.J., Birren, B.W., Buchy, P.,
humans transmit dengue virus to mosquitoes. PNAS Early Ed. www.pnas.org/cgi/ Henn, M.R., Simmons, C., 2010. Emergence of the Asian 1 genotype of dengue
doi/10.1073/pnas.1508114112. virus serotype 2 in Viet Nam: in vitro fitness advantage and lineage replacement in
Esu, E., Lenhart, A., Smith, L., Horstick, O., 2010. Effectiveness of peridomestic space South East Asia. PLoS NTD 4, 1–11.
spraying with insecticide on dengue transmission; systematic review. Trop. Med. Hidari, K.P., Suzuki, T., 2011. Dengue virus receptor. Trop. Med. Health 39, 37–43.
Int. Health 15, 619–631. Holmes, E.C., Burch, S.S., 2000. The causes and consequences of genetic variation in
Farrar, J., Hien, T.T., Horstick, O., Hung, N.T., Jaenisch, T., Junghanns, T., dengue virus. Trends Microbiol. 8, 74–77.
Kroeger, A., Laksono, I.S., Lum, L., Martinez, E., Simmons, C.P., Tami, A., Holmes, E.C., Twiddy, S.S., 2003. The origin, emergence and evolutionary genetics of
Tomashek, K.M., Wills, B., 2013. Dogma in classifying dengue disease. Am. J. dengue virus. Infect. Genet. Evol. 3, 19–28.
Trop. Med. Hyg. 89, 198–201. Hombach, J., Barrett, A.D., Cardosa, M.J., Deubel, V., Guzman, M., Kurane, I.,
Fischil, W., Bartenschlager, R., 2011. Exploitation of cellular pathways by dengue Roehrig, J.T., Sabchareon, A., Kieny, M.P., 2005. Review on flavivirus vaccine
virus. Curr. Opin. Microbiol. 14, 470–475. development. In: Proceedings of a Meeting Jointly Organised by the World Health
Franco, L., Palacios, G., Martinez, J.A., Vazquez, A., Savji, N., De Ory, F., Organization and the Thai Ministry of Public Health, 26–27 April 2004, vol. 23.
Sanchez-Seco, M.P., Martin, D., Lipkin, W.I., Tenorio, A., 2011. First report of Vaccine, Bangkok, Thailand, pp. 2689–2695.
sylvatic DENV-2-associated dengue hemorrhagic fever in West Africa. PLoS Negl. Hunsperger, E.A., Yoksan, S., Buchy, P., Nguyen, V.C., Sekaran, S.D., Enria, D.A.,
Trop. Dis. 5, e1251. Pelegrino, J.L., Vazquez, S., Artsob, H., Drebot, M., Gubler, D.J., Halstead, S.B.,
Friberg, H., Jaiswal, S., West, K., O’ketch, M., Rothman, A.L., Mathew, A., 2012. Guzman, M.G., Margolis, H.S., Nathanson, C.M., Rizzo Lic, N.R., Bessoff, K.E.,
Analysis of human monoclonal antibodies generated by dengue virus-specific Kliks, S., Peeling, R.W., 2009. Evaluation of commercially available anti-dengue
memory B cells. Viral Immunol. 25, 348–359. virus immunoglobulin M tests. Emerg. Infect. Dis. 15, 436–440.
Gamble, J., Bethell, D., Day, N.P., Loc, P.P., Phu, N.H., Gartside, I.B., Farrar, J.F., Hunsperger, E.A., Yoksan, S., Buchy, P., Nguyen, V.C., Sekaran, S.D., Enria, D.A.,
White, N.J., 2000. Age-related changes in microvascular permeability: a signif- Vazquez, S., Cartozian, E., Pelegrino, J.L., Artsob, H., Guzman, M.G., Olliaro, P.,
icant factor in the susceptibility of children to shock? Clin. Sci. (Lond.) 98, Zwang, J., Guillerm, M., Kliks, S., Halstead, S., Peeling, R.W., Margolis, H.S.,
211–216. 2014. Evaluation of commercially available diagnostic tests for the detection of
Garcia, G., Puerto, F.D., Perez, A.B., Sierra, B., Aguirre, E., Kikuchi, M., Sanchez, L., dengue virus NS1 antigen and anti-dengue virus IgM antibody. PLoS Negl. Trop.
Hirayama, K., Guzman, M.G., 2011. Association of MICA and MICB alleles with Dis. 8, e3171. http://dx.doi.org/10.1371/journal.pntd.0003171.
symptomatic dengue infection. Hum. Immunol. http://dx.doi.org/10.1016/ Kalayanarooj, S., Nimmannitya, S., 2005. Is dengue severity related to nutritional
j.humimm.2011.06.010. status? Southeast Asian J. Trop. Med. Public Health 36, 378–384.
Garcia, G., Sierra, B., Perez, A.B., Aguirre, E., Rosado, I., Gonzalez, N., Izquierdo, A., Kay, B., 1999. Dengue vector surveillance and control. Curr. Opin. Infect. Dis. 12,
Pupo, M., Danay Diaz, D.R., Sanchez, L., Marcheco, B., Hirayama, K., 425–432.
Guzman, M.G., 2010. Asymptomatic dengue infection in a Cuban population Khan, N.A., Azhar, E.I., El-Fiky, S., Madani, H.H., Abuljadial, M.A., Ashshi, A.M.,
confirms the protective role of the RR variant of the FcgammaRIIa polymorphism. Turkistani, A.M., Hamouh, E.A., 2008. Clinical profile and outcome of hospitalized
Am. J. Trop. Med. Hyg. 82, 1153–1156. patients during first outbreak of dengue in Makkah, Saudi Arabia. Acta Trop. 105,
Gibson, G., Souza-Santos, R., Brasil, P., Pacheco, A.G., Cruz, O.G., Honório, N.A., 39–44.
Kubelka, C., Carvalho, M.S., 2013. From Primary Care to Hospitalization: Clinical Khor, C.C., Chau, T.N., Pang, J., Davila, S., Long, H.T., Ong, R.T.,
Warning Signs of Severe Dengue Fever in Children and Adolescents during an Dunstan, S.J., Wills, B., Farrar, J., Van Tram, T., Gan, T.T., Binh, N.T., Tri
Outbreak in Rio de Janeiro, Brazil, vol. 29. Rev Saúde Pública, Rio de Janeiro, Le, T., Lien Le, B., Tuan, N.M., Tham, N.T., Lanh, M.N., Nguyet, N.M.,
pp. 82–90. Hieu, N.T., Van, N.V.C.N., Thuy, T.T., Tan, D.E., Sakuntabhai, A., Teo, Y.Y.,
Gubler, D., 1997. Dengue and dengue hemorrhagic fever: its history and resurgence Hibberd, M.L., Simmons, C.P., 2011. Genome-wide association study iden-
as a global public health problem. In: Gubler, D., Kuno, G. (Eds.), Dengue and tifies susceptibility loci for dengue shock syndrome at MICB and PLCE1. Nat.
Dengue Hemorrhagic Fever. CAB International. Genet. 43, 1139–1141.
Dengue 257

Kouri, G.P., Guzman, M.G., Bravo, J.R., 1987. Why dengue haemorrhagic fever Rodriguez-Roche, R., Villegas, E., Cook, S., Poh Kim, P.A.W., Hinojosa, Y., Rosario, D.,
in Cuba? 2. An integral analysis. Trans. R. Soc. Trop. Med. Hyg. 81, Villalobos, I., Bendezu, H., Hibberd, M.L., 2012. Population structure of the dengue
821–823. viruses, Aragua, Venezuela, 2006–2007. Insights into dengue evolution under
Kouri, G.P., Guzman, M.G., Bravo, J.R., Triana, C., 1989. Dengue haemorrhagic fever/ hyperendemic transmission. Infect. Genet. Evol. 12, 332–344.
dengue shock syndrome: lessons from the Cuban epidemic, 1981. Bull. World Sabchareon, A., Wallace, D., Sirivichayakul, C., Limkittikul, K., Chanthavanich, P.,
Health Organ. 67, 375–380. Suvannadabba, S., et al., 2012. Protective efficacy of the recombinant,
Kroeger, A., Lenhart, A., Ochoa, M., Villegas, E., Levy, M., Alexander, N., Mccall, P.J., live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a rando-
2006. Effective control of dengue vectors with curtains and water container covers mised, controlled phase 2b trial. Lancet 1–9.
treated with insecticide in Mexico and Venezuela: cluster randomised trials. BMJ San Martin, J.L., Brathwaite, O., Zambrano, B., Solorzano, J.O., Bouckenooghe, A.,
332, 1247–1252. Dayan, G.H., Guzman, M.G., 2010. The epidemiology of dengue in the americas
Lan, N.T.P., Hirayama, K., 2011. Host genetic susceptibility to severe dengue infection. over the last three decades: a worrisome reality. Am. J. Trop. Med. Hyg. 82,
Trop. Med. Health 39, 73–81. 128–135.
Lima, R.F., Croda, M.G., Araujo Muniz, D., Gomes, I.T., De Moraes Soares, K.R., Santiago, G., Vergne, E., Quiles, Y., Cosme, J., Vazquez, J., Medina, J.F., Medina, F.,
Rodrigues Cardoso, M., Marques Tauro, R.L.A., Croda, J., 2013. Evaluation of the Colon, C., Margolis, H., Muñoz-Jordan, J.L., 2013. Analytical and clinical perfor-
traditional and revised world health organization classifications of dengue cases in mance of the CDC realtime RT-PCR assay for detection and typing of dngue virus.
Brazil. Clinics 68, 1299–1304. PLoS NTD 7, e2311.
Loke, P.H., Hammond, S.N., Leung, J.M., Kim, C.C., Batra, S., Rocha, C., Sierra, B.D.L.C., Garcia, G., Perez, A.B., Morier, L., Alvarez, M., Kouri, G.,
Balmaseda, A., Harris, E., 2010. Gene expression patterns of dengue Guzman, M.G., 2006. Ethnicity and difference in dengue virus-specific memory
virus-infected children from nicaragua reveal a distinct signature of increased T cell responses in Cuban individuals. Viral Immunol. 19, 662–668.
metabolism. PLoS Negl. Trop. Dis. 4, e710. Simmons, C., Farrar, J., Chau, V.N., Wills, B., 2012. Dengue. N. Engl. J. Med. 366,
Lourenco, J., Recker, M., 2010. Viral and epidemiological determinants of the invasion 1423–1432.
dynamics of novel dengue genotypes. PLoS Negl. Trop. Dis. 4, e894. Smith, S.A., De Alwis, A.R., Kose, N., Jadi, R.S., De Silva, A.M., Crowe Jr., J.E., 2014.
McMeniman, Conor J., Lane, Roxanna V., Cass, Bodil N., Fong, Amy W.C., Isolation of dengue virus-specific memory B cells with live virus antigen from
Sidhu, Manpreet, Feng Wang, Yu, O’neill, Scott L., 2009. Stable introduction of human subjects following natural infection reveals the presence of diverse novel
a life-shortening Wolbachia infection into the mosquito Aedes aegypti. Science functional groups of antibody clones. J. Virol. 88, 12233–12241.
323, 141. St. John, A.L., Abraham, S.N., Gubler, D.J., 2013. Barriers to preclinical investigations
Montoya, M., Gresh, L., Mercado, J.C., Williams, K., Vargas, M.J., Gutierrez, G., of anti-dengue immunity and dengue pathogenesis. Nature 11, 420–426.
Kuan, G., Gotdon, A., Balmaseda, A., Harris, E., 2013. Symptomatic versus Sun, P., Kochel, T.J., 2013. The battle between infection and host immune responses of
inapparent outcome in repeat dengue virus infections is influenced by the time dengue virus and its implication in dengue disease pathogenesis. Sci. World J. 2013,
interval between infections and study year. PLoS NTD 7, 1–10. 843469. http://dx.doi.org/10.1155/2013/843469. Epub Feb 10, 2013. Review.
Narvaez, F., Gutierrez, G., Perez, M.A., Elizondo, D., Nunez, A., Balmaseda, A., Thisyakorn, U., Nimmannitya, S., 1993. Nutritional status of children with dengue
Harris, E., 2011. Evaluation of the traditional and revised WHO classifications of hemorrhagic fever. Clin. Infect. Dis. 16, 295–297.
dengue disease severity. PLoS Negl. Trop. Dis. 5, e1397. Thomas, S.J., 2014. Developing a dengue vaccine: progress and future challenges.
Nguyen, T.H., Nguyen, T.L., Lei, H.Y., Lin, Y.S., Le, B.L., Huang, K.J., Lin, C.F., Ann. N.Y. Acad. Sci. 1323, 140–159. E pub ahead of print.
Do, Q.H., Vu, T.Q., Lam, T.M., Yeh, T.M., Huang, J.H., Liu, C.C., Halstead, S.B., Villar, L., Dayan, G.H., Arredondo-García, J.L., Rivera, D.M., Cunha, R., Deseda, C.,
2005. Association between sex, nutritional status, severity of dengue hemorrhagic et al., 2015. Efficacy of a tetravalent dengue vaccine in children in Latin America.
fever, and immune status in infants with dengue hemorrhagic fever. Am. J. Trop. N. Engl. J. Med 372, 113–123. http://dx.doi.org/10.1056/NEJMoa1411037.
Med. Hyg. 72, 370–374. Voorham, J.M.S., Rodenhuis-Zybert, I.A., Ayala, N.V., Colpitts, T.M., Van Der
Pamplona, L.G.C., Mota, L.A., Lustosa, G.P., Fortes, M.C., Mota, D.A.M., Lima, A.A.B., Ende-Metselaar, H., Fikrig, E., Diamond, M.S., Wilschut, J., Smit, J.M., 2012.
Coelho, I.C.B., Mourão, M.P.G., 2014. Evaluation of the WHO Classification of Antibodies againts the envelope glycoprotein promote infectivity of immature
Dengue Disease Severity during an Epidemic in 2011 in the State of Ceará, Brazil. dengue virus serotype 2. PLoS One 7, 1–10.
Mem Inst Oswaldo Cruz, Rio de Janeiro, pp. 1–6. Wan, S.-W., Lin, C.-F., Yeh, T.-M., Liu, C.-C., Liu, H.-S., Wang, S., Ling, P.,
Pang, J., Salim, A., Lee, V.J., Hibberd, M.L., Chia, K.S., Leo, Y.S., Lye, D.C., 2012. Anderson, R., Lei, H.-Y., Lin, Y.-S., 2013. Autoimmunity in dengue pathogenesis.
Diabetes with hypertension as risk factors for adult dengue hemorrhagic fever in J. Formos. Med. Assoc. 112, 3–11.
a predominantly dengue serotype 2 epidemic: a case control study. PLoS NTD 6, Weiskopf, D., Sette, A., 2014. T-cell immunity to infection with dengue virus in
e1641. humans. Front. Immunol. 5, 1–6.
Perez, A.B., Sierra, B., Garcia, G., Aguirre, E., Babel, N., Alvarez, M., Sanchez, L., Weiskopf, D., Angelo, M.A., Sidney, J., Peters, B., Shresta, S., Sette, A., 2014.
Valdes, L., Volk, H.D., Guzman, M.G., 2010. Tumor necrosis factor-alpha, Immunodominance changes as a function of the infecting dengue virus serotype
transforming growth factor-beta1, and interleukin-10 gene polymorphisms: impli- and primary versus secondary infection. J. Virol. 88, 11383–11394. http://dx.doi.
cation in protection or susceptibility to dengue hemorrhagic fever. Hum. Immunol. org/10.1128/jvi.01108-14.
71, 1135–1140. Were, F., 2012. The dengue situation in Africa. Paediatr. Int. Child Health 32, 18–21.
Prasad, D., Kumar, C., Jain, A., Kumar, R., 2013. Accuracy and applicability of the Whitehorn, J., Simmons, C.P., 2011. The pathogenesis of dengue. Vaccine 29, 7221–7228.
revised WHO classification (2009) of dengue in children seen at a tertiary WHO, 1997. Dengue Haemorrhagic Fever: Diagnosis, Treatment, Prevention and
healthcare facility in northern India. Infection 41, 775–782. Control, second ed. WHO, Geneva.
Raghwani, J., Rambaut, A., Holmes, E.C., Hang, V.T., Hien, T.T., Farrar, J., Wills, B., WHO, 2003. Pyriproxyfen in Drinking Water. Background Document for Preparation of
Lennon, N.J., Birren, B.W., Henn, M.R., Simmons, C.P., 2011. Endemic dengue WHO Guidelines for Drinking-Water Quality. WHO/SDE/WSH/03-04/113. Geneva,
associated with the co-circulation of multiple viral lineages and localized Switzerland.
density-dependent transmission. PLoS Pathog. 7, e1002064. WHO, 2010. Working to Overcome the Global Impact of Neglected Tropical Diseases.
Rigau-Perez, J.G., 2006. Severe dengue: the need for new case definitions. Lancet First WHO report on neglected tropical diseases, Geneva.
Infect. Dis. 6, 297–302. WHO, 2012. Global Strategy for Dengue Prevention and Control. World Health Orga-
Rigau-Perez, J.G., Laufer, M.K., 2006. Dengue-related deaths in Puerto Rico, nization, ISBN 9789241504034.
1992–1996: diagnosis and clinical alarm signals. Clin. Infect. Dis. 42, WHO/TDR, 2009. Dengue Guidelines for Diagnosis, Treatment, Prevention and Control.
1241–1246. New ed. WHO Press, Geneva, ISBN 9789241547871.
Rodenhuis-Zybert, I.A., Moesker, B., Da Silva Voorham, J.M., Van Der Wilder-Smith, A., Quam, M., Sessions, O., Rocklov, J., Liu-Helmersson, J., Franco, L.,
Ende-Metselaar, H., Diamond, M.S., Wilschut, J., Smit, J.M., 2011a. A fusion-loop Khan, K., 2014. The 2012 dengue outbreak in Madeira: exploring the origins. Euro
antibody enhances the infectious properties of immature flavivirus particles. Surveill. 9 (8), 19. Available online: http://www.eurosurveillance.org/ViewArticle.
J. Virol. 85, 11800–11808. aspx?ArticleId¼20718.
Rodenhuis-Zybert, I.A., Wilschut, J., Smit, J.M., 2011b. Partial maturation: an Ye, J., Z, B., Fu, Z.F., Chen, H., Cao, S., 2013. Immune evasion strategies of flavi-
immune-evasion strategy of dengue virus? Trends Microbiol. 19, 248–254. viruses. Vaccine 31, 461–471.

Вам также может понравиться