- Access to this full-text is provided by Hindawi.
- Learn more
Download available
Content available from BioMed Research International
This content is subject to copyright. Terms and conditions apply.
Review Article
Production, Transmission, Pathogenesis, and Control of Dengue
Virus: A Literature-Based Undivided Perspective
Muhammad Torequl Islam,
1
Cristina Quispe,
2
Jesús Herrera-Bravo ,
3,4
Chandan Sarkar,
1
Rohit Sharma ,
5
Neha Garg,
6
Larry Ibarra Fredes,
7
Miquel Martorell ,
8,9
Mohammed M. Alshehri ,
10
Javad Sharifi-Rad ,
11
Sevgi Durna Daştan,
12,13
Daniela Calina ,
14
Radi Alsafi ,
15
Saad Alghamdi ,
15
Gaber El-Saber Batiha,
16
and Natália Cruz-Martins
17,18,19
1
Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University,
Gopalganj (Dhaka)8100, Bangladesh
2
Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
3
Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Chile
4
Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera,
Temuco 4811230, Chile
5
Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University,
Varanasi-221005, Uttar Pradesh, India
6
Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005,
Uttar Pradesh, India
7
European Institute of Traditional Chinese Studies, 4000-501 Porto, Portugal
8
Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción,
4070386 Concepción, Chile
9
Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepción 4070386, Chile
10
Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
11
Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
12
Department of Biology, Faculty of Science, Sivas Cumhuriyet University, 58140 Sivas, Turkey
13
Beekeeping Development Application and Research Center, Sivas Cumhuriyet University, 58140 Sivas, Turkey
14
Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
15
Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
16
Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
17
Faculty of Medicine, University of Porto, Porto, Portugal
18
Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
19
Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317,
4585-116 Gandra PRD, Portugal
Correspondence should be addressed to Javad Sharifi-Rad; javad.sharifirad@gmail.com, Daniela Calina; calinadaniela@gmail.com,
and Natália Cruz-Martins; ncmartins@med.up.pt
Received 13 July 2021; Revised 11 November 2021; Accepted 26 November 2021; Published 15 December 2021
Academic Editor: Cassiano Felippe Gonçalves-de-Albuquerque
Copyright © 2021 Muhammad Torequl Islam et al. This is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work
is properly cited.
Dengue remains one of the most serious and widespread mosquito-borne viral infections in human beings, with serious health
problems or even death. About 50 to 100 million people are newly infected annually, with almost 2.5 billion people living at
risk and resulting in 20,000 deaths. Dengue virus infection is especially transmitted through bites of Aedes mosquitos, hugely
Hindawi
BioMed Research International
Volume 2021, Article ID 4224816, 23 pages
https://doi.org/10.1155/2021/4224816
spread in tropical and subtropical environments, mostly found in urban and semiurban areas. Unfortunately, there is no particular
therapeutic approach, but prevention, adequate consciousness, detection at earlier stage of viral infection, and appropriate medical
care can lower the fatality rates. This review offers a comprehensive view of production, transmission, pathogenesis, and control
measures of the dengue virus and its vectors.
1. Introduction
Dengue regards one of the utmost serious arboviral infec-
tions around the world. dengue virus (DENV) is transmitted
through bites of female Aedes mosquitos especially Aedes
(Stegomyia) aegypti,Ae. alpopictus [1], Ae. niveus, and Ae.
polynesiensis [2]. DENV infection is almost similar to flu-
like infection and sometimes develops into possibly lethal
difficulties or severe illness including dengue shock syn-
drome and dengue hemorrhagic fever. World Health Orga-
nization (WHO) reports that DENV infection has been
shown to 30-fold increase around the globe over the past five
decades, and approximately 100 million newly infected peo-
ple are estimated in over 100 endemic countries with 20,000
deaths annually [3].
DENV and its vectors are primarily noticed in tropical
and subtropical environments globally, frequently in urban
and semiurban areas. In Bangladesh, DENV has been
detected as a severe health hazard. Between 2000 and 2008,
50,148 people were hospitalized for dengue in Bangladesh.
But in August 2019, nearly 60,000 dengue patients have been
hospitalized, and approximately 100 deaths have been
reported. Severe DENV infection is a leading cause of tena-
cious sickness and deaths of people of all ages in Asian and
Latin American countries. Unfortunately, we have no partic-
ular treatment strategy for dengue infection. It may be
because historically our pharmaceutical section did not
come up with much attention to this vector-borne viral
disease.
This review is aimed at sketching a current scenario on
DENV, dengue infection, and dengue vectors along with
the production, transmission, pathogenesis, and ways of con-
trol of DENV, and its vectors offers a comprehensive view of
production, transmission, pathogenesis, and control mea-
sures of DENV and its vectors.
2. Dengue Virus (DENV)
DENV, a pathogenic arthropod-borne flavivirus (arbovirus),
is a single-stranded and positive-sense RNA molecule
belonging to the family Flaviviridae.
The Flaviviridae family includes viruses transmitted by
arthropods that cause serious illness in humans. The family
includes a single genus-Flavivirus, with several types [4].
Recently, another subdivision of the family into three genera
has been proposed as follows: genus Flavivirus includes
arboviruses (yellow fever virus, dengue fever virus); genus
Pestivirus-viruses involved in animal pathology; and genus
Hepacavirus-the proposed name for different variants of
hepatitis C virus [4].
To date, 47 strains of DENV have been identified. The
total number of four closely linked serotypes (from DENV
-1 to -4) of DENV has been identified to date, but they are
lightly antigenically distinct [5, 6], and those can be subdi-
vided into several genotypes according to their gene
sequences [7]. These serotypes are generally progressed from
a mutual ancestor, and all are considered as the causative
agent of approximately similar disease spectrum in humans
due to DENV selecting different receptors based on cell
types and virus strains [8]. Developed viral particles have a
spherical shape with 11 kb in length and 40-50 nm in diam-
eter, containing single-stranded and positive-sense RNA
molecule, which has a 5-methyl cap with a single open read-
ing frame [2]. Dengue virus and its common four serotypes
have shown in Figure 1.
2.1. DENV Vectors. Dengue virus infection usually spreads
through bites of infected female mosquitos of genus Aedes,
especially by the Aedes aegypti and Ae. albopictus [1]. How-
ever, the other two vectors such as Ae. polynesiensis and Ae.
niveus have been identified as the secondary vectors in some
regions throughout the world [2] (Figure 2).
Adult Ae. aegypti has a white scale that forms a lyre or
violin shape at the dorsal side of the thorax, while the adult
Ae. albopictus forms a white stripe at the middle point of the
top of the thorax region. The white bands of every tarsal seg-
ment of the hind legs of these mosquitos are known as the
white stripe. The abdomen is generally found to be black
or dark brown, but sometimes, it also bears white scales.
Females are usually larger than males; on the other way,
through finding small palps tipped with silver or white
scales, they can be discriminated against properly. Males
are specially identified by the plumose type of antennae.
On the other hand, females are seen to bear short hair.
Under a microscope, the mouthparts of the male are
watched as a structural modification for nectar feeding,
and female mouthparts are viewed as a modified structure
for feeding on blood. The darkly coloured proboscis is found
to be present in both sexes. In addition, two clusters of white
scales presented on the segment above the proboscis are
known as clypeus. The tip of the abdomen is pointed out
as a distinctive feature of all Aedes species [9].
2.2. Geographical Distribution. DENV mainly originated
from monkeys, then jumped to humans in Southeast Asia
or Africa between 100 and 800 years ago. Geographically
DENV has been restricted till the 1950s, but after the Second
World War caused a rapid distribution throughout the
world. Firstly, DENV infection was recognized in the Thai-
land and Philippines in the 1850s, and after the 1980s
towards Latin America and the Caribbean. Presently, DENV
is prevalent throughout the different countries (at least 100
countries) including in Asia, the Pacific, the Americas,
Africa, and the Caribbean. DENV epidemics occurred in
26 states [10]. Scientific reports demonstrate that DENV-2
and 3 serotypes were mostly outbroken before 2000 and
2 BioMed Research International
between 2000 and 2009, respectively. DENV-1 serotype
started to dominate worldwide dengue outbreaks and after
2010, the DENV-4 [11].
The geographical distribution of DENV worldwide has
been shown in Figure 3.
Ae. Aegypti is scattered in tropical areas geographically,
and it breeds artificially in containers (such as tyres, drums,
flower vases including plastic food containers, tin cans, and
old motor parts) that are filled with water [12].
Ae. aegypti is an insect of holometabolous type, which is
fully developed after completing metamorphosis (i.g., four
growing phases from egg to adult period). The duration of
the life span of an adult may be 2 to 4 weeks; however, it
depends on the environmental conditions, at least 4-5 times
a female mosquito lays eggs throughout her whole life span
and the average 10 to 100 eggs in a single spawn. Three
diverse polytypic forms are found in Ae. aegypti such as syl-
van, domestic, and peridomestic [13].
A sylvan type is generally a rural form which breeds in
tree holes, normally in forests; the domestic type commonly
breeds in municipal surroundings, frequently inside or
around houses; and the peridomestic type usually survives
in biologically modified regions as groves and coconut farms
[14]. Ae. aegypti can survive above 4
°
C [15]; on the other
hand, about 15-37
°
C temperature is required for a complete
life cycle [16].
The extent of DENV epidemics not only depends on the
presence of DENV and mosquito genotypes but also
depends on how they interrelate with local temperature
[17]. Nevertheless, a current study demonstrated that DENV
infection can alter gene expression in the Ae. aegypti mos-
quito’s head that causes a loss of their olfactory preferences,
thereby modifying oviposition site choice [18]. Now, the
question is how safe is the host nervous system’s homeosta-
sis during Dengue infection?
2.3. Life Cycle. Primarily, the DENV was transmitted via syl-
vatic cycles in Asia and Africa by Aedes mosquito and the
nonhuman primates, with occasional appearances of human
populations [19]. However, nowadays, the global spread of
DENV follows its emergence of all types of transmissions
(e.g., sylvatic cycles and vertical: mosquito to mosquito).
Thus, its primary life cycle entirely involves the transmission
between Aedes mosquitoes and humans [20]. One report
suggests that dogs or other animals may act as incidental
hosts and may serve as reservoirs of DENV infection [21].
Life cycles of mosquitoes have been shown in Figure 4.
2.4. Immune Defensive Pathways. The Toll pathway is one of
the well-known potential immune defensive pathways
against the DENV and its serotypes bearing Ae. aegypti
[22]. In a study, after ten days postinfection of DENV, the
antioxidant enzymes were found to suppress, while upregu-
lated the expressions of Toll, JAK-STAT, and pathogen rec-
ognition receptor (PRR) [23]. It has also been analyzed that
the JAK-STAT pathway is another important DENV defen-
sive pathway in invertebrates [24, 25]. The mosquitoes of
genus Aedes should be more vulnerable to DENV infection
if the receptor JAK homolog HOP or Dome is inhibited by
RNA inference (RNAi, e.g., ds RNA and prM RNA) [25, 26].
Capsid
E & M proteins
Viral genome
Viral envelope
Serotype 1
0.002
Serotype 2
0.002
Serotype 3
0.002
Serotype 4
0.002
Figure 1: Diagram with dengue virus and its four serotypes.
Aedes aegypti Aedes albopictus
Aedes polynesiensis Aedes niveus
Figure 2: Aedes mosquitoes (dengue virus vectors).
3BioMed Research International
In a study, miR-375 was found to enhance DENV2 rep-
lication capacity [27]. In another study, in the period of
DENV infection, miRNAs were identified in different forms
(about sixty-six) in Ae. albopictus, where upregulated miR-
34-5p targets the Toll pathway signalling protein (REL-1)
[28]. Conversely, downregulated peptidoglycan recognition
protein LE, and AMP defensin D. miR87 targets the Toll
pathway [28].
Average of number of reported cases, 2010–2016
≥100 000 0 cases reported
No data
Not applicable
10 000 – 99 999
1 000 – 99 999
<1 000
Figure 3: Geographical distribution of dengue worldwide.
Eggs
Mosquito Life Cycle
Adult
Pupal stage
Fourth larval stage
ird larval stage
Second larval stage
First larval stage
Figure 4: Mosquito life cycle.
4 BioMed Research International
Differential expression of miRNAs in DENV has been
also reported by Yen et al. [29]. In this study, the authors
highlighted the possibility of using artificial antiviral miR-
NAs to reduce the transmission of two major arboviruses
in transgenic Ae. Aegypti. The miRNA-based approach
resulted in a dual resistance phenotype for Dengue serotype
3 viruses (DENV-3).
The piRNAs also plays essential roles in the innate anti-
viral response in DENV [30–33]. Moreover, nonretroviral
integrated RNA viruses (NIRVS) were recognized in Ae.
aegypti and Ae. albopictus in a larger number [34].
The expression of cecropin-like AMPs was expressively
upregulated by the infection of DENV [24]. In Ae. aegypti,
the immune deficiency (IMD) pathway shows a significant
role to resist DENV susceptibility, while the increase in viral
replication [35]. The ubiquitin variant (Ub3881) residues
may inhibit the DENV envelope protein, thereby and
decrease the production rate of DENV in Aedes vectors
[36, 37].
The DENV-containing blood meal first appears in the
midgut of the vectors, which has the first line of defense sys-
tems, such as the infection barrier and the escape barriers
[38, 39]. It is evident that, after a successful entry of DENV,
something has happened inside the midgut cells such as
uncoating, replication, and new virus particle assembly.
The innate immune signalling pathways have been seen to
be effective during the infection of DENV in Ae. aegypti.
Exogenous siRNA pathways also play a substantial role
against DENV infections in the Aedes midgut [40]. DENV
infection causes the production of NO in the hemolymph,
where the virus is released into the hemocoel from the mid-
gut. The hemocytes allow replication other than the distribu-
tion of DENV. Interestingly, DENV replication in
hemocytes is extensively inhibited by NO [41]. It has been
reported that about 40 differentially bacterial types have
been isolated from the gut of Ae. aegypti through a gut
microbiome study [42]. In another study, colonization of
Csp_P in the midgut of the Ae. aegypti also inhibited DENV
infection [43]. The Talaromyces (Tsp) secretome shows a
considerable modulating effect on the midgut transcriptome.
Tsp secretome may display a significant role in the advance-
ment of DENV infection in the midgut through downregu-
lating trypsin encoding genes involved in the digestion of
blood and through reducing the enzymatic activity of trypsin
[44]. It is cited that the presence of gram-negative endosym-
biotic bacteria Wolbachia spp. in Aedes mosquitoes effec-
tively suppressed the DENV infection [45]. Wolbachia
activates antimicrobial peptides defensin and cecropin Toll
pathway through producing reactive oxygen species (ROS)
after inducing a reduction-oxidation (Redox) reaction in
the mosquitos [46]. Wolbachia also upsurges vago1 expres-
sion in Ae. aegypti by acting as a ligand of the JAK-STAT
pathway [47].
Ae. aegypti macroglobulin complement related factor
(AaMCR) recognizes DENV particles. An anti-DENV effect
on Aedes mosquitoes has been found to link with the upreg-
ulation of AMP expression in the hemocytes [48]. The sali-
vary glands also contain the infection barrier and the
escape barriers [49]. Moreover, incomplete apoptosis of
DENV occurs here, which is required for the virus to release
via saliva [50]. A study revealed that multiple immune
defensive pathways (e.g., Toll and IMD) can be found here,
and this can rise putative antibacterial proteins/peptides
(e.g., attacin, cecropins, defensins,and gambicin) [24, 28,
35, 48, 51]. In the brain of Ae. aegypti, a homolog of Hikaru
Genki (AaHig)has been found to express ubiquitously [52].
Lipid droplets (LDs) containing a few exclusive struc-
tural proteins (Perilipin 1, 2, and 3) and a fatty acid mono-
layer are exclusively found to present in a variety of
organisms including DENV. These have been found to pro-
vide immunological defense of Aedes mosquitoes [53–55].
3. DENV Infection
3.1. Transmission. After initial midgut infection, DENV dis-
tributes systemically through the body cavity (commonly
known as hoemocel) of Aedes vectors, after that way dissem-
inates in secondary tissues. The time taken between initial
midgut infection and successive transmission of DENV by
its vector (e.g., Ae. aegypti) is termed as extrinsic incubation
period (7 to 14 days at 25-30
°
C). DENV stays in the midgut
of the vectors which it may be due to the viral genome being
stable here [36].
The ubiquitin-proteasome, an important pathway, acts
significant activity in the regulation of infectious DENV pro-
duction in vectors [36]. Finally, an infection of the salivary
glands and the release of virions into the host’s saliva occur
throughout the DENV transmission to the host [56]. Blood
cells and plasma are important media for the four serotypes
of DENV spreading into the host. A relation of domain III
from the envelope glycoprotein of DENV-II with human
plasma proteins has been identified by Huerta et al. [57]
[57]. DENV infection inductees after the attachment of the
dengue virus to the target cell through interfaces between
the various cell surface receptors and viral envelope (E) pro-
tein [58]. In mammalian cells, all categorized serotypes
interact with mannose, heparan sulfate, nLc4Cer, and DC-
SIGN/L-SIGN receptors.
Additionally, the DENV-2 serotype is found to intensity
of binding with GRP78, CD14-associated protein, HSP70/
HSP90, and two other unidentified receptor proteins. Con-
versely, serotypes DENV 1-3 bind with the laminin receptor
while serotypes DENV 2-4 attach with an unknown protein
receptor [59].
DENV after receptor-mediated endocytosis, virion fuses
with acidic lysosomes, and its genomic RNA is released into
the cytoplasm and translated into a polyprotein of ~3400
amino acids (genome is about 11000 bases of positive-sense,
a single-stranded RNA (ssRNA)) that are further cleaved by
viral and host proteases into three structural (capsid: C,
membrane: M, and envelope: E) and seven nonstructural
(NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) pro-
teins [60].
C protein is a foremost structural component of DENV
that is localized in the cytoplasm and nuclei [61, 62]. The
nuclear localization of this protein is thought to be crucial
for its well-organized replication [61, 63].
5BioMed Research International
The lipid bilayer of virions is formed by lipid (approxi-
mately 17% by weight) between the nucleocapsid core and
E/M outer shell [64, 65]. During the replication of DENV,
a membrane-bound replication complex formation helps to
incorporate host factors, viral proteins, and genomic RNA
[66]. In this case, positive-strand (+) DENV genomic RNA
acts as a template to synthesize complementary negative-
strand (-) RNA, which is sequentially used for multiple (+)
RNA genomes production that is obtainable for translation
and regulation of replication cycles or packaging into virions
[67]. However, in a study, Raquin and Lambrechts [68]
showed the presence of DENV genomic RNA in the salivary
galnds of Ae. aegypti, indicating an active replication of
DENV in its vector prior to transmission [68]. DENV itself
encodes RNA-dependent RNA polymerases, and the infec-
tion cycle of this virus is catalyzed by other cellular fac-
tors [69].
DENV infections can alter many important proteins in
the subcellular locales, including the Alix apoptosis-linked
gene-2-interacting protein X; therefore, blocking this step
may be one of the innovative beneficial approaches to reduce
DENV replication in the host [70]. Instead, several genes
have been identified that reduce infection of DENV when
silenced by at least 60% in its most important vector Ae.
aegypti. Among them, a putative cysteine-rich venom pro-
tein SeqID AAEL000379 (CRVP379) silencing has been
found to reduce DENV infection significantly in the cells
of midgut tissues of Ae. aegypti [71].
Loqs2, a gene has been found only in Aedes mosquitoes,
which is essential for the appropriate effectiveness of RNA
interference in this type of mosquito. However, without
Loqs2, the viruses can multiply and consequently infect their
salivary glands [72].
An interaction between DENV nonstructural protein 4A
(NS4A) and host cellular vimentin has been demonstrated in
localizing and concentrating the viral replication complex at
the perinuclear site, in consequence assisting well-organized
replication of viral RNA [73].
Figure 5 shows a general DENV transmission mode in
its vector to hosts.
3.2. Pathogenesis. DENV is usually greater in tropical and
subtropical environments throughout the world, frequently
in urban and semiurban zones. People, exposed to infected
mosquitoes of all ages are susceptible to DENV infection.
DENV infection causes dandy fever, breakbone fever, and
dengue hemorrhagic fever; and in severe cases, dengue shock
syndrome has occurred. The rainy season is the most
favorable climate for DENV infection outbreaks in tropical
countries in Asia and South America. Generally, the
infected female Aedes mosquitoes transmit DENV in
humans. Although humans are not capable of transmitting
DENV, it can be transmitted during the blood transfusion
between an infected person to a noninfected (healthy) per-
son [74, 75].
3.3. Physiological Data. After the incubation period (3 to 14
days) of DENV, the person may experience one or more
early symptoms such as nausea, headache, rash, fever, mus-
culoskeletal pain, and joint pain [76]. In classic dengue fever,
body temperature ranges from 39 to 40
°
F (5-7 days) [77]. In
the meantime, the DENV may enter systematically into the
bloodstream at the peripheral zones and sequentially dam-
age lymph nodes and blood vessels resulting in dengue hem-
orrhagic fever [78]. Symptoms of the latter case include
bleeding under the skin and from the gums and nose. On
the other hand, difficulty in breathing appears in patients
having dengue hemorrhagic fever, and severe progress of it
can lead to dengue shock syndrome, if left untreated, can
result in death.
3.4. Micronutrient Imbalance. The morphogenesis and
translation and/or replication of DENV occur in the endo-
plasmic reticulum (ER) [79], where Ca
2+
plays a significant
activity in cell signaling. The immune response of T-cell
has been drawn in DENV infection. At the time of secondary
infection (i.e., infection after 1-2 days of fever onset), high
concentration of interferon-alpha (IFN-α) is found, while
high levels of soluble interferon γ(IFN-γ), interleukin 2
receptor (IL-2R), and soluble CD4 and CD8 were reported
throughout the outset of vascular permeability [80, 81]. Den-
gue antigen is evident to increase the influx of Ca
2+
into T-
cells, thus reducing blood Ca
2+
levels [82, 83].
A multifunctional intermediate messenger protein cal-
modulin is well known as a primary sensor of intracellular
Ca2+ in the eukaryotic cells, which plays imperative utility
for proper decoding of Ca2+ signalling [84]. DDX3X is a
DEAD-box RNA helicase, which binds with the TRPV4 cat-
ion channel that regulates its functions. DDX3X is released
by the TRPV4-mediated Ca
2+
influx; at the same time,
DDX3X nuclear translocation is derived through a process
involving calmodulin and its kinase II-dependent pathway.
Therefore, pharmacological inhibition or genetic deple-
tion of TRPV4 can diminish DDX3X-dependent functions,
including translation and nuclear viral export. Thus, target-
ing TRPV4 may reduce the infectivity of some viruses,
including dengue, Zika viruses, and hepatitis C [85]. In a
study, the effect of W-7, a calmodulin antagonist in DENV
infection in Huh-7 cells, was seen, where W7 was inhibited
viral yield, NS1 secretion and viral RNA, and protein synthe-
sis, possibly through direct inhibition of NS2B-NS3 activity
and/or inhibition of the interaction between NS2A with
Ca
2+
-calmodulin complex [86]. Calcium depletion can mod-
ulate cardiac functions, immunopathogenesis, and platelet
functions in dengue infection [82]. Another study on 36 h
postinfection of Huh7 cells has been demonstrated that cal-
cium modulating cyclophilin-binding ligand influences the
apoptosis process by changing the activation of caspase-3
and the potentiation of mitochondrial membrane [87].
3.5. Clinical Aspects. In most cases, asymptomatic or mildly
symptomatic pathways are promising ways for transiting
DENV infection [88].
The most common signs and symptoms include pain of
bone, joint, muscle, and retro-orbital; headache; fever
(40
°
C); maculopapular or macular rash; and minor hemor-
rhagic manifestations including purpura, malaise, ecchymo-
sis, petechiae, epistaxis, hematuria, bleeding gums, aches or
6 BioMed Research International
pain, or a positive tourniquet test result. Dengue fever lasts
from 3 to 7 days. Before appearing the warning signs of
severe DENV infection, a slight portion of the infected
patients goes to life-threatening conditions [89].
Severe DENV infection can cause organ impairment,
bleeding, and plasma leakage. The warning signs during
dengue infection include vomiting, abdominal pain, respira-
tory distress, clinical/fluid accumulation, lethargic condition,
mucosal bleeding, liver enlargement (>2 cm), restlessness,
lethargic condition, and rapid decrease in platelet count.
An intensive care should be taken for the patients having
infancy, pregnancy, chronic hemolytic diseases, renal failure,
diabetes, obesity, and old age [2]. Chronic infections of
DENV may preserve in the central nervous system and can
be considered in progressive dementia patients [90].
In a recent study by Suppiah et al., the link between clin-
ical manifestation characteristic of Dengue fever and geno-
types, respectively, and DENV-specific phenotypes, was
highlighted. Thus, it was found that the clinical symptoms
are more severe in patients infected with DENV 2 serotype,
compared to patients infected with DENV1 serotype. Mus-
culoskeletal manifestations are characteristic of DENV
Dengue virus
Aedes mosquito
Blood transfusion
Human Human
Aedes mosquito
Other animals
(e.g., dog and monkey)
Figure 5: DENV transmission mode between the vector and hosts.
Table 1: Laboratory diagnostic approaches for DENV infection detection.
Clinical sample Diagnostic approach Methodology
Acute serum (1-5 days of DF) and necropsy
tissue
Virus isolation Mosquito or mosquito cell culture inoculation
Nucleic acid detection RT-PCR, real-time PCR
Antigen detection NS1 Ag rapid test, NS1 Ag capture ELISA,
immunohistochemistry
Paired sera
S1: acute serul from 1 to 5 days
S2: convalescent serum 15-21 days
IgG or IgM seroconversion (S1
to S2)
ELISA
HI
Plaque reduction neutralization test
Serum after day 5 of DF IgM detection MAC-ELISA, IgM rapid tests (lateral flow)
IgG detection IgG ELISA, HI, IgG rapid tests (lateral flow)
Abbreviations: Ag: antigen; DF: dengue fever; ELISA: enzyme-linked immunosorbent assay; HI: hemagglutination inhibition assay; IgG: immunoglobulin G;
IgM: immunoglobulin M; MAC: immunoglobulin M antibody capture; NS1: non-structural protein 1; RT-PCR: reverse-transcription polymerase chain
reaction.
7BioMed Research International
serotype 3 infection [91]. Also, nonstructural proteins (e.g.,
NS1, NS3, and NS5) can be targeted to develop a novel vac-
cine strategy [92, 93].
3.6. Diagnosis. Unfortunately, still, the signs and symptoms
are the foremost tools for the DENV infection diagnosis
[94, 95]. Fever or flu-like fever is the initial tool of DENV
infection.
To date, the well-known tests for detecting the presence
of DENV include identification of the responsible viral
genomic sequences, DENV serotype, viral antigen(s) (e.g.,
NS1 by MAC-ELISA assays) and/or antibodies in response
to it (e.g., IgG, IgM), and platelet counts.
Other important diagnosis includes viral RNA detection
(by nucleic acid amplification tests (NAAT) or RT-PCR),
detection of dengue specific monoclonal antibodies, IgM
captured ELISA, alive and/or viral isolation from mosquito
cell lines [96–100]. Immune-fluorescence tests, capture
ELISA, and hemagglutination assays are the commonly used
laboratory methods [101]. Other test includes +ve tourni-
quet test, leukopenia, HCT concurrent with a rapid decrease
in platelet count, AST or ALT ≥1000 IU/L, and impaired
consciousness [102]. Some important diagnostic approaches
and methodology have been shown in Table 1.
4. Control of DENV and Its Vectors
Public awareness counts as one of the major consequences of
the management of DENV, which essentially helps to avoid
or inhibit the contacts of the infected Aedes mosquitoes or
other animals and their derivatives [103]. In this way, Ae.
aegypti was properly eradicated during the 1960s from dif-
ferent areas of the USA. For this, a well-educated society
needs the strongest collaborative activities with skilful and
well-trained mosquito control staff[104].
It is possible to control DENV infection by using differ-
ent interesting methods.
4.1. Preventive Measures. Preventive measures should be the
first and best choice in this case, such as the prevention of
direct contact of blood or blood-derived products from the
infected patients and infected vectors from the infected host
[105]. Daytime is the most suitable time for biting Aedes
mosquitoes; consequently, its contact can be diminished or
avoided using the following techniques:
(i) By using nets (e.g., insecticide-treated nets) and
mosquito repellents (e.g., coils, solids (sticks), aero-
sols, liquids, pump sprays, and nonsticky creams)
(ii) By wearing gloves and other defensive clothing
(iii) Through well-planed management of wastes and
stored water
(iv) By destroying the mature Aedes mosquitoes or lar-
vae through applying some protective chemicals
(e.g., N,N-Diethyl-3-Methylbenzamide, diethyl car-
bonate, metofluthrin, oil of lemon-eucalyptus,
Smooth Endoplasmic
Reticulum
Terpenoids Virus Release
Virus
Maturation
Uncoating
+ss RNA
Translation &
Polyprotein Processing
Replication Compled
+ss RNA
Immature
Virion
Virus
assembly
MR Receptor
DC-SIGN
Receptor
Endocytosis
pH-dependent
fusion with endocytosis
Flavonoids
Alkaloids
Polyphenols
Chalcone
Exocytosis
Flavonoids
Coumarins
Polyphenols
Flavonoids
Fusion & Entry
Nucleolus
Nucleolus
Nuclear
Membrane
Ion
Channel
Figure 6: Potential antiviral mechanism and molecular targets of the bioactive compounds inhibiting viral entry and replication of dengue
virus.
8 BioMed Research International
Table 2: Antidengue activities of natural compounds.
Botanical name Plants part Isolated compounds Model Results References
Garcinia
mangostana Fruits α-Mangostin
DENV infection in human
peripheral blood mononuclear cells
(PBMC) in vitro
↓virus replication, ↓TNF-α,↓IFN-γ,↓IL-6, ↓MIP-1β,↓IP-10 [196, 197]
Anacolosa
pervilleana Leaves Octadeca-9,11,13-triynoic acid DENV NS
5
RNA-dependent RNA
polymerase (RdRp) assay in vitro IC50 =3μM[198]
Streptomyces
aureofaciens Fermentation Narasin DENV2-infected hepatocytes Huh-
7 cells in vitro IC50 =1μM↓viral protein synthesis [199]
Glycyrrhiza glabra Root
Glycyrrhizin DENV serotypes1-3 in vitro EC50 = 450, 174.2, 632.7 μg/mL [200]
Glycyrrhizic acid DENV2 infected Vero E6 cells
in vitro IC50 =8:1μM[201]
Squalus acanthias Liver Squalamine Human endothelial cells HMEC-1
in vitro ↓viral infection IC50 = 100 μg/mL [202]
Zastera marina.
Rees
Marine
eelgrass Zoasteric acid DENV serotypes (1–4) in vitro IC50 =24, 46, 14, 47 μmol/L [137]
Quercus lusitanica Galls Methyl gallate DENV-2 infected C6/36 cells
in vitro
↓DENV-2 NS2B/3 protease IC50 =0:3 mg/mL
↓NS1 protein [203]
Flacourtia
ramontchi Stem bark Flacourtosides A, E DENV NS
5
polymerase RdRp
in vitro IC50 =9:3−9:5μmol/L [204]
Gymnochrinus
richeri
Stalked fossil
crinoïd Gymnochrome B DENV-2, DENV-4 infected PS cells
in vitro ED50 =0:029 nmol/mL [205]
Gymnochrinus
richeri
Living fossil
crinoid
Gymnochrome D,
isogymnochrome D DENV-1 infected PS cells in vitro Reduction of foci was smaller than 1 μg/mL [206]
Arrabidaea
pulchra Leaves Verbascoside,
caffeoylcalleryanin, ursolic acid DENV-2 infected Vero cells in vitro EC50 =3:2, 2.8, 3.4 μg/mL [207]
Trigonostemon
cherrieri
Bark and
wood
Trigocherrin A,
trigocherriolides A and B
DENV NS
5
polymerase RdRp
in vitro IC50 =12:7, 3.1, 16.0 μmol/L [208]
Micromonospora
rhodorangea Whole part Geneticin DENV-2 infected BHK cells in vitro EC50 =2μg/mL [209]
Castanospermum
australe Seeds Castanospermine
DENV-2 infection of Huh-7 and
BHK-21 cells 10
5
PFU of mouse-
adapted DENV-2 in vitro/in vivo
IC50 =1μM↓mortality in a mouse model
Dose = 10, 50, and 250 mg/kg [210]
Coptis chinensis
Franch Rhizomes Palmatine DENV-2 infected Vero cells in vitro EC50 =26:4μmol/L [211]
Psychotria
Ipecacuanha Roots Emetine hydrochloride DENV-2 infected Huh-7, BHK-21
in vitro IC50 =0:5μM[212]
Distictella elongate
(Vahl) Urb
Leaves and
fruits
Petcolinarin and acacetin-7-O-
Rutinoside
DENV-2 infected Vero, LLCMK2
cells in vitro EC50 =86:4 and 11:1μg/mL [213, 214]
Scutellaria
baicalensis Roots Baicalein DENV-2 infected Vero cells in vitro IC50 =6:46 μg/mL [180, 215]
9BioMed Research International
Table 2: Continued.
Botanical name Plants part Isolated compounds Model Results References
Cryptocarya
chartacea Barks Chartaceones C-F Dengue virus NS
5
RdRp inhibition
in vitro IC50 =1:8to4:2μM[216, 217]
Boesenbergia
rotunda (L.) Rhizomes Panduratin A 4-
hydroxypanduratin B
DENV-2 NS2B/NS3 protease
in vitro Ki inhibitory constants
ðÞ
= 21, 25 μmol/L [123]
Tephrosia s.p. Aerial parts Glabranine 7-O-methyl-
glabranine DENV-2 serotype in vitro 70% inhibition IC50 =25mM [134]
Mimosa scabrella
Seeds
Mannose/galactose (1 : 1) DENV-1 (Hawaii strain) virus
in vitro
↓virus titer IC50 = 347 mg/L [114, 131]
Leucaena
leucocephala Mannose/galactose (1 : 4) ↓virus titer IC50 = 37 mg/L [114, 131]
Gymnogongrus
torulosus
Red seaweed
DL-galactan hybrids
DENV-2 serotype infected Vero
cells in vitro
IC50 =0:19 −1:7μg/mL [128]
G. griffithsiae and
Cryptonemia
crenulata
Sulfated G3d and C2S-3
polysaccharides IC50 =1μg/mL [125]
Cladosiphon
okamuranus
Brown
seaweeds Fucoidan DENV-2 infected BHK-21 cells
in vitro IC50 =4:7μg/mL [78, 218]
Nephelium
lappaceum L. Whole plant Geraniin DENV-2 E domain III (rE-DIII)
protein in vitro IC50 =1:75 μM[219, 220]
Scutellaria
baicalensis Radices Baicalin DENV-2 (NGC strain) infected
Vero cells in vitro IC50 =13:5±0:08 μg/mL [221, 222]
Camellia sinensis Dried leaves Epigallocatechin gallate Dengue virus (serotypes 1–4)
infected Vero cells in vitro EC50 =14:8, 18, 11:2, and 13:6μM[223]
Zoanthus spp. Animal
materials Zoanthone A DENV-2 NS
5
polymerase in vitro EC50 =19:61 ± 2:46 μM[224]
Mammea
americana Seeds Coumarin A
Coumarin B DENV-2/NG strain in vitro
EC50 =9:6 and 2:6μg/mL [225]
Tabernaemontana
cymosa Seeds Lupeol acetate
Voacangine EC50 =37:5 and 10:1μg/mL [225]
Angelica keiskei Roots Brefeldin A DENV serotypes (1–4) in vitro IC50 =54:6±0:9nM (DENV-2)
IC50 =61:32 ± 13:5, 57:9±0:1, and 65:7±6:3nM DENV−1, 3, 4
ðÞ
[226]
Uncaria
rhynchophylla Leaves Hirsutine DENV-1 infected A549 cells
in vitro EC50 =1:97 μM[227]
Viola yedoensis
Makino Aerial parts Luteolin DENV infected HEK-293 T, A549,
and BHK-21 cells in vitro EC50 =4:36 to 39:16 mM [228]
Persea americana Fruits (2 R,4 R)-1,2,4-
Trihydroxyheptadec-16-yne DENV serotypes (1–4) in vitro EC50 =14:61, 10:98, 12:87, and 14:61 μM[229]
Nephelium
lappaceum Rind Geraniin DENV-2 RNA synthesis in Vero
cells in vitro IC50 =1:78 μM[195]
Palythoa mutuki Peridinin DENV NS2B/NS3 protease in vitro IC50 =4:50 ± 0:46 μg/mL [230]
10 BioMed Research International
Table 2: Continued.
Botanical name Plants part Isolated compounds Model Results References
Formosan
zoanthid
Ganoderma
lucidum
Fruiting
bodies Ganodermanotriol IC50 = 50, 25 μM[231]
Faramea bahiensis Leaves
5-Hydroxy-4′-methoxy-
flavanone-7-O-ß-D-
apiofuranosyl-(1 →6)-β-D-
glucopyranoside
DENV-2 in HepG2 cells in vitro ↓viral replication
↓infected cell number [232]
Rhodiola rosea Roots Salidroside
DENV serotype-2 infection in vitro
↓DENV envelope protein
↑RNA helicases [233]
Swietenia
macrophylla Seeds Swielimonoid B EC50 =7:2±1:33 μM[234]
11BioMed Research International
Figure 7: Chemical structures of natural compounds acting against dengue.
12 BioMed Research International
diethyl phthalate, ethyl hexanediol, and picaridin)
[106, 107].
CYD-TDV (brand name Dengvaxia), an one and only
FDA approved live-attenuated dengue vaccine prepared by
applying rDNA technology through substituting the pre-
membrane (PrM) and envelope (E) structural proteins of
the 17D strain of attenuated yellow fever vaccine with those
from the dengue serotypes excepting DENV-5 serotype, is
manufactured by SanofiPasteur [108, 109]. Other vaccines
under development are DENVax/TAK-003 (recombinant
chimeric vaccine with DENV-1, -3, and -4 components on
the DENV-2 backbone, developed at Mahidol University in
Bangkok) [110, 111], TetraVax-DV (tetravalent admixture
of monovalent vaccines, being tested in Brazil and Thailand
in phase II trial) [112], TDEN PIV (inactivated tetravalent
vaccine, being experimented by the Walter Reed Army Insti-
tute of Research and GSK in phase I clinical trials) [113],
V180 (recombinant subunit vaccines expressed in Drosoph-
ila cells, undergoing phase I trial by Merck [114], and DNA
vaccines (the Naval Medical Research Center attempted to
develop a monovalent DNA plasmid vaccine) [111].
4.2. The Role of Natural Products and Their Bioactive
Constituents in Controlling DENV Infection. Natural prod-
ucts are the potential sources of many important modern
medicines [115–117]. Plants and/or their extracts having
antidengue activities are also distributed worldwide
[118–120]. To date, a number of medicinal plants have been
reported to act against DENV and/or their vectors, for
example, Alternanthera philoxeroides (Fam: Amarantha-
ceae) [121], Azidarachta indica (Fam: Meliaceae) [122], Boe-
senbergia rotunda (Fam: Zingiberaceae) [123], Carica
papaya (Fam: Caricaceae) [124], Cladosiphon okamuranus
(Fam: Chordariaceae) [78], Cryptonemia crenulata (Fam:
Halymeniaceae) and Gymnogongrus griffithsiae (Fam: Phyl-
lophoraceae) [125], Cymbopogon citratus (Fam: Poaceae),
Andrographis paniculata (Fam: Acanthaceae), Momordica
charantia (Fam: Cucurbitaceae), Ocimum sanctum (Fam:
Labiatae), Piper retrofractum (Fam: Piperaceae) [126], Fla-
gellaria indica (Fam: Flagellariaceae), Cladogynos orientalis
(Fam: Euphorbiaceae), Rhizophora apiculata (Fam: Rhizo-
phoraceae) and Houttuynia cordata (Fam: Saururaceae)
[127], Gymnogongrus torulosus (Fam: Phyllophoraceae)
[128], Lippia alba and L. citriodora (Fam: Verbenaceae)
[129], Meristiella gelidium (Fam: Solieriaceae) [130],
Mimosa scabrella (Fam: Fabaceae) [131], Psidium guajava
(Fam: Myrtaceae) and Euphorbia hirta (Fam: Euphorbia-
ceae) (Abd [132]), Quercus lusitanica (Fam: Fagaceae)
[133], Tephrosia crassifolia,Tephrosia madrensis, Leucaena
leucocephala, and Tephrosia viridiflora (Fam: Fabaceae)
[131, 134, 135], Uncaria tomentosa (Fam: Rubiaceae)
[136], Zostera marina (Fam: Zosteraceae) [137], Myristica
fatua,Cymbopogon citratus and Acorus calamus [138], Dor-
atoxylum apetalum [139], Psiloxylon mauritianum [140],
Acorus calamus (Fam: Acoraceae) [141], Cinnamosma fra-
grans [142], Pedalium murex [143], Aesculus hippocastanum
[144], Norantea brasiliensis [145], Azadirachta indica [146],
Spondias mombin [147], Angelica sinensis [148], Phyllanthus
spp. [149], Solanum xanthocarpum,Mesocyclops thermocy-
clopoides (Mahesh [150]), Delonix elata (Fam: Fabaceae)
[151], Acalypha alnifolia (Fam: Euphorbiaceae) [152], Com-
bretum collinum [153], and Solanum villosum [154].
The aqueous extract of Houttuynia cordata (10-100 mg/
mL) against DENV-2 with human hepatocarcinoma cell lin-
eage (HepG2) cells showed that extract significantly
decreased intracellular DENV-2 RNA production with the
reduction in the expression of dengue protein. It also
showed a potential role in the release of the virion from
infected LLC-MK2 cells at 10-40 mg/mL concentrations
[155]. 9 N-methylamine and Harmol may selectively inhibit
DENV-2 multiplication without virucidal effect in cell cul-
tures [156].
The ethyl acetate fraction of H. cordata and quercetin
showed in vitro activity against mouse hepatitis virus
(MHV) and DENV-2 with IC
50
0.98 and 125 μg/mL for
MHV while 7.50 and 176.76 μg/mL for DENV-2 [157]. Del-
phinidin and epigallocatechin gallate showed a direct effect
on against West Nile virus (WNV) and also reduced the
infectivity of ZIKV and DENV. The effect of delphinidin
and, particularly of epigallocatechin gallate, was found
higher for the African strain (MR766) than for the American
strain (PA259459) [158].
In another study, it was found an absence of anti-DENV
activity in chemical constituents like acetyl-L-carnitine,
melatonin, α-tocopherol, and folic acid while resveratrol
exhibited some limited anti-DENV activity [159]. Organo-
sulfur compounds in garlic were tested against DENV-2
NGC (New Guinea C) virus U937 human macrophage-
like cells and Huh-7 human liver cells. The organosulfur
compounds reduced the levels of proinflammatory cyto-
kines (TNF-α, IL-8 and IL-10) and affect the oxidative
stress response [160].
The methanol extract of Rumex dentatus showed the
highest antiviral efficacy by inhibiting DENV-2 replication,
with IC
50
of 0.154 and 0.234 μg/mL, while gallic acid showed
with IC
50
of 0.191 μg/mL and 0.522 μg/mL at 45 and 90 PFU
of DENV-2 infection, respectively [161].
Naringenin (citrus flavanone) was evaluated against den-
gue viruses (serotypes 1–4) in Huh7.5 cells which impaired
virus replication in human cells with IC50 =35:81, 17:97,
117:1, and 177:5μM, respectively [162]. Annona muricata
aqueous leave extract was evaluated against dengue virus
type 2. Selectivity index of the extract was found more than
10 against DENV-2 which showed potential as a nature-
based antiviral drug [163]. Three spirotetronate compounds
(2EPS-A, -B, -C) isolated from Actinomadura strain showed
strong DENV-2 NS2B-NS3 protease inhibition with IC
50
values of 1:94 ± 0:18,1:47 ± 0:15, and 2:51 ± 0:21 μg/mL,
respectively [164].
In vitro activity of essential oils of β-caryophyllene was
evaluated against DENV-2. β-caryophyllene acts on the ini-
tial steps of the viral replication cycle and showed inhibition
with IC50 =22:5±5:6μMagainst DENV-2 [165]. The etha-
nol extract of polyherbal formulation Nilavembu kudineer
showed antiviral activity against DENV-2 virus infection in
Vero and human macrophage cell line (THP-1 cells) from
0.78% till 0.01% of the human dose [166].
13BioMed Research International
The aqueous leave extract of Orthosiphon stamineus was
evaluated against DENV-2. The extract exhibited the ability
to reduce DENV-2 replication in the pretreated cell while
ineffective in inhibiting cell death in the posttreated cell
[167]. Antiviral activity of natural alkaloid anisomycin was
evaluated against DENV and ZIKV viruses. The compound
prevented DENV and ZIKV multiplication in human cell
lines, inhibited viral protein expression, and also impaired
viral replication in the posttreated cell. In a lower dose, it
also showed a significant decrease in viremia levels in ZIKV
infected AG129 mice [168]. A natural antimicrobial agent
(latarcin peptide) was evaluated against DENV replication
in infected cells. The peptide exhibited a significant inhibi-
tory effect (IC50 =12:68 ± 3:2μM) against the dengue prote-
ase NS2B-NS3pro at room temperature and also reduced the
viral RNA in a dose-dependent manner [169].
The crude extract of Rhodiola imbricata showed an antiviral
immune response against the dengue virus. It induced inter-
feron (IFN) b and other cytokines and also upregulated MIF-
2a,PKR,andNF-κB phosphorylation in infected cells [170].
The antiviral effect of C. longa extract showed low cytotoxicity
and effective inhibitor (IC50 =17:91 μg/mL)againstDENV-2
infected Huh7it-1 cells [171]. The hydroalcoholic extracts of
leaves and bark of Uncaria guinanensis DENV-2-infected
Huh-7 cells reduced intracellular viral antigen and inhibited
the secretion of viral nonstructural protein [172]. The chemical
compound 5-hydroxy-7-methoxy-6-methylflavanone inhibited
DENV2 infectivity in LLC/MK2 (EC50 =15:99 ± 5:38)aswell
as Vero cell lines (EC50 =12:31 ± 1:64 μM)andDENV4
(EC50 =11:70 ± 6:04 μM). Phospholipase A
2
, a chemical con-
stituent of Crotalus durissus terrificus venom, was inhibited den-
gue virus and yellow fever virus infection in Vero cells, inducing
a partial exposure of genomic RNA through glycerophospholip-
ids cleavage [173]. Tomatidine has inhibited dengue virus
mainly at late stages of infection towards all dengue virus sero-
types and controlled the activating transcription factor 4
(ATF4) expression. It showed inhibition of DENV2
(EC50 =0:82 μM) infection mainly independent of ATF4 [174].
In silico analysis showed that Nimbin is found to be
effective and reducing the morbidity and mortality against
the envelope protein of DENV 1-4 infection [175]. Quinic
acid derivatives were found effective against DENV1-4 and
exhibited impaired dengue virus replication in infected
Huh7.5 cell lines [176]. The antiviral activity of isobutyl gal-
late was evaluated against DENV. Isobutyl gallate exhibited
no cytotoxic activity against Huh 7 and possessed strong
activity (IC50 =4:45 μg/mL against DENV [177]. Gallic acid,
fisetin, quercetin, and catechin inhibited infectious viral par-
ticles production against DENV-2 infected Vero cells
[178–180]. Gedunin was evaluated against the DENV
infected BHK-15 cells. Gedunin showed a significant reduc-
tion (EC50 =10μM). In addition, the molecular docking study
showed the strong interaction of the compound with the ATP/
ADP binding site of the host protein (Hsp90) [181].
The study of thrombocytopenia (≤30,000/μl) in adult
dengue infections, leaves of Carica papaya extract, was
enhanced platelet counts, TNFα, and IFNγlevels while it
reduced IL-6 levels in patients [182]. In another pilot study
was done in Srilanka, two doses of leaves extract at 8 h of
intervals were found an increase in total WBC and platelet
count within 24 h of treatment [183]. The 25 mL leave juice
of C. papaya (two times a day for 5 days) was found an
increase in total WBC and platelet count after 2 days of drug
administration in Pakistani patients [124].
The aqueous leave extract of C. papaya was tested
against DENV-infected THP-1 cells and its role on platelet
augmentation. The leave extract was facilitated in platelet
augmentation and showed antidengue activity by a signifi-
cant reduction in the envelope expression, erythrocyte dam-
age, nonstructural (NS1) proteins, lipid peroxidation, and
intracellular viral load. In addition, thrombocytopenic rats
administered with aqueous extract exhibited increased IL-6
and thrombopoietin levels [184]. In Indonesia, hydroalco-
holic leave extract of C. papaya was given in capsule form
to the patient having a thrombocyte count of <150,000/μL
along with 20% hematocrit. The recovery of patients was
found faster via speedy increase platelets levels [185].
The leave extract of Hippophae rhamnoides was tested in
DENV-2 infected BHK-21 cells. The extract was found
potential antidengue activity by sustaining the cell viability
in infected cells, decreasing TNF-αand increasing IFN-γ
levels [186]. Aqueous extract of the Scutellaria baicalensis
roots was tested against DENV 1-4 serotype-infected Vero
cells. The extract exhibited strong virus replication property
(IC50 =74:33 to 95.83 μg/mL for DENV 1-4 serotypes)
[187]. The aqueous extract of Solanum villosum green
berries showed the highest mortality against Stegomyia
aegypti [154]. The silver nanoparticles (AgNPs) were synthe-
sized from Holarrhena antidysenterica bark extract which
showed strong larvicidal activity (LC50 =9:3 ppm) as com-
pared with other organic solvents and aqueous extract alone
[188]. The synthesized AgNPs from Gmelina asiatica leave
extracts showed potent larvicidal activity
(LC50 =22:44 μg/mL) against Anopheles stephensi lar-
vae [189].
4.3. Chemicobiological Data. Isolated compounds of medici-
nal plants and/or their derivatives may be one of the poten-
tial tools for the treatment of DENV and act against the
vectors [190], such as essential oils [191], polyphenols
[192], flavonoids [140], alkaloids [193], glycosides [194],
and tannins [195].
The antiviral mechanism of natural compounds inhibit-
ing viral entry and replication is shown in Figure 6.
Infection of virus involves various stages:
(i) In the initial steps, DENV binds to cell receptors
including mannose-binding receptor (MR) and
DC-SIGN (dendritic cell-specific ICAM3 grabbing
nonintegrin) receptor present at the surface of the
cell, followed by fusion and entry
(ii) Clathrin-mediated endocytosis and transport of
DENV take place along with pH-dependent fusion
with endocytosis
(iii) The genomic ssRNA (positive-sense) is translated
hooked on a polyprotein, which is smitten into all
proteins
14 BioMed Research International
(iv) Transcription and ribonucleic acid (RNA) replica-
tion occurs at the endoplasmic reticulum (ER)
surface
(v) A synthesized dsRNA genomic virus is taking place
at ER. At the ER, virions bud and are passaged to
the Golgi, where DENV prM (membrane) protein
is cleaved, and virion maturation takes place and is
released by exocytosis
Natural compounds inhibit several proteins involved in
the transcription as well as translation machinery essential
in the DENV life cycle.
Furthermore, natural compounds block the virus repli-
cation by modulating the inflammatory redox-sensitive
pathways and host cell signaling. Details of plant-derived
natural compounds and their antidengue activities are stipu-
lated in Table 2, and their chemical structures have been dis-
played in Figure 7.
Monocyte macrophages are thought to be the principal
target cells for the DENV, the cause of dengue fever and
hemorrhagic fever. Besides Ca
2+
, depletion of Mg
2+
is also
evident during binding of DENV to monocyte macrophages
and cells of T cell and B cell lineages in in vitro studies [8]. It
has been seen that the monocyte-derived macrophages dis-
criminated in the presence of vitamin D3 restrict DENV
infection and moderate the classical inflammatory cytokine
(e.g., TNF-αand IL-1β, -4, and -10) response, where a
reduced surface expression of C-type lectins, including the
mannose receptor [214].
In another report, 1,25(OH)2D3 is evident to suppress
the levels of IL-4 and IL-17A and modulate the levels of
IL-12p70 and IL-10 in DENV infected U937-DC-SIGN cells
and THP-1 macrophages, suggesting an immunomodula-
tory power that can ameliorate inflammation during dengue
infections [196]. These findings have also complied with an
earlier report [217]. In a clinical study, patients (n=64),
received a single dose of 200,000 IU vitamin D, was found
to decrease the risk of dengue fever [220]. A challenge test
done with 10 healthy individuals supplemented with 1000
or 4000 international units (IU)/day of vitamin D during
10 days suggested that 4000 IU/day of vitamin D represents
an adequate dose to control DENV progression and replica-
tion [222].
5. Conclusions and Perspectives
To date, it is not possible to recognize intricate details and
the complexity of the target of DENV of the other suitable
vectors/secondary or hosts for its entrance, production,
transmission, and pathogenesis. Several preventive measures
have been taken; however, still, there is a deficiency of oper-
ative treatment modalities of DENV infections in human
and pet animals. DENV-mediated imbalance of micronutri-
ents may be one of the effective significances of numerous
pathophysiological situations, such as Ca
+2
depletion for
muscle pain, irregular heartbeats, muscle weakness, fatigue,
painful signs and symptoms, and deficiency of vitamin D
in case of inflammatory conditions. The deficiency of vita-
min D leads to oxidative stress and the inflammatory process
[235], which may contribute to lowering the standard levels
of platelet in patients with DENV. The internal bleeding and
fatigue of the infected patients perform the leading functions
for platelet deficiency.
Taken together, the biochemical markers and immuno-
logical parameters can be considered as important diagnos-
tic tools in DENV infection. Besides preventive measures,
the medicinal plants and their derivatives might be alterna-
tive tools in the treatment of DENV infection. Calcium
along with vitamin D supplements can be used in DENV
infection. More researches are necessary regarding to the
successful diagnosis, prevention, and control of dengue
worldwide.
Data Availability
The data used to support the findings of this study are avail-
able from the corresponding author upon request.
Conflicts of Interest
The authors declare that they have no conflicts of interest.
Acknowledgments
This work was supported by CONICYT PIA/APOYO CCTE
AFB170007.
References
[1] V. H. Ferreira-de-Lima and T. N. Lima-Camara, “Natural
vertical transmission of dengue virus in Aedes aegypti and
Aedes albopictus: a systematic review,”Parasites & Vectors,
vol. 11, no. 1, p. 77, 2018.
[2] M. K. Zahoor, A. Rasul, M. A. Zahoor et al., Dengue fever: A
general perspective. Chapter 1, IntechOpen, 2018.
[3] WHO, Dengue and Severe Dengue, World Health Organiza-
tion (WHO) Report, 2019.
[4] A. Wilder-Smith, E. E. Ooi, O. Horstick, and B. Wills, “Den-
gue,”Lancet, vol. 393, no. 10169, pp. 350–363, 2019.
[5] L. C. Katzelnick, J. M. Fonville, G. D. Gromowski et al., “Den-
gue viruses cluster antigenically but not as discrete sero-
types,”Science, vol. 349, no. 6254, pp. 1338–1343, 2015.
[6] D. Normile, “Tropical medicine. Surprising new dengue virus
throws a spanner in disease control efforts,”Science, vol. 342,
no. 6157, p. 415, 2013.
[7] E. Holmes and S. Twiddy, “The origin, emergence and evolu-
tionary genetics of dengue virus,”Infection, Genetics and Evo-
lution, vol. 3, no. 1, pp. 19–28, 2003.
[8] H. Bielefeldt-Ohmann, M. Meyer, D. R. Fitzpatrick, and J. S.
Mackenzie, “Dengue virus binding to human leukocyte cell
lines: receptor usage differs between cell types and virus
strains,”Virus Research, vol. 73, no. 1, pp. 81–89, 2001.
[9] I. C. dos Reis, G. Gibson, T. Ayllón et al., “Entomo-virological
surveillance strategy for dengue, Zika and chikungunya arbo-
viruses in field-caught Aedes mosquitoes in an endemic
urban area of the Northeast of Brazil,”Acta Tropica,
vol. 197, p. 105061, 2019.
15BioMed Research International
[10] J. P. Messina, O. J. Brady, N. Golding et al., “The current and
future global distribution and population at risk of dengue,”
Nature Microbiology, vol. 4, no. 9, pp. 1508–1515, 2019.
[11] V. P. Waman, S. M. Kasibhatla, M. M. Kale, and U. Kulkarni-
Kale, “Population genomics of dengue virus serotype 4:
insights into genetic structure and evolution,”Archives of
Virology, vol. 161, no. 8, pp. 2133–2148, 2016.
[12] M. Castillo-Méndez and V. Valverde-Garduño, “Aedes
aegypti Immune response and its potential impact on dengue
virus transmission,”Viral Immunology, vol. 33, no. 1, pp. 38–
47, 2020.
[13] A. Wilder-Smith, “The expanding geographic range of den-
gue in Australia,”The Medical Journal of Australia, vol. 215,
no. 4, pp. 171-172, 2021.
[14] N. A. Johari, K. Voon, S. Y. Toh, L. H. Sulaiman, I. K. S. Yap,
and P. K. C. Lim, “Sylvatic dengue virus type 4 in Aedes
aegypti and Aedes albopictus mosquitoes in an urban setting
in peninsular Malaysia,”PLoS Neglected Tropical Diseases,
vol. 13, no. 11, article e0007889, 2019.
[15] I. M. Kramer, A. Kreß, D. Klingelhöfer et al., “Does winter
cold really limit the dengue vector Aedes aegypti in Europe?,”
Parasites & Vectors, vol. 13, no. 1, p. 178, 2020.
[16] R. A. Marinho, E. B. Beserra, M. A. Bezerra-Gusmão, V. D. S.
Porto, R. A. Olinda, and C. A. C. Dos Santos, “Effects of tem-
perature on the life cycle, expansion, and dispersion of Aedes
aegypti (Diptera: Culicidae) in three cities in Paraiba, Brazil,”
Journal of Vector Ecology, vol. 41, no. 1, pp. 1–10, 2016.
[17] M. Hau, R. E. Ricklefs, M. Wikelski, K. A. Lee, and J. D.
Brawn, “Corticosterone, testosterone and life-history strate-
gies of birds,”Proceedings of the Royal Society B: Biological
Sciences, vol. 277, no. 1697, pp. 3203–3212, 2010.
[18] J. Gaburro, P. N. Paradkar, M. Klein, A. Bhatti, S. Nahavandi,
and J. B. Duchemin, “Dengue virus infection changes Aedes
aegypti oviposition olfactory preferences,”Scientific Reports,
vol. 8, no. 1, pp. 1–11, 2018.
[19] K. A. Hanley, T. P. Monath, S. C. Weaver, S. L. Rossi, R. L.
Richman, and N. Vasilakis, “Fever versus fever: the role of
host and vector susceptibility and interspecific competition
in shaping the current and future distributions of the sylvatic
cycles of dengue virus and yellow fever virus,”Infection,
Genetics and Evolution, vol. 19, pp. 292–311, 2013.
[20] A. D. Haddow, H. Guzman, V. L. Popov et al., “First isolation
of Aedes flavivirus in the Western Hemisphere and evidence
of vertical transmission in the mosquito Aedes (Stegomyi)
albopictus (Diptera: Culicidae),”Virology, vol. 440, no. 2,
pp. 134–139, 2013.
[21] S. Thongyuan and P. Kittayapong, “First evidence of dengue
infection in domestic dogs living in different ecological set-
tings in Thailand,”PLoS One, vol. 12, no. 8, article
e0180013, 2017.
[22] J. L. Ramirez and G. Dimopoulos, “The Toll immune signal-
ing pathway control conserved anti-dengue defenses across
diverse _Ae. aegypti_ strains and against multiple dengue
virus serotypes,”Developmental and Comparative Immunol-
ogy, vol. 34, no. 6, pp. 625–629, 2010.
[23] Z. Xi, J. L. Ramirez, and G. Dimopoulos, “The Aedes aegypti
toll pathway controls dengue virus infection,”PLoS Patho-
gens, vol. 4, no. 7, article e1000098, 2008.
[24] N. Luplertlop, P. Surasombatpattana, S. Patramool et al.,
“Induction of a peptide with activity against a broad spec-
trum of pathogens in the Aedes aegypti salivary gland, follow-
ing infection with dengue virus,”PLoS Pathogens, vol. 7,
no. 1, article e1001252, 2011.
[25] J. A. Souza-Neto, S. Sim, and G. Dimopoulos, “An evolution-
ary conserved function of the JAK-STAT pathway in anti-
dengue defense,”Proceedings of the National Academy of Sci-
ences of the United States of America, vol. 106, no. 42,
pp. 17841–17846, 2009.
[26] N. J. Caplen, Z. Zheng, B. Falgout, and R. A. Morgan, “Inhi-
bition of viral gene expression and replication in mosquito
cells by dsRNA-triggered RNA interference,”Molecular
Therapy, vol. 6, no. 2, pp. 243–251, 2002.
[27] M. Hussain, T. Walker, S. L. O'Neill, and S. Asgari, “Blood
meal induced microRNA regulates development and
immune associated genes in the Dengue mosquito vector,
_Aedes aegypti_,”Insect Biochemistry and Molecular Biology,
vol. 43, no. 2, pp. 146–152, 2013.
[28] X. Liu, C. Guo, Y. Huang, X. Zhang, and Y. Chen, “Inhibition
of porcine reproductive and respiratory syndrome virus by
Cecropin D in vitro,”Infection, Genetics and Evolution,
vol. 34, pp. 7–16, 2015.
[29] P.-S. Yen, A. James, J. C. Li, C. H. Chen, and A. B. Failloux,
“Synthetic miRNAs induce dual arboviral-resistance pheno-
types in the vector mosquito Aedes aegypti,”Communications
Biology, vol. 1, no. 1, p. 11, 2018.
[30] P. Miesen, A. Ivens, A. H. Buck, and R. P. van Rij, “Small RNA
profiling in dengue virus 2-infected Aedes Mosquito cells
reveals viral piRNAs and novel host miRNAs,”PLoS Neglected
Tropical Diseases, vol. 10, no. 2, article e0004452, 2016.
[31] J. C. Scott, D. E. Brackney, C. L. Campbell et al., “Comparison of
dengue virus type 2-specific small RNAs from RNA
interference-competent and -incompetent mosquito cells,”PLoS
Neglected Tropical Diseases,vol.4,no.10,articlee848,2010.
[32] Y. Wang, B. Jin, P. Liu, J. Li, X. Chen, and J. Gu, “piRNA pro-
filing of dengue virus type 2-infected Asian Tiger mosquito
and midgut tissues,”Viruses, vol. 10, no. 4, p. 213, 2018.
[33] X. Wu, H. Hong, J. Yue et al., “Inhibitory effect of small inter-
fering RNA on dengue virus replication in mosquito cells,”
Virology Journal, vol. 7, no. 1, pp. 270–270, 2010.
[34] U. Palatini, P. Miesen, R. Carballar-Lejarazu et al., “Compar-
ative genomics shows that viral integrations are abundant
and express piRNAs in the arboviral vectors Aedes aegypti
and Aedes albopictus,”BMC Genomics, vol. 18, no. 1,
p. 512, 2017.
[35] S. Sim, N. Jupatanakul, J. L. Ramirez et al., “Transcriptomic
profiling of diverse Aedes aegypti strains reveals increased
basal-level immune activation in dengue virus-refractory
populations and identifies novel virus-vector molecular inter-
actions,”PLoS Neglected Tropical Diseases, vol. 7, no. 7, article
e2295, 2013.
[36] M. M. Choy, O. M. Sessions, D. J. Gubler, and E. E. Ooi, “Pro-
duction of infectious dengue virus in Aedes aegypti is depen-
dent on the ubiquitin proteasome pathway,”PLoS Neglected
Tropical Diseases, vol. 9, no. 11, article e0004227, 2015.
[37] A. Troupin, B. Londono-Renteria, M. J. Conway et al., “A
novel mosquito ubiquitin targets viral envelope protein for
degradation and reduces virion production during dengue
virus infection,”Biochimica et Biophysica Acta, vol. 1860,
no. 9, pp. 1898–1909, 2016.
[38] W. C. T. Black, K. E. Bennett, N. Gorrochótegui-Escalante
et al., “Flavivirus Susceptibility in Aedes aegypti,”Archives
of Medical Research, vol. 33, no. 4, pp. 379–388, 2002.
16 BioMed Research International
[39] N. Kato, C. R. Mueller, J. F. Fuchs et al., “Evaluation of the
function of a type I peritrophic matrix as a physical barrier
for midgut epithelium invasion by mosquito-borne patho-
gens in Aedes aegypti,”Vector Borne and Zoonotic Diseases,
vol. 8, no. 5, pp. 701–712, 2008.
[40] I. Sánchez-Vargas, J. C. Scott, B. K. Poole-Smith et al., “Den-
gue virus type 2 infections of Aedes aegypti are modulated by
the mosquito's RNA interference pathway,”PLoS Pathogens,
vol. 5, no. 2, article e1000299, 2009.
[41] J. Ramos-Castañeda, C. González, M. A. Jiménez et al., “Effect
of nitric oxide on dengue virus replication in <i>Aedes
aegypti</i>and <i>Anopheles</i><i>albimanus</i>,”
Intervirology, vol. 51, no. 5, pp. 335–341, 2009.
[42] J. L. Ramirez, J. Souza-Neto, R. T. Cosme et al., “Reciprocal
tripartite interactions between the Aedes aegypti midgut
microbiota, innate immune system and dengue virus influ-
ences vector competence,”PLoS Neglected Tropical Diseases,
vol. 6, no. 3, article e1561, 2012.
[43] J. L. Ramirez, S. M. Short, A. C. Bahia et al., “Chromobacter-
ium Csp_P reduces malaria and dengue infection in vector
mosquitoes and has entomopathogenic and in vitro anti-
pathogen activities,”PLoS Pathogens, vol. 10, no. 10, article
e1004398, 2014.
[44] Y. I. Angleró-Rodríguez, O. A. C. Talyuli, B. J. Blumberg
et al., “An Aedes aegypti-associated fungus increases suscep-
tibility to dengue virus by modulating gut trypsin activity,”
eLife, vol. 6, 2017.
[45] S. Thomas, J. Verma, M. Woolfit, and S. L. O’Neill, “Wolba-
chia-mediated virus blocking in mosquito cells is dependent
on XRN1-mediated viral RNA degradation and influenced
by viral replication rate,”PLoS Pathogens, vol. 14, no. 3, arti-
cle e1006879, 2018.
[46] X. Pan, G. Zhou, J. Wu et al., “Wolbachia induces reactive
oxygen species (ROS)-dependent activation of the toll path-
way to control dengue virus in the mosquito Aedes aegypti,”
Proceedings of the National Academy of Sciences of the United
States of America, vol. 109, no. 1, pp. E23–E31, 2012.
[47] S. Asad, R. Parry, and S. Asgari, “Upregulation of Aedes
aegypti Vago1 by Wolbachia and its effect on dengue virus
replication,”Insect Biochemistry and Molecular Biology,
vol. 92, pp. 45–52, 2018.
[48] X. Xiao, Y. Liu, X. Zhang et al., “Complement-related pro-
teins control the flavivirus infection of Aedes aegypti by
inducing antimicrobial peptides,”PLoS Pathogens, vol. 10,
no. 4, article e1004027, 2014.
[49] E. J. Houk, J. L. Hardy, S. B. Presser, and L. D. Kramer, “Dis-
semination barriers for western equine encephalomyelitis
virus in Culex tarsalis infected after ingestion of low viral
doses,”The American Journal of Tropical Medicine and
Hygiene, vol. 30, no. 1, pp. 190–197, 1981.
[50] P. R. Grimstad, S. L. Paulson, and G. B. Craig Jr., “Vector
competence of Aedes hendersoni (Diptera: Culicidae) for La
Crosse virus and evidence of a salivary-gland escape barrier,”
Journal of Medical Entomology, vol. 22, no. 4, pp. 447–453,
1985.
[51] G. K. Christophides, E. Zdobnov, C. Barillas-Mury et al.,
“Immunity-related genes and gene families in Anopheles
gambiae,”Science, vol. 298, no. 5591, pp. 159–165, 2002.
[52] M. Hoshino, F. Matsuzaki, Y.-i. Nabeshima, and C. Hama,
“hikaru genki, a CNS-specific gene identified by abnormal
locomotion in drosophila, encodes a novel type of protein,”
Neuron, vol. 10, no. 3, pp. 395–407, 1993.
[53] A. B. Barletta, L. R. Alves, M. C. L. Nascimento Silva et al.,
“Emerging role of lipid droplets in Aedes aegypti immune
response against bacteria and Dengue virus,”Scientific
Reports, vol. 6, no. 1, p. 19928, 2016.
[54] J. C. Molloy, U. Sommer, M. R. Viant, and S. P. Sinkins,
“Wolbachia modulates lipid metabolism in Aedes albopictus
Mosquito cells,”Applied and Environmental Microbiology,
vol. 82, no. 10, pp. 3109–3120, 2016.
[55] M. Wu, L. V. Sun, J. Vamathevan et al., “Phylogenomics of
the reproductive parasite Wolbachia pipientis wMel: a
streamlined genome overrun by mobile genetic elements,”
PLoS Biology,vol. 2, no. 3, p. E69, 2004.
[56] N. B. Tjaden, S. M. Thomas, D. Fischer, and C. Beierkuhnlein,
“Extrinsic incubation period of dengue: knowledge, backlog,
and applications of temperature dependence,”PLoS
Neglected Tropical Diseases, vol. 7, no. 6, 2013.
[57] V. Huerta, Y. Ramos, A. Yero et al., “Novel interactions of
domain III from the envelope glycoprotein of dengue 2 virus
with human plasma proteins,”Journal of Proteomics, vol. 131,
pp. 205–213, 2016.
[58] C. Cruz-Oliveira, J. M. Freire, T. M. Conceição, L. M. Higa,
M. A. R. B. Castanho, and A. T. da Poian, “Receptors and
routes of dengue virus entry into the host cells,”FEMS Micro-
biology Reviews, vol. 39, no. 2, pp. 155–170, 2015.
[59] K. I. Hidari and T. Suzuki, “Dengue virus receptor,”Medicine
& Health, vol. 39, 4SUPPLEMENT, pp. S37–S43, 2011.
[60] N. Nanaware, A. Banerjee, S. M. Bagchi, P. Bagchi, and
A. Mukherjee, “Dengue virus infection: a tale of viral exploi-
tations and host responses,”Viruses, vol. 13, no. 10, p. 1967,
2021.
[61] Y. Mori, T. Okabayashi, T. Yamashita et al., “Nuclear local-
ization of Japanese encephalitis virus core protein enhances
viral replication,”Journal of Virology, vol. 79, no. 6,
pp. 3448–3458, 2005.
[62] W.-k. Oh and J. Song, “Hsp70 functions as a negative regula-
tor of West Nile virus capsid protein through direct interac-
tion,”Biochemical and Biophysical Research
Communications, vol. 347, no. 4, pp. 994–1000, 2006.
[63] R. Bhuvanakantham, M. K. Chong, and M. L. Ng, “Specific
interaction of capsid protein and importin-α/βinfluences
West Nile virus production,”Biochemical and Biophysical
Research Communications, vol. 389, no. 1, pp. 63–69, 2009.
[64] K. A. Hanley, J. J. Lee, J. E. Blaney Jr., B. R. Murphy, and S. S.
Whitehead, “Paired charge-to-alanine mutagenesis of dengue
virus type 4 NS5 generates mutants with temperature-sensi-
tive, host range, and mouse attenuation phenotypes,”Journal
of Virology, vol. 76, no. 2, pp. 525–531, 2002.
[65] R. J. Kuhn, W. Zhang, M. G. Rossmann et al., “Structure of
dengue virus: implications for flavivirus organization, matu-
ration, and fusion,”Cell, vol. 108, no. 5, pp. 717–725, 2002.
[66] S. Welsch, S. Miller, I. Romero-Brey et al., “Composition and
three-dimensional architecture of the dengue virus replica-
tion and assembly sites,”Cell Host & Microbe, vol. 5, no. 4,
pp. 365–375, 2009.
[67] E. G. Westaway, A. A. Khromykh, and J. M. Mackenzie,
“Nascent Flavivirus RNA Colocalized _in Situ_ with
Double-Stranded RNA in Stable Replication Complexes,”
Virology, vol. 258, no. 1, pp. 108–217, 1999.
[68] V. Raquin and L. Lambrechts, “Dengue virus replicates and
accumulates in _Aedes aegypti_ salivary glands,”Virology,
vol. 507, pp. 75–81, 2017.
17BioMed Research International
[69] C. V. Filomatori, M. F. Lodeiro, D. E. Alvarez, M. M. Samsa,
L. Pietrasanta, and A. V. Gamarnik, “A 5' RNA element pro-
motes dengue virus RNA synthesis on a circular genome,”
Genes & Development, vol. 20, no. 16, pp. 2238–2249, 2006.
[70] S.-n. Pattanakitsakul, J. Poungsawai, R. Kanlaya,
S. Sinchaikul, S.-T. Chen, and V. Thongboonkerd, “Associa-
tion of Alix with late endosomal lysobisphosphatidic acid is
important for dengue virus infection in human endothelial
cells,”Journal of Proteome Research, vol. 9, no. 9, pp. 4640–
4648, 2010.
[71] B. Londono-Renteria, A. Troupin, M. J. Conway et al., “Den-
gue virus infection of Aedes aegypti requires a putative cyste-
ine rich venom protein,”PLoS Pathogens, vol. 11, no. 10,
article e1005202, 2015.
[72] T. W. Scott and A. C. Morrison, “Aedes aegypti density and
the risk of dengue-virus. Ecological aspects for application
of genetically modified mosquitoes,”FRONTIS, vol. 2, 2003.
[73] C. S. H. Teo and J. J. H. Chu, “Cellular vimentin regulates
construction of dengue virus replication complexes through
interaction with NS4A protein,”Journal of Virology, vol. 88,
no. 4, pp. 1897–1913, 2014.
[74] S. M. Eick, A. P. Dale, B. McKay et al., “Seroprevalence of
dengue and Zika virus in blood donations: A systematic
review,”Transfusion Medicine Reviews, vol. 33, no. 1,
pp. 35–42, 2019.
[75] R. Kulkarni, D. Tiraki, D. Wani, A. C. Mishra, and V. A. Ara-
nkalle, “Risk of transfusion-associated dengue: screening of
blood donors from Pune, western India,”Transfusion,
vol. 59, no. 2, pp. 458–462, 2019.
[76] A. Guzman and R. E. Istúriz, “Update on the global spread of
dengue,”International Journal of Antimicrobial Agents,
vol. 36, Suppl 1, pp. S40–S42, 2010.
[77] A. Goel, D. Patel, K. Lakhani et al., “Dengue fever-a danger-
ous foe,”Journal Indian Academy of Clinical Medicine,
vol. 5, pp. 247–258, 2004.
[78] K. I. P. J. Hidari, N. Takahashi, M. Arihara, M. Nagaoka,
K. Morita, and T. Suzuki, “Structure and anti-dengue virus
activity of sulfated polysaccharide from a marine alga,”Bio-
chemical and Biophysical Research Communications,
vol. 376, no. 1, pp. 91–95, 2008.
[79] P. D. Uchil and V. Satchidanandam, “Architecture of the Fla-
viviral Replication Complex:,”The Journal of Biological
Chemistry, vol. 278, no. 27, pp. 24388–24398, 2003.
[80] I. Kurane, B. L. Innis, S. Nimmannitya, A. Nisalak, F. A.
Ennis, and A. Meager, “High levels of interferon alpha in
the sera of children with dengue virus infection,”The Amer-
ican Journal of Tropical Medicine and Hygiene, vol. 48,
no. 2, pp. 222–229, 1993.
[81] I. Kurane, B. L. Innis, S. Nimmannitya et al., “Activation of T
lymphocytes in dengue virus infections. High levels of soluble
interleukin 2 receptor, soluble CD4, soluble CD8, interleukin
2, and interferon-gamma in sera of children with dengue,”
The Journal of Clinical Investigation, vol. 88, no. 5,
pp. 1473–1480, 1991.
[82] M. C. Shivanthan and S. Rajapakse, “Dengue and calcium,”
International Journal of Critical Illness and Injury Science,
vol. 4, no. 4, pp. 314–316, 2014.
[83] G. P. Zaloga and B. Chernow, “The multifactorial basis for
hypocalcemia during sepsis. Studies of the parathyroid
hormone-vitamin D axis,”Annals of Internal Medicine,
vol. 107, no. 1, pp. 36–41, 1987.
[84] R. Bravo, V. Parra, D. Gatica et al., “Chapter Five - Endoplas-
mic Reticulum and the Unfolded Protein Response: Dynam-
ics and Metabolic Integration,”in International Review of Cell
and Molecular Biology, K. W. Jeon, Ed., Academic Press,
2013.
[85] N. Meng, X. Ren, G. Santagiuliana et al., “Ultrahigh β-phase
content poly(vinylidene fluoride) with relaxor-like ferroelec-
tricity for high energy density capacitors,”Nature Communi-
cations, vol. 10, no. 1, pp. 1–9, 2019.
[86] P. Bautista-Carbajal, R. Soto-Acosta, A. H. Angel-Ambrocio
et al., “The calmodulin antagonist W-7 (N-(6-aminohexyl)-
5-chloro-1-naphthalenesulfonamide hydrochloride) inhibits
DENV infection in Huh-7 cells,”Virology, vol. 501,
pp. 188–198, 2017.
[87] J. Li, R. Huang, W. Liao, Z. Chen, S. Zhang, and R. Huang,
“Dengue virus utilizes calcium modulating cyclophilin-
binding ligand to subvert apoptosis,”Biochemical and Bio-
physical Research Communications, vol. 418, no. 4, pp. 622–
627, 2012.
[88] R. George and L. Lum, Clinical Spectrum of Dengue Infection.
Dengue and Dengue Haemorrhagic Fever,CAB International,
New York, 1997.
[89] I.-K. Lee, J.-W. Liu, and K. D. Yang, “Fatal dengue hemor-
rhagic fever in adults: emphasizing the evolutionary pre-
fatal clinical and laboratory manifestations,”PLoS Neglected
Tropical Diseases, vol. 6, no. 2, article e1532, 2012.
[90] T. P. Johnson, H. B. Larman, M.‐. H. Lee et al., “Chronic den-
gue virus panencephalitis in a patient with progressive
dementia with extrapyramidal features,”Annals of Neurology,
vol. 86, no. 5, pp. 695–703, 2019.
[91] J. Suppiah, S.-M. Ching, S. Amin-Nordin et al., “Clinical
manifestations of dengue in relation to dengue serotype and
genotype in Malaysia: A retrospective observational study,”
PLoS Neglected Tropical Diseases, vol. 12, no. 9,
p. e0006817, 2018.
[92] F. Giovannoni, M. F. Ladelfa, M. Monte, D. A. Jans,
P. Hemmerich, and C. García, “Dengue non-structural pro-
tein 5 polymerase complexes with promyelocytic leukemia
protein (PML) isoforms III and IV to disrupt PML-nuclear
bodies in infected cells,”Frontiers in Cellular and Infection
Microbiology, vol. 9, pp. 284–284, 2019.
[93] Y.-S. Kao, C.-Y. Yu, H.-J. Huang et al., “Combination of mod-
ified NS1 and NS3 as a novel vaccine strategy against dengue
virus infection,”Journal of Immunology, vol. 203, no. 7,
pp. 1909–1917, 2019.
[94] D. S. Burke, R. M. N. Scott, D. E. Johnson, and A. Nisalak, “A
prospective study of dengue infections in Bangkok,”The
American Journal of Tropical Medicine and Hygiene, vol. 38,
no. 1, pp. 172–180, 1988.
[95] T. P. Endy, S. Chunsuttiwat, A. Nisalak et al., “Epidemiology
of inapparent and symptomatic acute dengue virus infection:
a prospective study of primary school children in Kamphaeng
Phet, Thailand,”American Journal of Epidemiology, vol. 156,
no. 1, pp. 40–51, 2002.
[96] S. Datta and C. Wattal, “Dengue NS1 antigen detection: a
useful tool in early diagnosis of dengue virus infection,”
Indian Journal of Medical Microbiology, vol. 28, no. 2,
pp. 107–110, 2010.
[97] P. Dussart, B. Labeau, G.̀
. Lagathu et al., “Evaluation of an
enzyme immunoassay for detection of dengue virus NS1 anti-
gen in human serum,”Clinical and Vaccine Immunology,
vol. 13, no. 11, pp. 1185–1189, 2006.
18 BioMed Research International
[98] A. Khan, “Frequency and clinical presentation of dengue
fever at tertiary Care Hospital of Hyderabad/Jamshoro,”
Journal of the Liaquat University of Medical and Health Sci-
ences, vol. 9, pp. 88–94, 2010.
[99] D. H. Libraty, P. R. Young, D. Pickering et al., “High circulat-
ing levels of the dengue virus nonstructural protein NS1 early
in dengue illness correlate with the development of dengue
hemorrhagic fever,”The Journal of Infectious Diseases,
vol. 186, no. 8, pp. 1165–1168, 2002.
[100] P.-Y. Shu, L.-K. Chen, S.-F. Chang et al., “Comparison of cap-
ture immunoglobulin M (IgM) and IgG enzyme-linked
immunosorbent assay (ELISA) and nonstructural protein
NS1 serotype-specific IgG ELISA for differentiation of pri-
mary and secondary dengue virus infections,”Clinical and
Diagnostic Laboratory Immunology, vol. 10, no. 4, pp. 622–
630, 2003.
[101] N. Butt, A. Abbassi, S. M. Munir, S. M. Ahmad, and Q. H.
Sheikh, “Haematological and biochemical indicators for the
early diagnosis of dengue viral infection,”Journal of the Col-
lege of Physicians and Surgeons–Pakistan, vol. 18, no. 5,
pp. 282–285, 2008.
[102] D. A. Muller, A. C. I. Depelsenaire, and P. R. Young, “Clinical
and laboratory diagnosis of dengue virus infection,”The Jour-
nal of Infectious Diseases, vol. 215, suppl_2, pp. S89–s95,
2017.
[103] A. Wilder-Smith, “Dengue vaccine development by the year
2020: challenges and prospects,”Current Opinion in Virology,
vol. 43, pp. 71–78, 2020.
[104] N. G. Gratz, “Critical review of the vector status of Aedes
albopictus,”Medical and Veterinary Entomology, vol. 18,
no. 3, pp. 215–227, 2004.
[105] A. Wilder-Smith, “Dengue vaccine development: status and
future,”Bundesgesundheitsblatt - Gesundheitsforschung -
Gesundheitsschutz, vol. 63, no. 1, pp. 40–44, 2020.
[106] G. Clark and M. F. Suárez, “Mosquito vector control and biol-
ogy in Latin America–a second symposium,”Journal of the
American Mosquito Control Association, vol. 8, no. 3,
pp. 305–317, 1992.
[107] N. C. Tan and S. Ho, “Treat-to-target approach in managing
modifiable risk factors of patients with coronary heart disease
in primary care in Singapore: what are the issues?,”Asia
Pacific Family Medicine, vol. 10, no. 1, p. 12, 2011.
[108] U. Thisyakorn and C. Thisyakorn, “Latest developments and
future directions in dengue vaccines,”Therapeutic Advances
in Vaccines, vol. 2, no. 1, pp. 3–9, 2014.
[109] L. E. Yauch and S. Shresta, “Dengue virus vaccine develop-
ment,”Advances in Virus Research, vol. 88, pp. 315–372,
2014.
[110] J. E. Osorio, C. Y.-H. Huang, R. M. Kinney, and D. T. Stinch-
comb, “Development of DENVax: a chimeric dengue-2 PDK-
53-based tetravalent vaccine for protection against dengue
fever,”Vaccine, vol. 29, no. 42, pp. 7251–7260, 2011.
[111] L. M. Schwartz, M. E. Halloran, A. P. Durbin, and I. M. Long-
ini, “The dengue vaccine pipeline: implications for the future
of dengue control,”Vaccine, vol. 33, no. 29, pp. 3293–3298,
2015.
[112] S. Swaminathan and N. Khanna, “Dengue vaccine develop-
ment: global and Indian scenarios,”International Journal of
Infectious Diseases, vol. 84, pp. S80–S86, 2019.
[113] M. A. McArthur, M. B. Sztein, and R. Edelman, “Dengue vac-
cines: recent developments, ongoing challenges and current
candidates,”Expert Review of Vaccines, vol. 12, no. 8,
pp. 933–953, 2013.
[114] N. K. Tripathi and A. Shrivastava, “Recent developments in
recombinant protein–based dengue vaccines,”Frontiers in
Immunology, vol. 9, 2018.
[115] J. Sharifi-Rad, S. Kamiloglu, B. Yeskaliyeva et al., “Pharmaco-
logical activities of psoralidin: a comprehensive review of the
molecular mechanisms of action,”Frontiers in Pharmacology,
vol. 11, 2020.
[116] J. Sharifi-Rad, C. Quispe, J. Herrera-Bravo et al., “A pharma-
cological perspective on plant-derived bioactive molecules for
epilepsy,”Neurochemical Research, vol. 46, no. 9, pp. 2205–
2225, 2021.
[117] D. Tsoukalas, O. Zlatian, M. Mitroi et al., “A novel nutraceu-
tical formulation can improve motor activity and decrease
the stress level in a murine model of middle-age animals,”
Journal of Clinical Medicine, vol. 10, no. 4, p. 624, 2021.
[118] B. Salehi, A. Rescigno, T. Dettori et al., “Avocado-soybean
unsaponifiables: a panoply of potentialities to be exploited,”
Biomolecules, vol. 10, no. 1, p. 130, 2020.
[119] B. Salehi, M. S. Shetty, N. V. Anil Kumar et al., “Veronica
plants-drifting from farm to traditional healing, food applica-
tion, and phytopharmacology,”Molecules, vol. 24, no. 13,
p. 2454, 2019.
[120] T. A. Sani, “Cytotoxic and APOPTOGENIC properties of
DRACOCEPHALUM KOTSCHYI aerial part different frac-
tions on CALU-6 and MEHR-80 lung cancer cell lines,”
Farmácia, vol. 65, pp. 189–199, 2017.
[121] W. L. Jiang, X. L. Luo, and S. J. Kuang, “Effects of Alter-
nanthera philoxeroides Griseb against dengue virus
in vitro,”Di Yi Jun Yi Da Xue Xue Bao, vol. 25, no. 4,
pp. 454–456, 2005.
[122] M. M. Parida, C. Upadhyay, G. Pandya, and A. M. Jana,
“Inhibitory potential of neem (Azadirachta indica Juss) leaves
on Dengue virus type-2 replication,”Journal of Ethnophar-
macology, vol. 79, no. 2, pp. 273–278, 2002.
[123] T. S. Kiat, R. Pippen, R. Yusof, H. Ibrahim, N. Khalid, and
N. A. Rahman, “Inhibitory activity of cyclohexenyl chalcone
derivatives and flavonoids of fingerroot, _Boesenbergia
rotunda_ (L.), towards dengue-2 virus NS3 protease,”Bioor-
ganic & Medicinal Chemistry Letters, vol. 16, no. 12,
pp. 3337–3340, 2006.
[124] N. Ahmad, H. Fazal, M. Ayaz, B. H. Abbasi, I. Mohammad,
and L. Fazal, “Dengue fever treatment with Carica papaya
leaves extracts,”Asian Pacific Journal of Tropical Biomedi-
cine, vol. 1, no. 4, pp. 330–333, 2011.
[125] L. Talarico, C. Pujol, R. Zibetti et al., “The antiviral activity of
sulfated polysaccharides against dengue virus is dependent
on virus serotype and host cell,”Antiviral Research, vol. 66,
no. 2-3, pp. 103–110, 2005.
[126] L. I. C. Tang, A. P. K. Ling, R. Y. Koh, S. M. Chye, and K. G. L.
Voon, “Screening of anti-dengue activity in methanolic
extracts of medicinal plants,”BMC Complementary and
Alternative Medicine, vol. 12, no. 1, 2012.
[127] N. Klawikkan, “Effect of Thai medicinal plant extracts against
dengue virus in vitro,”MU J Pharm, vol. 38, pp. 13–18, 2011.
[128] C. A. Pujol,J. M. Estevez, M. J. Carlucci, M.Ciancia, A. S. Cer-
ezo, and E. B. Damonte, “Novel DL-galactan hybrids from the
red seaweed Gymnogongrus torulosus are potent inhibitors of
herpes simplex virus and dengue virus,”Antiviral Chemistry &
Chemotherapy,vol.13,no.2,pp.83–89, 2002.
19BioMed Research International
[129] R. M. Lopez, R. E. Ocazionez, J. R. Martinez, and E. E. Sta-
shenko, “Inhibitory effect of essential oils obtained from
plants grown in Colombia on yellow fever virus replication
in vitro,”Annals of Clinical Microbiology and Antimicrobials,
vol. 8, no. 1, pp. 8–8, 2009.
[130] P. C. De Sf-Tischer, L. B. Talarico, M. D. Noseda, S. M. Gui-
marães, E. B. Damonte, and M. E. Duarte, “Chemical struc-
ture and antiviral activity of carrageenans from Meristiella
gelidium against herpes simplex and dengue virus,”Carbohy-
drate Polymers, vol. 63, pp. 459–465, 2006.
[131] L. Ono, W. Wollinger, I. M. Rocco, T. L. M. Coimbra, P. A. J.
Gorin, and M.-R. Sierakowski, “In vitro and in vivo antiviral
properties of sulfated galactomannans against yellow fever
virus (BeH111 strain) and dengue 1 virus (Hawaii strain),”
Antiviral Research, vol. 60, no. 3, pp. 201–208, 2003.
[132] S. L. A. Kadir, H. Yaakob, and R. M. Zulkifli, “Potential anti-
dengue medicinal plants: a review,”Journal of Natural Medi-
cines, vol. 67, no. 4, pp. 677–689, 2013.
[133] S. Y. Muliawan, L. S. Kit, S. Devi, O. Hashim, and R. Yusof,
“Inhibitory potential of Quercus lusitanica extract on dengue
virus type 2 replication,”The Southeast Asian Journal of
Tropical Medicine and Public Health, vol. 37, Suppl 3,
pp. 132–135, 2006.
[134] I. Sánchez, F. Gómez-Garibay, J. Taboada, and B. H. Ruiz,
“Antiviral effect of flavonoids on the dengue virus,”Phy-
totherapy Research, vol. 14, no. 2, pp. 89–92, 2000.
[135] M. Srivastava and V. P. Kapoor, “Seed galactomannans: an
overview,”Chemistry & Biodiversity,vol.2,pp.295–317, 2005.
[136] S. R. I. N. Reis, L. M. M. Valente, A. L. Sampaio et al., “Immu-
nomodulating and antiviral activities of _Uncaria tomen-
tosa_ on human monocytes infected with Dengue Virus-2,”
International Immunopharmacology, vol. 8, no. 3, pp. 468–
476, 2008.
[137] C. Rees, J. Costin, R. Fink et al., “In vitro inhibition of dengue
virus entry by _p_ -sulfoxy-cinnamic acid and structurally
related combinatorial chemistries,”Antiviral Research,
vol. 80, no. 2, pp. 135–142, 2008.
[138] R. Rosmalena, B. Elya, B. E. Dewi et al., “The Antiviral Effect
of Indonesian Medicinal Plant Extracts against Dengue Virus
In Vitro and in Silico,”Pathogens, vol. 8, no. 2, p. 85, 2019.
[139] J. G. Haddad, A. C. Koishi, A. Gaudry et al., “Doratoxylon
apetalum, an indigenous medicinal plant from Mascarene
Islands, is a potent inhibitor of Zika and dengue virus infec-
tion in human cells,”International Journal of Molecular Sci-
ences, vol. 20, no. 10, p. 2382, 2019.
[140] E. Clain, J. G. Haddad, A. C. Koishi et al., “The polyphenol-
rich extract from Psiloxylon mauritianum, an endemic
medicinal plant from Reunion Island, inhibits the early stages
of dengue and Zika virus infection,”International Journal of
Molecular Sciences, vol. 20, no. 8, p. 1860, 2019.
[141] X. Yao, Y. Ling, S. Guo et al., “Tatanan A from the _Acorus
calamus L_. root inhibited dengue virus proliferation and
infections,”Phytomedicine, vol. 42, pp. 258–267, 2018.
[142] E. A. Inocente, M. Shaya, N. Acosta, L. H. Rakotondraibe, and
P. M. Piermarini, “A natural agonist of mosquito TRPA1
from the medicinal plant Cinnamosma fragrans that is toxic,
antifeedant, and repellent to the yellow fever mosquito Aedes
aegypti,”PLoS Neglected Tropical Diseases, vol. 12, no. 2, arti-
cle e0006265, 2018.
[143] R. Ishwarya, B. Vaseeharan, R. Anuradha et al., “Eco-friendly
fabrication of Ag nanostructures using the seed extract of
_Pedalium murex_ , an ancient Indian medicinal plant: His-
topathological effects on the Zika virus vector _Aedes
aegypti_ and inhibition of biofilm- forming pathogenic bac-
teria,”Journal of Photochemistry and Photobiology. B,
vol. 174, pp. 133–143, 2017.
[144] F. M. Salinas, L. Vázquez, M. V. Gentilini et al., “Aesculus hip-
pocastanum_ L. seed extract shows virucidal and antiviral
activities against respiratory syncytial virus (RSV) and
reduces lung inflammation _in vivo,”Antiviral Research,
vol. 164, pp. 1–11, 2019.
[145] L. G. Fialho, V. P. da Silva, S. R. N. I. Reis et al., “Antiviral and
Immunomodulatory Effects of Norantea brasiliensis Choisy
on Dengue Virus-2,”Intervirology, vol. 59, no. 4, pp. 217–
227, 2017.
[146] V. D. Dwivedi, I. P. Tripathi, and S. K. Mishra, “In silico eval-
uation of inhibitory potential of triterpenoids from Azadir-
achta indica against therapeutic target of dengue virus,
NS2B-NS3 protease,”Journal of Vector Borne Diseases,
vol. 53, no. 2, pp. 156–161, 2016.
[147] E. E. Ajaegbu, S. P. Danga, I. U. Chijoke, and F. B. Okoye,
“Mosquito adulticidal activity of the leaf extracts of Spondias
mombin L. against Aedes aegypti L. and isolation of active
principles,”Journal of Vector Borne Diseases, vol. 53, no. 1,
pp. 17–22, 2016.
[148] D. Champakaew, A. Junkum, U. Chaithong et al., “Angelica
sinensis (Umbelliferae) with proven repellent properties
against Aedes aegypti, the primary dengue fever vector in
Thailand,”Parasitology Research, vol. 114, no. 6, pp. 2187–
2198, 2015.
[149] S. H. Lee, Y. Q. Tang, A. Rathkrishnan et al., “Effects of cock-
tail of four local Malaysian medicinal plants (Phyllanthus
spp.) against dengue virus 2,”BMC Complementary and
Alternative Medicine, vol. 13, no. 1, 2013.
[150] P. M. Kumar, K. Murugan, K. Kovendan et al., “Mosquitoci-
dal activity of solanum xanthocarpum fruit extract and cope-
pod Mesocyclops thermocyclopoides for the control of
dengue vector Aedes aegypti,”Parasitology Research,
vol. 111, no. 2, pp. 609–618, 2012.
[151] G. Marimuthu, S. Rajamohan, R. Mohan, and
Y. Krishnamoorthy, “Larvicidal and ovicidal properties of
leaf and seed extracts of Delonix elata (L.) gamble (family:
Fabaceae) against malaria (Anopheles stephensi Liston) and
dengue (Aedes aegypti Linn.) (Diptera: Culicidae) vector
mosquitoes,”Parasitology Research, vol. 111, no. 1, pp. 65–
77, 2012.
[152] K. Kovendan, K. Murugan, and S. Vincent, “Evaluation of
larvicidal activity of Acalypha alnifolia Klein ex Willd.
(Euphorbiaceae) leaf extract against the malarial vector,
Anopheles stephensi, dengue vector, Aedes aegypti and Ban-
croftian filariasis vector, Culex quinquefasciatus (Diptera:
Culicidae),”Parasitology Research, vol. 110, no. 2, pp. 571–
581, 2012.
[153] J. Odda, S. Kristensen, J. Kabasa, and P. Waako, “Larvicidal
activity of Combretum collinum Fresen against Aedes
aegypti,”Journal of Vector Borne Diseases, vol. 45, no. 4,
pp. 321–324, 2008.
[154] N. Chowdhury, A. Ghosh, and G. Chandra, “Mosquito Larvi-
cidal Activities Of Solanum Villosum Berry Extract Against
The Dengue Vector Stegomyia Aegypti,”Bmc Complemen-
tary And Alternative Medicine, vol. 8, no. 1, p. 10, 2008.
[155] V. Leardkamolkarn, W. Sirigulpanit, C. Phurimsak,
S. Kumkate, L. Himakoun, and B. Sripanidkulchai, “The
20 BioMed Research International
inhibitory actions of houttuynia cordata aqueous extract on
dengue virus and dengue-infected cells,”Journal of Food Bio-
chemistry, vol. 36, no. 1, pp. 86–92, 2012.
[156] V. M. Quintana, L. E. Piccini, J. D. Panozzo Zénere, E. B.
Damonte, M. A. Ponce, and V. Castilla, “Antiviral activity
of natural and synthetic β-carbolines against dengue virus,”
Antiviral Research, vol. 134, pp. 26–33, 2016.
[157] K. H. Chiow, M. C. Phoon, T. Putti, B. K. H. Tan, and V. T.
Chow, “Evaluation of antiviral activities of Houttuynia cor-
data Thunb. extract, quercetin, quercetrin and cinanserin
on murine coronavirus and dengue virus infection,”Asian
Pacific Journal of Tropical Medicine, vol. 9, no. 1, pp. 1–7,
2016.
[158] Á. Vázquez-Calvo, N. Jiménez de Oya, M. A. Martín-Acebes,
E. Garcia-Moruno, and J. C. Saiz, “Antiviral properties of the
natural polyphenols delphinidin and epigallocatechin gallate
against the Flaviviruses West Nile virus, Zika virus, and den-
gue virus,”Frontiers in Microbiology, vol. 8, pp. 1314–1314,
2017.
[159] A. Paemanee, A. Hitakarun, S. Roytrakul, and D. R. Smith,
“Screening of melatonin, α-tocopherol, folic acid, acetyl-L-
carnitine and resveratrol for anti-dengue 2 virus activity,”
BMC Research Notes, vol. 11, no. 1, p. 307, 2018.
[160] A. Hall, A. Troupin, B. Londono-Renteria, and T. Colpitts,
“Garlic organosulfur compounds reduce inflammation and
oxidative stress during dengue virus infection,”Viruses,
vol. 9, no. 7, p. 159, 2017.
[161] R. Batool, E. Aziz, T. Mahmood, B. K. H. Tan, and V. T. K.
Chow, “Inhibitory activities of extracts ofRumex dentatus,
Commelina benghalensis, Ajuga bracteosa, Ziziphus mauri-
tianaas well as their compounds of gallic acid and emodin
against dengue virus,”Asian Pacific Journal of Tropical Med-
icine, vol. 11, no. 4, p. 265, 2018.
[162] S. Frabasile, A. C. Koishi, D. Kuczera et al., “The citrus flava-
none naringenin impairs dengue virus replication in human
cells,”Scientific Reports, vol. 7, no. 1, pp. 41864–41864, 2017.
[163] N. Z. Wahab, N. Ibrahim, M. K. Kamarudin et al., “Cytotox-
icity and antiviral activity of Annona muricata aqueous leaves
extract against dengue virus type 2,”Journal of Fundamental
and Applied Sciences, vol. 10, pp. 580–589, 2018.
[164] J. Euanorasetr, B. Intra, N. Thunmrongsiri et al., “In vitro
antiviral activity of spirotetronate compounds against dengue
virus serotype 2,”The Journal of General and Applied Micro-
biology, vol. 65, no. 4, pp. 197–203, 2019.
[165] N. P. Castro, M. C. Flechas, R. Ocazionez, E. Stashenko, and
J. Olivero-Verbel, “Potential interaction of components from
essential oils with dengue virus proteins,”Boletin Latinoa-
mericano y del Caribe de Plantas Medicinales y Aromaticas,
vol. 14, pp. 141–155, 2015.
[166] J. Jain, A. Kumar, V. Narayanan et al., “Antiviral activity of
ethanolic extract of _Nilavembu Kudineer_ against dengue
and chikungunya virus through _in vitro_ evaluation,”Jour-
nal of Ayurveda and Integrative Medicine, vol. 11, no. 3,
pp. 329–335, 2020.
[167] N. Z. Wahab, N. Ibrahim, M. K. Kamarudin, F. Lananan,
H. Juahir, and A. Ghazali, “In vitro antiviral activity of Ortho-
siphon stamineus extract against dengue virus type 2,”Journal
of Fundamental and Applied Sciences, vol. 10, pp. 541–551,
2018.
[168] V. M. Quintana, B. Selisko, J. E. Brunetti et al., “Antiviral
activity of the natural alkaloid anisomycin against dengue
and Zika viruses,”Antiviral Research, vol. 176, p. 104749,
2020.
[169] H. A. Rothan, H. Bahrani, N. Rahman, and R. Yusof, “Identi-
fication of natural antimicrobial agents to treat dengue infec-
tion: in vitro analysis of latarcin peptide activity against
dengue virus,”BMC Microbiology, vol. 14, no. 1, p. 140, 2014.
[170] D. Diwaker, K. P. Mishra, L. Ganju, and S. B. Singh, “Rho-
diola inhibits dengue virus multiplication by inducing innate
immune response genes RIG-I, MDA5 and ISG in human
monocytes,”Archives of Virology, vol. 159, no. 8, pp. 1975–
1986, 2014.
[171] M. Ichsyani, A. Ridhanya, M. Risanti et al., “Antiviral effects
ofCurcuma longaL. against dengue virus In Vitro and In
Vivo,”IOP Conference Series: Earth and Environmental Sci-
ence, vol. 101, p. 012005, 2017.
[172] C. da Silva Mello, L. M. M. Valente, T. Wolffet al., “Decrease
in dengue virus-2 infection and reduction of cytokine/che-
mokine production by Uncaria guianensis in human hepato-
cyte cell line Huh-7,”Memórias do Instituto Oswaldo Cruz,
vol. 112, no. 6, pp. 458–468, 2017.
[173] V. D. Muller, R. O. Soares, N. N. dos Santos-Junior et al.,
“Phospholipase A2 isolated from the venom of Crotalus dur-
issus terrificus inactivates dengue virus and other enveloped
viruses by disrupting the viral envelope,”PLoS One, vol. 9,
no. 11, article e112351, 2014.
[174] M. Diosa-Toro, B. Troost, D. van de Pol et al., “Tomatidine, a
novel antiviral compound towards dengue virus,”Antiviral
Research, vol. 161, pp. 90–99, 2019.
[175] P. Lavanya, S. Ramaiah, and A. Anbarasu, “Computational
analysis reveal inhibitory action of nimbin against dengue viral
envelope protein,”Virus, vol. 26, no. 4, pp. 243–254, 2015.
[176] P. R. Zanello, A. C. Koishi, C. . O. Rezende Júnior et al., “Qui-
nic acid derivatives inhibit dengue virus replication in vitro,”
Virology Journal, vol. 12, no. 1, p. 223, 2015.
[177] B. E. Dewi, M. Angelina, F. Nuwwaaridya, H. Desti, and T. M.
Sudiro, “Antiviral activity of isobutyl gallate to dengue virus
serotype 2In vitro,”IOP Conference Series: Earth and Envi-
ronmental Science, vol. 251, p. 012018, 2019.
[178] A. I. Trujillo-Correa, D. C. Quintero-Gil, F. Diaz-Castillo,
W. Quiñones, S. M. Robledo, and M. Martinez-Gutierrez,
“In vitro and in silico anti-dengue activity of compounds
obtained from Psidium guajava through bioprospecting,”
BMC Complementary and Alternative Medicine, vol. 19,
no. 1, p. 298, 2019.
[179] K. Zandi, B. T. Teoh, S. S. Sam, P. F. Wong, M. R. Mustafa,
and S. AbuBakar, “Antiviral activity of four types of bioflavo-
noid against dengue virus type-2,”Virology Journal, vol. 8,
no. 1, p. 560, 2011.
[180] K. Zandi, B. T. Teoh, S. S. Sam, P. F. Wong, M. R. Mustafa,
and S. Abubakar, “Novel antiviral activity of baicalein against
dengue virus,”BMC Complementary and Alternative Medi-
cine, vol. 12, no. 1, p. 214, 2012.
[181] D. Amraiz, N. U. S. S. Zaidi, and M. Fatima, “Antiviral eval-
uation of an Hsp90 inhibitor, gedunin, against dengue virus,”
Tropical Journal of Pharmaceutical Research, vol. 16, no. 5,
pp. 997–1004, 2017.
[182] D. T. Sathyapalan, A. Padmanabhan, M. Moni et al., “Efficacy
& safety of Carica papaya leaf extract (CPLE) in severe
thrombocytopenia (≤30,000/μl) in adult dengue - results of
a pilot study,”PLoS One, vol. 15, no. 2, pp. e0228699–
e0228699, 2020.
21BioMed Research International
[183] S. Hettige, “Salutary effects of Carica papaya leaf extract in
dengue fever patients a pilot study,”Sri Lankan Fam Physi-
cian, vol. 29, pp. 17–19, 2008.
[184] N. Sharma, K. P. Mishra, S. Chanda et al., “Evaluation of anti-
dengue activity of Carica papaya aqueous leaf extract and its
role in platelet augmentation,”Archives of Virology, vol. 164,
no. 4, pp. 1095–1110, 2019.
[185] F. Yunita, E. Hanani, and J. Kristianto, “The effect of Carica
papaya L leaves extract capsule on platelet count and hemat-
ocrit level in dengue fever patient,”International Journal of
Medicinal and Aromatic Plants, vol. 2, pp. 573–578, 2012.
[186] M. Jain, L. Ganju, A. Katiyal et al., “Effect of _Hippophae
rhamnoides_ leaf extract against Dengue virus infection in
human blood-derived macrophages,”Phytomedicine,
vol. 15, no. 10, pp. 793–799, 2008.
[187] K. Zandi, T. H. Lim, N. A. Rahim et al., “Extract of Scutellaria
baicalensis inhibits dengue virus replication,”BMC Complemen-
tary and Alternative Medicine,vol.13,no.1,pp.91–91, 2013.
[188] D. Kumar, G. Kumar, and V. Agrawal, “Green synthesis of
silver nanoparticles using Holarrhena antidysenterica (L.)
Wall.Bark extract and their larvicidal activity against dengue
and filariasis vectors,”Parasitology Research, vol. 117, no. 2,
pp. 377–389, 2018.
[189] U. Muthukumaran, M. Govindarajan, M. Rajeswary, and S. L.
Hoti, “Synthesis and characterization of silver nanoparticles
using Gmelina asiatica leaf extract against filariasis, dengue,
and malaria vector mosquitoes,”Parasitology Research,
vol. 114, no. 5, pp. 1817–1827, 2015.
[190] V. Roa-Linares, Y. Miranda-Brand, V. Tangarife-Castaño
et al., “Anti-herpetic, anti-dengue and antineoplastic activi-
ties of simple and heterocycle-fused derivatives of terpenyl-
1,4-naphthoquinone and 1,4-anthraquinone,”Molecules,
vol. 24, no. 7, p. 1279, 2019.
[191] H.-T. Huang, C.-C. Lin, T.-C. Kuo, S.-J. Chen, and R.-
N. Huang, “Phytochemical composition and larvicidal activ-
ity of essential oils from herbal plants,”Planta, vol. 250,
no. 1, pp. 59–68, 2019.
[192] C. Jasso-Miranda, I. Herrera-Camacho, L. K. Flores-Men-
doza et al., “Antiviral and immunomodulatory effects of poly-
phenols on macrophages infected with dengue virus
serotypes 2 and 3 enhanced or not with antibodies,”Infection
and Drug Resistance, vol. 12, pp. 1833–1852, 2019.
[193] D.-L. Wu, H. J. Li, D. Smith et al., “Polyketides and alkaloids
from the marine-derived fungus dichotomomyces cejpii F31-1
and the antiviral activity of scequinadoline A against dengue
virus,”Marine Drugs, vol. 16, no. 7, p. 229, 2018.
[194] A. M. S. Ahmed, R. A. I. Abou-Elkhair, A. M. el-Torky, and
A. E. A. Hassan, “3-Trifluoromethylpyrazolones derived
nucleosides: synthesis and antiviral evaluation,”Nucleosides,
Nucleotides & Nucleic Acids, vol. 38, no. 8, pp. 590–603, 2019.
[195] S. A. Abdul Ahmad, U. D. Palanisamy, J. J. Khoo, A. Dhanoa,
and S. Syed Hassan, “Efficacy of geraniin on dengue virus
type-2 infected BALB/c mice,”Virology Journal, vol. 16,
no. 1, p. 26, 2019.
[196] Z. Sugiyanto, B. Yohan, S. Hadisaputro et al., “Inhibitory
effect of alpha-mangostin to dengue virus replication and
cytokines expression in human peripheral blood mononu-
clear cells,”Natural Products and Bioprospecting, vol. 9,
no. 5, pp. 345–349, 2019.
[197] M. Tarasuk, P. Songprakhon, P. Chimma, P. Sratongno,
K. Na-Bangchang, and P.-t. Yenchitsomanus, “Alpha-man-
gostin inhibits both dengue virus production and cytokine/
chemokine expression,”Virus Research, vol. 240, pp. 180–
189, 2017.
[198] M. Bourjot, P. Leyssen, C. Eydoux et al., “Chemical constitu-
ents of Anacolosa pervilleana and their antiviral activities,”
Fitoterapia, vol. 83, no. 6, pp. 1076–1080, 2012.
[199] J. S. Y. Low, K. X. Wu, K. C. Chen, M. M.-L. Ng, and J. J. H.
Chu, “Narasin, a novel antiviral compound that blocks den-
gue virus protein expression,”Antiviral Therapy, vol. 16,
no. 8, pp. 1203–1218, 2011.
[200] J. M. Crance, N. Scaramozzino, A. Jouan, and D. Garin,
“Interferon, ribavirin, 6-azauridine and glycyrrhizin: antiviral
compounds active against pathogenic flaviviruses,”Antiviral
Research, vol. 58, no. 1, pp. 73–79, 2003.
[201] L. A. Baltina, Y. T. Tasi, S. H. Huang et al., “Glycyrrhizic acid
derivatives as dengue virus inhibitors,”Bioorganic & Medici-
nal Chemistry Letters, vol. 29, no. 20, article 126645, 2019.
[202] M. Zasloff, A. P. Adams, B. Beckerman et al., “Squalamine as
a broad-spectrum systemic antiviral agent with therapeutic
potential,”Proceedings of the National Academy of Sciences
of the United States of America, vol. 108, no. 38, pp. 15978–
15983, 2011.
[203] A. Rahman, S. Muliawan, N. Rashid, and Y. Muhamad,
“Studies on Quercus Iusitanica Extract on DENV-2 Replica-
tion,”Dengue Bulletin, vol. 30, pp. 260–269, 2006.
[204] M. Bourjot, P. Leyssen, C. Eydoux et al., “Flacourtosides A-F,
phenolic glycosides isolated from Flacourtia ramontchi,”
Journal of Natural Products, vol. 75, no. 4, pp. 752–758, 2012.
[205] D. Laurent, F. Baumann, A. G. Benoit et al., “Structure-activ-
ity relationships of dengue antiviral polycyclic quinones,”The
Southeast Asian Journal of Tropical Medicine and Public
Health, vol. 36, no. 4, pp. 901–905, 2005.
[206] M. Laille, F. Gerald, and C. Debitus, “In vitro antiviral activity
on dengue virus of marine natural products,”Cellular and
Molecular Life Sciences, vol. 54, no. 2, pp. 167–1670, 1998.
[207] G. C. Brandão, E. Kroon, D. Souza, J. Filho, and A. Oliveira,
“Chemistry and antiviral activity of Arrabidaea pulchra
(Bignoniaceae),”Molecules, vol. 18, no. 8, pp. 9919–9932,
2013.
[208] P.-M. Allard, P. Leyssen, M. T. Martin et al., “Antiviral chlo-
rinated daphnane diterpenoid orthoesters from the bark and
wood of Trigonostemon cherrieri,”Phytochemistry, vol. 84,
pp. 160–168, 2012.
[209] X. G. Zhang, P. W. Mason, E. J. Dubovi et al., “Antiviral activ-
ity of geneticin against dengue virus,”Antiviral Research,
vol. 83, no. 1, pp. 21–27, 2009.
[210] K. Whitby, T. C. Pierson, B. Geiss et al., “Castanospermine, a
potent inhibitor of dengue virus infection in vitro and
in vivo,”Journal of Virology, vol. 79, no. 14, pp. 8698–8706,
2005.
[211] F. Jia, G. Zou, J. Fan, and Z. Yuan, “Identification of palma-
tine as an inhibitor of West Nile virus,”Archives of Virology,
vol. 155, no. 8, pp. 1325–1329, 2010.
[212] J. S. Y. Low, K. C. Chen, K. X. Wu, M. M.-L. Ng, and J. J. H.
Chu, “Antiviral activity of emetine Dihydrochloride against
dengue virus infection,”Journal of Antivirals & Antiretrovir-
als, vol. 1, pp. 62–71, 2009.
[213] L. R. Simões, G. M. Maciel, G. C. Brandão, E. G. Kroon, R. O.
Castilho, and A. B. Oliveira, “Antiviral activity of Distictella
elongata (Vahl) Urb. (Bignoniaceae), a potentially useful
source of anti-dengue drugs from the state of Minas Gerais,
22 BioMed Research International
Brazil,”Brazil. Lett Appl Microbiol, vol. 53, no. 6, pp. 602–
607, 2011.
[214] J. F. Arboleda Alzate, I. A. Rodenhuis-Zybert, J. C. Hernán-
dez, J. M. Smit, and S. Urcuqui-Inchima, “Human macro-
phages differentiated in the presence of vitamin D3 restrict
dengue virus infection and innate responses by downregulat-
ing mannose receptor expression,”PLoS Neglected Tropical
Diseases, vol. 11, no. 10, article e0005904, 2017.
[215] M. Jadhav, M. Nayak, S. Kumar et al., “Clinical proteomics
and cytokine profiling for dengue fever disease severity bio-
markers,”Omics: a journal of integrative biology, vol. 21,
no. 11, pp. 665–677, 2017.
[216] P. M. Allard, E. T. H. Dau, C. Eydoux et al., “Alkylated flava-
nones from the bark of Cryptocarya chartacea as dengue
virus NS5 polymerase inhibitors,”Journal of Natural Prod-
ucts, vol. 74, no. 11, pp. 2446–2453, 2011.
[217] H. Puerta-Guardo, S. I. De la Cruz Hernández, V. H. Rosales,
J. E. Ludert, and R. M. del Angel, “The 1α,25-dihydroxy-vita-
min D3 reduces dengue virus infection in human myelomo-
nocyte (U937) and hepatic (Huh-7) cell lines and cytokine
production in the infected monocytes,”Antiviral Research,
vol. 94, no. 1, pp. 57–61, 2012.
[218] T. G. Floore, “Mosquito larval control practices: past and
present,”Journal of the American Mosquito Control Associa-
tion, vol. 22, no. 3, pp. 527–533, 2006.
[219] S. A. Abdul Ahmad, U. D. Palanisamy, B. A. Tejo, M. F.
Chew, H. W. Tham, and S. Syed Hassan, “Geraniin extracted
from the rind of Nephelium lappaceum binds to dengue virus
type-2 envelope protein and inhibits early stage of virus rep-
lication,”Virology Journal, vol. 14, no. 1, p. 229, 2017.
[220] S. Zaman, “Effectiveness of vitamin D in prevention of den-
gue Haemorrhagic fever and dengue shock syndrome,”Jour-
nal of Rawalpindi Medical College, vol. 21, pp. 205–207, 2017.
[221] E. Moghaddam, B.-T. Teoh, S.-S. Sam et al., “Baicalin, a
metabolite of baicalein with antiviral activity against dengue
virus,”Scientific Reports, vol. 4, 2015.
[222] D. M. Giraldo, A. Cardona, and S. Urcuqui-Inchima, “High-
dose of vitamin D supplement is associated with reduced sus-
ceptibility of monocyte-derived macrophages to dengue virus
infection and pro-inflammatory cytokine production: an
exploratory study,”Clinica Chimica Acta, vol. 478, pp. 140–
151, 2018.
[223] M. Raekiansyah, C. C. Buerano, M. A. D. Luz, and K. Morita,
“Inhibitory effect of the green tea molecule EGCG against
dengue virus infection,”Archives of Virology, vol. 163, no. 6,
pp. 1649–1655, 2018.
[224] Y. B. Cheng, J. C. Lee, I. W. Lo et al., “Ecdysones from
Zoanthus spp with inhibitory activity against dengue virus
2,”Bioorganic & Medicinal Chemistry Letters, vol. 26, no. 9,
pp. 2344–2348, 2016.
[225] C. Gómez-Calderón, C. Mesa-Castro, S. Robledo et al., “Anti-
viral effect of compounds derived from the seeds of Mammea
americana and Tabernaemontana cymosa on dengue and
chikungunya virus infections,”BMC Complementary and
Alternative Medicine, vol. 17, no. 1, pp. 57–57, 2017.
[226] M. Raekiansyah, M. Mori, K. Nonaka et al., “Identification of
novel antiviral of fungus-derived brefeldin A against dengue
viruses,”Tropical Medicine and Health, vol. 45, no. 1, 2017.
[227] T. Hishiki, F. Kato, S. Tajima et al., “Hirsutine, an indole alka-
loid of Uncaria rhynchophylla, inhibits late step in dengue
virus lifecycle,”Frontiers in Microbiology, vol. 8, pp. 1674–
1674, 2017.
[228] M. Peng, S. Watanabe, K. W. K. Chan et al., “Luteolin
restricts dengue virus replication through inhibition of the
proprotein convertase furin,”Antiviral Research, vol. 143,
pp. 176–185, 2017.
[229] Y.-H. Wu, C. K. Tseng, H. C. Wu et al., “Avocado (Persea
americana) fruit extract (2R,4R)-1,2,4-trihydroxyheptadec-
16-yne inhibits dengue virus replication via upregulation of
NF-κB-dependent induction of antiviral interferon
responses,”Scientific Reports, vol. 9, no. 1, p. 423, 2019.
[230] J.-C. Lee, F.-R. Chang, S.-R. Chen et al., “Anti-dengue virus
constituents from Formosan Zoanthid Palythoa mutuki,”
Marine Drugs, vol. 14, no. 8, p. 151, 2016.
[231] S. Bharadwaj, K. E. Lee, V. D. Dwivedi et al., “Discovery of
Ganoderma lucidum triterpenoids as potential inhibitors
against Dengue virus NS2B-NS3 protease,”Scientific Reports,
vol. 9, no. 1, article 19059, 2019.
[232] A. C. Nascimento, L. M. M. Valente, M. Gomes et al., “Anti-
viral activity of _Faramea bahiensis_ leaves on dengue virus
type-2 and characterization of a new antiviral flavanone gly-
coside,”Phytochemistry Letters, vol. 19, pp. 220–225, 2017.
[233] N. Sharma, K. P. Mishra, and L. Ganju, “Salidroside exhibits
anti-dengue virus activity by upregulating host innate
immune factors,”Archives of Virology, vol. 161, no. 12,
pp. 3331–3344, 2016.
[234] Y. B. Cheng, Y. T. Chien, J. C. Lee et al., “Limonoids from the
seeds of Swietenia macrophylla with inhibitory activity
against dengue virus 2,”Journal of Natural Products,
vol. 77, no. 11, pp. 2367–2374, 2014.
[235] J. P. Sales de Almeida, L. S. Liberatti, F. E. N. Barros et al.,
“Profile of oxidative stress markers is dependent on vitamin
D levels in patients with chronic hepatitis C,”Nutrition,
vol. 32, no. 3, pp. 362–367, 2016.
23BioMed Research International