Open access peer-reviewed chapter - ONLINE FIRST

Exploring Host Factors of the Human Metabolism as Promising Targets for Dengue Treatment

Written By

Naiara Clemente Tavares, Camila Sales Nascimento, Jaquelline Germano de Oliveira and Carlos Eduardo Calzavara-Silva

Submitted: 09 October 2023 Reviewed: 07 November 2023 Published: 05 January 2024

DOI: 10.5772/intechopen.113902

Viral Infectious Diseases - Annual Volume 2024 IntechOpen
Viral Infectious Diseases - Annual Volume 2024 Authored by Shailendra K. Saxena

From the Annual Volume

Viral Infectious Diseases - Annual Volume 2024 [Working Title]

Prof. Shailendra K. Saxena

Chapter metrics overview

77 Chapter Downloads

View Full Metrics

Abstract

The absence of specific therapy and the challenges posed by currently available palliative drugs, such as paracetamol, underscore the urgent need for targeting medications against dengue. Extensive research in the field of antiviral therapies has primarily focused on investigating viral proteins as potential targets. However, despite these efforts, finding an effective therapy for dengue fever remains a daunting task. Importantly, like all viruses, Dengue virus relies on human host proteins to enable infection. Recognizing this fact has prompted the consideration of host factors as viable targets for intervention strategies to combat the infection. This chapter aims to provide an overview of host-virus interactions during Dengue virus infection, emphasizing the importance of metabolic pathways, as well as molecular and cellular processes such as lipid metabolism, autophagy, apoptosis, and the immune system, which are critical for virus propagation. The main goal here is to expand the list of human factors that could serve as potential drug targets. Additionally, molecules that interact with these factors are explored for their therapeutic potential. This comprehensive exploration of host-virus interactions lays the groundwork for more effective dengue treatments. The molecules highlighted here hold promise as antiviral agents, and their inclusion in repurposing research could expedite the development of therapies for dengue fever.

Keywords

  • dengue virus
  • treatment
  • host factors
  • lipid metabolism
  • autophagy
  • apoptosis
  • immune system
  • drug targets

1. Introduction

Several arthropod-borne viruses, also known as arboviruses, are transmitted between vertebrate hosts and arthropod vectors, and pose a major threat to human health worldwide since they can cause significant morbidity and mortality, making it crucial to combat their spread and protect individuals globally [1, 2]. Most arboviruses causing human disease belong to four families: Flaviviridae, Togaviridae, Peribunyaviridae, and Phenuiviridae [3].

Among them, Dengue virus (DENV)—genus Flavivirus, Flaviviridae family—emerges as the most prevalent arbovirus worldwide. DENV is the primary causative agent responsible for dengue fever, a mosquito-borne viral illness that affects millions of people worldwide. Aedes aegypti and Aedes albopictus mosquitoes are the main vectors for DENV transmission [4, 5]. DENV is grouped into four serotypes (1–4), which display limited cross-protection immunity due to antigenic and genetic differences. Infection with any of these serotypes can result in self-limiting dengue fever or progress to severe dengue hemorrhagic fever and dengue shock syndrome [5].

The structural characteristics of DENV particles have been elucidated through electron microscopy, revealing virions that consist of an electron-dense core enveloped by a lipid bilayer [6]. The DENV genome is comprised of a single-stranded, positive-polarity RNA, spanning approximately 11 kb in length. This RNA encodes a total of ten proteins: three structural proteins, namely core (or capsid), pre-membrane (prM), and envelope (E), responsible for forming the viral particle, and seven nonstructural (NS) proteins, denoted as NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5, which play vital roles in viral genome replication and multiplication [6, 7].

Dengue has attained endemic status in various regions, including the Americas, the African continent, Eastern Mediterranean, South-East Asia, and the Western Pacific. Notably, over the past decade, cases have been reported in some European countries, showing its expanding geographical range [8]. Global records indicate a gradual increase in dengue incident cases, deaths, and disability-adjusted life years (DALYs) from 1990 to 2019, with larger epidemics occurring cyclically every 3 to 5 years [9, 10]. In 2019, the Americas reported the highest number of dengue cases, surpassing 3.1 million [5, 9]. The dengue fever burden is influenced by various factors, including socioeconomic development, climate warming, and population mobility. Research plays a pivotal role in reversing the escalating trend of DENV infection by improving methods, systems, and strategies for its treatment, prevention, and disease control.

To date, there are no antiviral agents or universal vaccines available for the treatment or prevention of dengue. Currently, the treatment for dengue primarily focuses on providing high-quality patient care to alleviate symptoms and minimize complications. Severe cases of dengue affect approximately 500,000 individuals per year and carry a mortality rate of up to 10% for hospitalized patients and 30% for non-hospitalized patients [11]. In cases where patients experience clinical manifestations, the use of paracetamol as an antipyretic and analgesic has been widely recommended [12, 13]. However, it is important to note that although it poses no significant risk at therapeutic doses, paracetamol can be hepatotoxic at high doses, and DENV infection often affects the liver and can lead to acute liver failure with fatal consequences. Then, considering these facts, when hepatocytes are already under stress due to DENV infection, the prolonged use of paracetamol can significantly impact the severity of the disease and exacerbate hepatic dysfunction [14, 15, 16].

Considering the lack of specific therapy, and the challenges associated with paracetamol use, there remains an urgent and imperative need to develop effective medications for dengue [11]. Extensive research in the field of antiviral therapies has explored both structural and nonstructural viral proteins as potential targets for inhibiting virus entry, viral genome replication, and virus maturation. Additionally, current research efforts have also focused on host factors as potential targets for the development of new therapies.

Investigating inhibitors of host processes involved in viral morphogenesis holds promise for the development of valuable therapeutics [17, 18]. However, traditional drug discovery entails a process that can take several years for development and approval. It involves significant time and resources, requiring meticulous research, rigorous testing, and regulatory approval before a new drug can be brought to market [19].

These challenges and issues can be addressed through the strategy of drug repositioning/repurposing, which involves exploring new indications for approved drugs or revitalizing failed drug candidates. This approach offers a promising solution by leveraging existing knowledge and resources to potentially identify alternative therapeutic applications for known drugs [20]. Presently, numerous studies are underway to identify drugs that have the potential to be repurposed for dengue treatment. One of the primary strategies employed involves identifying specific molecular targets associated with DENV multiplication and disease progression. A comprehensive understanding of the virus biology and its interactions with host cells is crucial for this approach [21]. Once these targets are identified, the studies focus on identifying existing drugs that can bind to these targets and regulate their activity, offering potential avenues for therapeutic intervention [22].

This chapter aims to provide a comprehensive overview of the current knowledge of the interaction of DENV with human host factors, as well as the modulation of these host factors during infection. The host factors discussed in this chapter play essential roles in lipid metabolism, the immune system pathways, and the autophagy and apoptosis processes. By identifying these elements, we can explore potential drug targets to develop effective therapeutics for dengue fever. Moreover, this review aims to identify existing molecules that interact with the host factors identified as potential drug targets such as antagonists, agonists, or inhibitors. These molecules could be explored in future studies to enhance our understanding of the role of host factors during infection and to investigate their potential as antiviral agents.

Advertisement

2. Dengue virus infection mechanisms and therapeutic implications

Dengue virus infection begins when the virus particle is released from the salivary gland of female mosquito vectors via saliva into the skin of a mammalian host [23]. DENV binds to a variety of host cell receptors, infecting a diverse cell type that includes but is not restricted to epithelial cells, fibroblast, monocytes, macrophages, dendritic cells, B cells, T cells, endothelial cells, and hepatocytes [23, 24]. Although there are several human host receptors that mediate DENV entry, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), cluster of differentiation 4 (CD4), heat shock protein 90 (HSP90), heat shock protein 70 (HSP70), mannose receptors, T cell immunoglobin and mucin domain 1 (TIM-1), AXL, Claudin-1, it is known that the viral surface E protein is responsible for interacting with the cell membrane receptors and virus attachment to the cell [23, 25, 26].

The envelope (E) protein is a membrane glycoprotein that is differentially glycosylated according to the DENV serotype and the cell type in which the virus is propagated, being important for humoral immunity. The interaction between the E protein and its receptors initiates a cascade of events that affect both the viral particle and the cell membrane, as well as the cytoskeleton, facilitating the virus entry [24]. DENV entry can occur through two main mechanisms: direct fusion of the virus with the cell membrane or clathrin-dependent endocytosis. The specific mechanism depends on the tropism or infectivity of the virus, which is influenced by the DENV serotype and the host cell type. Once inside the endosomes, which have a mildly acidic pH range of 6.2–6.4, conformational changes occur in the E protein, leading to the release of the viral nucleocapsid into the cytoplasm. Following uncoating, the DENV genome becomes available for translation and replication through the host cell machinery [24, 27, 28].

Then, the DENV RNA genome is transported to the endoplasmic reticulum (ER), where it becomes accessible for two crucial processes: translation to produce a polyprotein and genome replication [23, 24]. It is important to note that the replication of the viral genome is coupled to the translation of the polyprotein, as viral proteins are necessary for RNA replication.

During translation, the viral positive-stranded genomic RNA is utilized as a template to synthesize a polyprotein. This polyprotein is subsequently cleaved by a combination of host cell enzymes and the viral NS3 protease complex, resulting in the production of three structural and seven nonstructural proteins [29, 30, 31]. While structural proteins are constituents of the virion, nonstructural proteins assemble into the replication complex and drive the invagination of the ER membrane forming specialized compartments known as viral replication complexes [23, 32]. The RNA-dependent RNA polymerase (RdRp) activity of the viral NS5 protein, along with the viral protease/helicase NS3 and other viral NS proteins collectively known as the replication complex, catalyzes the enzymatic reaction involved in RNA synthesis [33]. Thus, the replication complex replicates the viral RNA through a negative-stranded RNA intermediate, producing a positive-stranded RNA that is packaged into new nucleocapsids and envelopes, forming immature particles [30, 31] that undergo maturation through the Golgi and trans-Golgi membrane networks and are released into extracellular space from the infected cell [34, 35].

These intricate steps within the viral entry and replication processes offer invaluable insights into the underlying mechanisms of DENV infection, spotlighting the complex interplay between viral and cellular constituents. Likewise, throughout this course and within the context of infection, the virus adeptly modulates several host proteins to facilitate optimal replication, virion production, and successful infection. This comprehensive understanding presents a significant opportunity for the development of focused antiviral strategies aimed at disrupting critical stages of infection and replication. Ultimately, this holds the promise of effectively controlling and managing DENV infections.

In the subsequent sections, we highlighted the involvement of lipid metabolism, autophagy, apoptosis processes, the immune system, and signal transduction pathways in the context of DENV infection. We also explored the complex interplay of DENV with genes and proteins within the host, intricately linked with these pathways. Additionally, we investigated the modulation of genes and proteins within these pathways during infection, thereby enhancing the virus’s propagation. Considering these findings, we pointed out host factors operating within these pathways that warrant closer examination as potential antiviral drug targets. Furthermore, we present a comprehensive exploration of molecules, compounds, and drugs that interact with these host factors. These potential therapeutic agents could serve as valuable tools in future studies aimed at therapeutic development or a deeper investigation into the multifaceted role of host factors in the landscape of DENV infection.

Advertisement

3. The intimate relationship between lipid metabolism and Dengue virus infection

Viruses do not possess the machinery for lipid synthesis, so most of them, such as flaviviruses, depend on and influence the host’s lipid metabolism pathways to obtain energy to complete their multiplication cycle [36, 37]. Indeed, lipid metabolism plays a crucial role in the cellular entry, replication, and assembly of the Dengue virus. Several key molecules and pathways involved in de novo fatty acid (FA) synthesis, lipid droplet formation, cholesterol biosynthesis, and lipoprotein metabolism have been identified as important factors in DENV infection. Understanding the interplay between DENV and lipid metabolism provides valuable insights into potential therapeutic targets and strategies for combating dengue fever. Further research in this field will contribute to unraveling the mechanisms underlying lipid-mediated processes and may lead to the development of novel antiviral interventions.

3.1 Fatty acid synthesis

Multiple studies have provided evidence demonstrating the involvement of molecules associated with de novo fatty acid synthesis in the successful infection of DENV.

De novo fatty acid synthesis occurs via acetyl coenzyme A (acetyl-CoA) carboxylation using adenosine triphosphate (ATP) and reduced nicotinamide adenine dinucleotide phosphate (NADPH). Acetyl-CoA carboxylase (ACC) catalyzes the carboxylation of acetyl-CoA to produce malonyl-CoA [37]. The enzyme fatty acid synthase (FAS) utilizes acetyl-CoA, malonyl-CoA, and NADPH as substrates to catalyze the synthesis of the saturated fatty acid palmitate. FAS can also produce shorter fatty acids such as stearate. Furthermore, the lipids synthesized by FAS are stored in lipid droplets [38].

The inhibition of ACC caused a notable reduction in DENV-2 proliferation both in vitro and in vivo [38, 39]. The NS3 protein interacts with FAS, facilitating the recruitment of this enzyme to sites where cytosolic replication complexes are formed, thereby enabling fatty acid generation. Moreover, DENV-2 infection leads to an upregulation of FAS activity [40]. In addition, Tongluan et al. described that the knockdown of FAS led to a decrease in DENV-2 and DENV-4 production, indicating that DENV exploits the fatty acid biosynthetic pathway to establish its replication complexes [41].

These reports underscore the crucial role of de novo fatty acid synthesis in DENV infection, revealing potential targets for antiviral strategies.

3.2 Lipid droplets

It is known that lipid droplets (LDs) have an important role in the process of de novo fatty acid synthesis. LDs are organelles that originate from the endoplasmic reticulum (ER) through the accumulation of neutral lipids such as triacylglycerols (TAGs) and sterol esters, which are synthesized by diacylglycerol acyltransferases (DGATs) and cholesterol acyltransferases (ACATs/SOATS), respectively [42, 43].

Studies already demonstrated that DENV infection activates lipophagy, leading to an increase in the release of free fatty acids (FFAs) from LDs. Concurrently, the virus induces β-oxidation, thus promoting energy production [44, 45]. Moreover, transcriptomic analysis showed a downregulation of acat2 in hepatocytes infected with DENV-2 [46].

In addition to lipids, LDs are composed of several proteins, such as seipin, perilipins, fat storage-inducing transmembrane proteins, and ER-shaping proteins [47]. Among these, the perilipins play a critical role in mediating the binding of DENV capsid to LDs, with particular significance attributed to perilipin 3 [48].

Transmembrane Protein 41B (TMEM41B) and Vacuole Membrane Protein 1 (VMP1) are ER-associated proteins that play a crucial role in the mobilization of LDs [49, 50]. TMEM41B- and VMP1-deficient human embryonal kidney cells resulted in the accumulation of lipids, FA and triglyceride decrease, impaired β-oxidation, and a significant reduction in DENV infection across all serotypes. These findings highlight the involvement of TMEM41B and VMP1 in lipid mobilization, β-oxidation, and consequently, in DENV replication [50].

The inhibition of subtilisin kexin isozyme-1/site-1 protease (SKI-1/S1P), a modulator of lipid homeostasis, resulted in decreased formation of LDs and attenuated DENV infection [51]. Likewise, the inhibition of FAS leads to a decrease in LD levels and impairs DENV-2 assembly [52].

Ancient ubiquitous protein 1 (AUP1), a type-III membrane protein, and regulator of LDs serves as a crucial host cofactor involved in DENV assembly and production [45, 53]. AUP1 interacts with NS4A and NS4B, stimulating lipophagy to generate the energy necessary for DENV replication. Notably, the knockdown of aup1 decreases viral replication and significantly impacts virus assembly [45].

Studies documenting the interaction between DENV-2 capsid protein and LDs have demonstrated the pivotal role of this interaction in manipulating specific intracellular ion concentrations, thereby establishing an advantageous environment for viral multiplication. Consequently, this interaction decreases the production of infectious virions [48, 54]. Additionally, individuals infected with DENV exhibit altered levels of LDs [55].

Hence, DENV exploits LDs and their modulators to promote viral infection and replication. Indeed, further studies to investigate the roles of ACATs and DGATs in DENV pathogenesis would contribute to unraveling the mechanisms underlying LD-mediated processes. These investigations can potentially provide valuable insights into additional targets within LD biogenesis.

3.3 Cholesterol biosynthesis

The process of cholesterol biosynthesis not only influences cellular cholesterol levels but also plays a significant role in DENV infection. The connection between cholesterol metabolism and DENV infection is multifaceted, with various key enzymes and pathways serving as potential targets for antiviral strategies. Understanding and manipulating these pathways offer promising avenues for combating dengue and mitigating its severity.

The biosynthesis of cholesterol begins with the conversion of two molecules of acetyl-CoA into acetoacetyl-CoA by ACATs. Subsequently, the condensation of acetoacetyl-CoA and acetyl-CoA, catalyzed by HMG-CoA synthase (HMGCS), forms 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA). HMG-CoA reductase (HMGCR) then synthesizes mevalonate from HMG-CoA. Mevalonate is further converted into activated isoprenes that undergo condensation reactions leading to the formation of squalene. Sequential reactions then transform squalene into cholesterol [56, 57].

It has been reported that hepatocytes infected with DENV-2 exhibit elevated cholesterol levels. The inhibition of the Niemann-Pick C1-Like 1 (NPC1L1) receptor, which is essential for cholesterol uptake, leads to a significant reduction in infection and viral RNA synthesis [58, 59]. Patients diagnosed with dengue fever exhibit lower levels of cholesterol in their blood, which has been associated with an increased risk of developing severe dengue [60].

Blocking cholesterol trafficking in DENV-2-infected cells resulted in the inhibition of cholesterol biosynthesis and a decrease in virus multiplication [61]. The inhibition of sterol regulatory element-binding protein 2 (SREBP2), a regulator of cholesterol biosynthesis, resulted in the downregulation of genes coding for other key enzymes involved in cholesterol biosynthesis, including HMGCS, HMGCR, and low-density lipoprotein receptor (LDLR). This downregulation led to a decrease in cholesterol biosynthesis, which impaired virus yield, genome replication, and viral protein synthesis [62].

The enzyme diphosphomevalonate decarboxylase (MVD) plays a crucial role in cholesterol biosynthesis, and mvd knockdown effectively inhibited endogenous cholesterol production as well as virus multiplication. Additionally, inhibiting key enzymes involved in the cholesterol syntheses pathway, such as HMGCR and squalene synthetase, had a significant impact on virus replication [63].

Furthermore, the inhibition of 17-beta-hydroxysteroid dehydrogenase type 12 (HSD17B12), an enzyme involved in the steroid metabolism pathway, resulted in decreased DENV replication and reduced production of infectious viral particles [64].

3.4 Lipoproteins and apolipoproteins

Triglycerides and cholesterol are transported through the bloodstream in association with proteins known as lipoproteins [65]. Lipoproteins are macromolecular complexes composed of lipids and specific proteins. They consist of a core of neutral lipids, including triglycerides and cholesterol esters, enclosed by phospholipids, free cholesterol, and apolipoproteins [66, 67]. The classes of lipoproteins include chylomicrons, very low-density lipoproteins (VLDL), low-density lipoproteins (LDL), intermediate-density lipoprotein (IDL), high-density lipoproteins (HDL), and lipoprotein (a) (Lp(a)) [66, 68, 69, 70, 71].

Plasma analysis of patients infected with DENV, regardless of the severity of the infection, revealed decreased levels of VLDL [72]. However, contrasting these findings, another study observed a notable increase in VLDL levels in the serum of patients with severe dengue [73]. Previous reports have documented the interaction between the capsid protein of DENV-2 and VLDL [74]. Moreover, patients diagnosed with dengue fever exhibit elevated levels of high-density lipoprotein (HDL) in their serum and lower concentrations of LDL in their plasma [72, 73]. Additionally, the interaction between NS1 and HDL has been documented in the plasma of individuals with dengue fever [75]. Interestingly, fluorescence microscopy analysis has provided evidence that synthetic peptides derived from the DENV envelope colocalize with LDL on the cell surface, indicating that LDL may serve as a mechanism for viral entry [76]. These findings provide valuable insights into the potential role of lipoproteins in the pathogenesis of dengue, highlighting their potential involvement in the early stages of DENV infection and the virus replication cycle.

Apolipoprotein AI (ApoAI) is the primary protein constituent of HDL and plays a crucial role in cellular cholesterol transport [77]. ApoAI is essential for HDL assembly by functioning as the receptor for cholesterol uptake from cells [77, 78]. Several studies have unveiled the involvement of ApoAI, as well as human proteins that interact with ApoAI, in DENV infection. A yeast two-hybrid assay study revealed the interaction between ApoAI and the protein NS2A of DENV-2 [79]. It has been suggested that ApoAI interacts with DENV-2, facilitating the virus’s attachment to host cells and increasing its infectivity in pro-monocytic cells. Coelho et al. demonstrated that NS1 interacts with ApoAI, and infected individuals exhibit decreased levels of this protein, implying that DENV infection reduces ApoAI levels and consequently increases inflammation [80]. Furthermore, the levels of ApoAI proteins are significantly increased in individuals infected with DENV [81]. Conversely, the transcript levels of apoaI and other apolipoproteins have been found to be downregulated in hepatocytes infected with DENV-2 [46]. The differential regulation of apoaI suggests a potential role for these molecules in the host response to DENV infection and may indicate a disruption in lipid metabolism and transport processes. Further investigation into the functional implications of these changes could provide valuable insights into the interplay between DENV and host lipid homeostasis. ApoAI interacts with membrane lipid transporters, including those of the ATP-binding cassette (ABC) family and scavenger receptor class B type 1 (SR-BI), in the extracellular environment to accumulate lipids and facilitate the formation of fully developed HDL particles [82]. Proteins from the ABC family and SR-BI have been implicated in the entry and cell infection of various viruses, including the human immunodeficiency virus (HIV), and hepatitis C virus (HCV) [83, 84, 85, 86]. Additionally, SR-BI has been identified as a cell surface binding receptor for NS1 and appears to mediate viral entry and enhance infectivity in hepatocytes [87, 88].

Regarding other apolipoproteins, interactions have been observed between DENV-2 PrM and the nonstructural proteins NS2A and NS5 with Apolipoprotein AII (ApoAII) and Apolipoprotein B (ApoB), respectively [79, 89]. Furthermore, Apolipoprotein E (ApoE) interacts with DENV protein C [90]. Interestingly, the knockdown of apoe did not lead to a decrease in DENV-infected particle production; however, it did have an impact on the assembly and maturation of HCV [91].

Lipoproteins’ and apolipoproteins’ interactions with DENV provide valued insights into their significant roles in various stages of the infection process, suggesting a multifaceted connection between lipid homeostasis and the virus. Further investigations are required to uncover potential targets for antiviral interventions.

3.5 Lipid rafts

Lipid rafts are microdomains on the cell membrane characterized by the association of cholesterol, sphingolipids, and protein receptors [92, 93].

Analysis of endothelial cells infected with DENV-2 revealed the colocalization of Caveolin 1 (Cav-1) and flotillin 1 (Flt-1), which are lipid raft markers, with NS2B and NS3 proteins. However, this association was found to be stronger with Cav-1 [94].

The protein disulfide isomerase (PDI), which is localized on lipid rafts, interacts with the NS1 protein, and, upon cholesterol inhibition, PDI and lipid raft expression are increased during DENV infection. These data provide evidence for the role of PDI and lipid rafts in DENV cell entry and potentially in its replication [95].

Following the use of lipid raft disruptors, Soto-Acosta et al. reported a decrease in DENV-2 and DENV-4 yield, as well as NS1 secretion, providing further evidence for the involvement of lipid rafts in virus cell entry. An increase in lipid raft concentration was also observed following DENV-4 infection [96]. Additionally, Coelho et al. also demonstrated that DENV infection triggers the accumulation of lipid rafts by activating inflammation through NS1, which facilitates virus attachment. Nevertheless, the induction of lipid rafts can be impeded by the presence of ApoAI [80].

In summary, lipid rafts emerge as crucial microdomains for DENV infection, with their characteristic components, such as cholesterol and sphingolipids, playing pivotal roles.

3.6 Acyl-CoA thioesterases and other lipids

Acyl-CoA thioesterases (ACOTs) are enzymes involved in the regulation of β-oxidation of fatty acids by hydrolyzing acyl-CoA into free fatty acids (FFAs) and coenzyme A [97, 98]. Acyl-CoA thioesterase 2 (ACOT2) and Acyl-CoA thioesterase 7 (ACOT7) have been implicated in DENV-2 replication and infectivity, as acot2 and acot7 knockdown resulted in reduced viral particle number and infectivity. However, the combined knockdown of acot1 and acot2 had the opposite effect, causing an increase in viral replication [99].

High expression of stearoyl-CoA-desaturase (SCD), an enzyme involved in the conversion of saturated fatty acids to monounsaturated fatty acids, has been observed during the early stages of DENV-2 infection. Interestingly, suppression of SCD significantly reduced DENV-2 replication [100, 101].

Inhibition of neutral sphingomyelinase 2 (nSMase2), an enzyme involved in the hydrolysis of sphingomyelin, a key phospholipid, has demonstrated potent antiviral effects against West Nile virus (WNV) and Zika virus (ZIKV) infections, as it led to a reduction in viral yield. However, the involvement of nSMase2 in DENV infection remains to be explored [102].

3.7 Molecules targeting host factors from lipid metabolism

Several host factors involved in lipid metabolism have been identified to interact with specific drugs and compounds such as statins, soraphen-A, orlistat, and hesperetin [103]. Additionally, extensive research has been carried out to investigate the effectiveness of these molecules against DENV.

Statins, a class of HMGCR inhibitors, have been extensively studied to explore their potential in treating dengue fever (Table 1). Soraphen-A, a compound that targets ACC2, demonstrated remarkable efficacy in reducing DENV-2 viremia in infected mice [119]. Orlistat, a FAS inhibitor, exhibited potent antiviral effects by suppressing replication and infection of all DENV serotypes [107]. Hesperetin, an Acyl CoA: cholesterol acyltransferase 1 (ACAT1) inhibitor, effectively interacted with and inhibited the activity of DENV-2 NS2B/NS3 [104].

Host targetMoleculeActionDrugbank IDStudies involving DENV
ABC subfamily A member 1GlyburideInhibitorDB01016
ProbucolInhibitorDB01599
ABC subfamily B member 5BiricodarDB04851
DofequidarInhibitorDB14067
ABC sub-family C member 8GliclazideBinderDB01120
NateglinideInhibitorDB00731
RepaglinideInhibitorDB00912
MitiglinideInhibitorDB01252
GlipizideInhibitorDB01067
TolbutamideInhibitorDB01124
ChlorpropamideInhibitorDB00672
GliquidoneInhibitorDB01251
GlimepirideInducerDB00222
GlymidineInducerDB01382
ABC sub-family C member 9GlyburideModulatorDB01016
NicorandilActivatorDB09220
ABC sub-family G member 2BiricodarDB04851
ACAT1HesperetinInhibitorDB01094[104, 105, 106]
EzetimibeInhibitorDB00973[60]
ACAT2HesperetinInhibitorDB01094[104, 105, 106]
ACC1BiotinCofactorDB00121
ACC2BiotinCofactorDB00122
Soraphen ADB02859[107]
Haloxyfop-PDB07870
ACOT2-UndecanoneDB08688
ApoAICopperDB09130[108, 109]
ZincDB01593[110, 111, 112]
Zinc acetateDB14487
Zinc chlorideInducer; LigandDB14533
Zinc sulfateInducer; LigandDB14548
ApoB and ApoEZinc acetateDB14487
Zinc chlorideAntagonistDB14533
Zinc sulfateAntagonistDB14548
FASCeruleninInhibitorDB01034[45]
OrlistatInhibitorDB01083[42, 107]
HMGCRLovastatinInhibitorDB00227[113, 114, 115, 116]
CerivastatinInhibitorDB00439
SimvastatinInhibitorDB00641[115, 117]
AtorvastatinInhibitorDB01076[115, 117]
RosuvastatinInhibitorDB01098
FluvastatinInhibitorDB01095[64, 117, 118]
PravastatinInhibitorDB00175[117]
MevastatinInhibitorDB06693[64, 118]
PitavastatinInhibitorDB08860
MeglutolDB04377
TAK-475DB05317
NabiximolsStimulatorDB14011
HMGCS(7R,12R,13R)-13-formyl-12,14-dihydroxy-3,5,7-trimethyltetradeca-2,4-dienoic acidDB07740
NPC1L1EzetimibeInhibitorDB00973[60]
PDIRibostamycinDB03615
ArtenimolLigandDB11638
CopperDB09130[110, 111, 112]
ZincDB01593[110, 111, 112]
Zinc acetateDB14487
Zinc chlorideBinderDB14533
Zinc sulfateBinderDB14548
Perilipin-3IdursulfaseDB01271
GalsulfaseDB01279
SR-BIPhosphatidylserineDB00144
SREBP2Omega-3-acid ethyl estersInhibitorDB09539
DoconexentInhibitorDB03756
Omega-3 fatty acidsInhibitorDB11133

Table 1.

Molecules targeting host lipid metabolism.

The table represents existing molecules that target the described host factors from the lipid metabolism, as identified through a search in the DrugBank Database [103]. The table also displays the specific action of each molecule on the targeted host factor, the corresponding DrugBank ID, and references to studies related to the molecule’s interaction with the Dengue virus or their role during the infection.

Nonetheless, numerous ligands associated with other host factors involved in lipid metabolism remain unexplored or require additional investigation. These include SREBP2 inhibitors, apolipoprotein antagonists, and ATP-binding cassette protein inhibitors (Table 1).

Further investigation into these molecules would contribute to a better understanding of their target role during DENV infection and could potentially establish them as promising candidates for dengue fever treatment.

Advertisement

4. Autophagy and apoptosis processes and their potential influence upon infection with Dengue virus

4.1 Autophagy

Autophagy acts as a defense mechanism against viral infections. However, certain viruses, such as DENV, are capable of modulating autophagy-related genes (ATGs) to enhance their own multiplication and facilitate successful infection [105].

Autophagy is a catabolic process that involves the formation of the autophagosome, which sequesters damaged proteins, organelles, and other cellular components. Subsequently, the autophagosome fuses with a lysosome, forming an autolysosome. Within the autolysosome, the captured material is degraded and broken down into smaller molecules and components that can be recycled and reused by the cell [106].

This process is regulated by a set of key proteins coded by ATGs including serine/threonine-protein kinase Unc-51 like autophagy activating kinase 1 (ULK1) Beclin-1, and microtubule-associated protein light-chain 3 (LC3). ULK1 is involved in autophagy initiation, while Beclin-1 plays critical roles in the generation of phosphatidylinositol 3-phosphate (PI3P) and autophagosome formation. LC3 is essential for autophagosome elongation and cargo recognition [108, 109]. For instance, infection with all DENV serotypes stimulates the expression and secretion of Macrophage Migration Inhibitory Factor (MIF) in hepatoma cells, thereby triggering autophagy and subsequently leading to increased replication and infection of DENV [110]. Depletion of atg5 in hepatomas resulted in a significant decrease in the DENV infection rate and the release of viral particles. These particles were found to be associated with LC3 [111].

The employment of mycophenolic acid, a DENV replication inhibitor, which effectively inhibits viral replication by targeting inosine monophosphate dehydrogenase, resulted in the downregulation of several crucial ATGs, including those coding for Unc-51 like autophagy activating kinase 2 (ULK2), Cathepsin D, transmembrane protein 74 (TMEM74), and estrogen receptor protein 1 (ESR1). Additionally, a significant reduction was observed in the expression levels of lipid-conjugated LC3 (an LC3-phospholipid conjugate (LC3-II)) and p62 proteins [112]. The p62 protein serves as an autophagy adaptor protein and is known to associate with LC3 [113, 114]. These findings indicate a direct correlation between the downregulation of autophagy genes and the decrease in viral replication. Furthermore, p62 specifically targets the capsid of DENV-2, facilitating autophagy. Interestingly, the absence of p62 resulted in heightened susceptibility to DENV-2 infection [115].

The serine/threonine-protein kinase known as the mammalian target of rapamycin (mTOR) comprises two distinct complexes called mTOR complex 1 (mTORCl) and mTOR complex 2 (mTORC2). This signaling pathway holds significant importance in various cellular processes, including the modulation of autophagy, which can be suppressed upon its activation [116]. Endothelial cells infected with DENV-2 exhibited reduced levels of mTOR phosphorylation, along with ATG13 and ULK1, while presenting elevated levels of LC3-II [117]. During DENV infection in vitro, mTORC1 becomes dysregulated, and its inhibition leads to an increase in viral replication through the activation of lipophagy [118, 120]. Inhibition of mTOR resulted in a reduction in the levels of the C protein of DENV-2 [115]. Moreover, Carter et al. observed that DENV infection stimulated mTORC2 signaling and that NS5 interacts with both mTORC1 and mTORC2 [121]. These findings imply that the modulation of the mTOR pathway is one potential mechanism through which DENV infection triggers autophagy.

Recently, high-mobility group box 1 (HMGB1) has emerged as a significant host factor involved in mediating autophagy [122]. The knockdown of hmgb1 in lung cells infected with DENV-2 led to decreased expression of NS5 and C proteins, as well as reduced levels of autophagy markers LC3-II, p62, autophagy-related protein 5 (ATG5), and autophagy-related protein 7 (ATG7). Conversely, stimulating HMGB1 resulted in increased LC3-II levels, decreased p62 levels, and enhanced viral production. Furthermore, when hmgb1 was knocked down, Beclin-1 levels exhibited a reduction, whereas Beclin-1 levels increased upon stimulation, indicating that HMGB1 mediates autophagy modulation during DENV infection through the Beclin-1 pathway [123].

In summary, autophagy initially acts as a cellular defense mechanism against viral replication, but the Dengue virus has evolved strategies to manipulate ATGs and exploit this process for its own benefit. The depletion of ATG proteins, downregulation of autophagy genes, dysregulation of the mTOR signaling pathway, and involvement of other host factors like HMGB1 all contribute to the modulation of autophagy during dengue infection.

4.2 Apoptosis

Apoptosis serves as a cellular process responsible for programmed cell death, predominantly executed by caspases [124]. This cellular death pathway can also serve as an antiviral response.

Apoptosis can be induced through either the extrinsic or intrinsic apoptotic pathway. The extrinsic pathway involves the activation of tumor necrosis factor receptor (TNFR) and FAS-ligand (FASL) receptor, leading to the formation of death receptor adaptor proteins that trigger the apoptotic cascade by activating caspase-8, −3, and − 7 [125, 126]. The intrinsic apoptotic pathway, which is regulated by B-cell lymphoma 2 (BCL-2) family proteins, involves the disruption of mitochondrial outer membrane permeabilization (MOMP) and the activation of caspase-9 [127128]. Indeed, viruses employ both pro-apoptotic and anti-apoptotic mechanisms to facilitate their replication and dissemination. Studies have already demonstrated the regulatory role of various DENV proteins.

Dengue virus proteins, C, M, E, NS2B-NS3, and NS5, interact with and modulate apoptosis factors such as death-associated protein 6 (Daxx), receptor-interacting serine/threonine-protein kinase 2 (RIPK2), and nuclear factor kappa B (NF-κB). C protein has been identified as a pro-survival protein, PrM, M, E, NS2A, NS2B, and NS3 have been found to possess pro-apoptotic properties [129, 130].

Apoptosis-linked gene-2-interacting protein X (ALIX) plays a role in vesicular trafficking associated with the initiation of apoptosis [131]. ALX interacts with and colocalizes with the NS3, and when alx is overexpressed, there is a significant increase in viral titer, indicating enhanced viral replication. Conversely, in the absence of alx expression, viral production is significantly reduced, suggesting that ALX plays a critical role in facilitating DENV multiplication [132].

The proteins, such as BCL2-like 1 (BCLXL/BCL2L1), BCL2-like 2 (BCLW/BCL2L2), and Myeloid cell leukemia 1 (MCL1) of the BCL-2 family, play a crucial role in preventing or delaying cell death as anti-apoptotic proteins. They inhibit the activation of pro-apoptotic proteins, thereby preserving cell survival [133]. MCL1 expression undergoes a reduction upon DENV-4 infection. Additionally, bcl2l1-deficient cells demonstrate higher levels of apoptosis, while mcl1-deficient cells do not exhibit such effects. Furthermore, the inhibition of BCL-2, BCL2L2, and BCL2L1 in DENV-4-infected cells results in diminished viral production alongside augmented cell death [134].

Thus, apoptosis serves as an intrinsic antiviral response. However, in the case of dengue infection, the virus employs both pro-apoptotic and anti-apoptotic mechanisms to support its replication and spread. Various DENV proteins interact with apoptosis factors and modulate their activity, promoting either cell survival or cell death. Proteins like ALIX and MCL1 have been implicated in facilitating DENV propagation or preventing apoptosis, respectively. Inhibiting the anti-apoptotic proteins BCL-2, BCLW, and BCLXL has shown promising results in reducing viral production and increasing cell death in DENV-infected cells.

4.3 Autophagy and apoptosis association

Several studies have revealed a compelling and well-established association between autophagy and apoptosis during DENV infection. Inhibition of autophagy in endothelial cells infected with DENV-2 resulted in an increase in apoptosis, while inhibition of apoptosis led to an elevation in autophagy characterized by elevated LC3 levels [135].

Autophagy is induced during early DENV-2 infection, while apoptosis is suppressed. In the late phase, an augmented cleavage of Beclin-1 coincided with an increase in apoptosis. Interestingly, the knockdown of becn1 led to a reduction in LC3-II levels, indicating a decrease in autophagy. This was accompanied by an increase in apoptosis and a subsequent decrease in DENV-2 replication. Notably, the interaction between NS1 and Beclin-1 was observed, which led to a decrease in Beclin-1 cleavage and subsequently increased autophagy. Additionally, inhibiting autophagy using 3-methyladenine (3-MA) in DENV-infected cells resulted in the accumulation of LC3-II and degradation of p62 during early infection and a further elevation in apoptosis in late infection [136]. Moreover, inactivation of mTORC2 signaling results in increased apoptosis and reduced viral replication. These findings indicate that the virus utilizes mTORC2 activation to suppress apoptosis and promote viral replication [121].

X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) was found to be involved in apoptosis induction during DENV-2 infection, whereas Interferon-alpha-inducible protein 6 (IFI6) is known to regulate apoptosis [135, 137]. DENV-2-infected cells that overexpressed xaf1 presented enhanced LC3 protein expression, indicating heightened autophagy. Conversely, DENV-2-infected cells overexpressing ifi6 exhibited lower levels of LC3, suggesting a correlation between autophagy and apoptosis during the infection [135].

Transcriptomic analysis revealed that upregulated genes involved in autophagy and apoptosis were expressed in hepatocytes infected with DENV-2. Among them, the genes encoding the proteins, including caspase-1, −2, and −4, as well as Daxx are already described. In addition, several other genes encoding proteins involved in these processes were also upregulated, for example, immunity-related GTPase family M member 1 (IRGM1), ring finger protein 152 (RNF152), La ribonucleoprotein 1 (LARP1), Endophilin-B1, TANK (TRAF-associated NF-kB)-binding kinase 1 (TBK1), lysosomal-associated membrane protein 2 (LAMP2), and Optineurin (OPTN) [46]. IRGM1 acts as an AMP-activated protein kinase (AMPK) activator, while RNF152 and LARP1 function as downstream regulators of mTORC1 [138, 139]. Endophilin-B1 is involved in the activation of BAX/BAK1 [140]. TBK1 mediates ATG8 and ATG7 phosphorylation and is inactivated by NS2A and NS4B of DENV-1, −2, and −4 [141, 142]. Furthermore, LAMP2 is required for the fusion of autophagosomes with lysosomes and OPTN plays a role in modulating proteins from the LC3 family [142, 143].

Therefore, a dynamic interplay between autophagy and apoptosis is a well-established aspect of DENV infection. Autophagy and apoptosis are intricately linked, with the inhibition of one pathway often resulting in the upregulation of the other. The timing of their activation during DENV-2 infection is distinct, with early autophagy induction and late apoptosis, highlighting the complex temporal regulation of these processes. This network of interactions offers merits further research into the mechanisms of DENV infection and the potential development of antiviral strategies.

4.4 Molecules targeting host factors involved in autophagy and apoptosis processes

Numerous molecules exhibit affinity for host factors involved in autophagy and apoptosis [103] (Table 2). For instance, minocycline, a negative modulator of caspase-1 and -3, demonstrated inhibitory effects on DENV infection and replication [110, 158]. In vitro studies revealed that minocycline effectively inhibits DENV infection by suppressing autophagy induced by MIF. Furthermore, minocycline reduced the secretion of MIF, inhibited the formation of autophagy, and diminished viremia induced by DENV in mice [110].

Host targetMoleculeActionDrugbank IDStudies involving DENV
BCLW/BCL2L2NavitoclaxDB12340
BCLXL-/BCL2L1FostamatinibInhibitorDB12010
Beclin-1/BECN1EstradiolBinderDB00783
Estradiol acetateDB13952
Estradiol benzoateDB13953
Estradiol cypionateDB13954
Estradiol dienanthateDB13955
Estradiol valerateDB13956
Caspase-1MinocyclineNegative modulatorDB01017[125, 144]
PralnacasanDB04875
EmricasanDB05408
VX-765DB05507
1-methyl-3-trifluoromethyl-1H-thieno[2,3-C] pyrazole-5-carboxylic acid (2-mercapto-ethyl)-amideDB07733
Z-Val-Ala-Asp fluoromethyl ketoneDB07744
3-{6-[(8-hydroxy-quinoline-2-carbonyl)-amino]-2-thiophen-2-YL-hexanoylamino}-4-oxo-butyric acidDB07916
Acetylsalicylic acidInhibitor/ DownregulatorDB00945
Caspase-3MinocyclineNegative modulatorDB01017[125, 144]
5-[4-(1-Carboxymethyl-2-Oxo-Propylcarbamoyl)-Benzylsulfamoyl]-2-Hydroxy-Benzoic AcidDB03124
EmricasanDB05408
2-hydroxy-5-(2-mercapto-ethylsulfamoyl)-benzoic acidDB06862
methyl (3S)-3-[(tert-butoxycarbonyl)amino]-4-oxopentanoateDB07696
[N-(3-dibenzylcarbamoyl-oxiranecarbonyl)-hydrazino]-acetic acidDB08229
4-[5-(2-carboxy-1-formyl-ethylcarbamoyl)-pyridin-3-YL]-benzoic acidDB08251
(1S)-2-oxo-1-phenyl-2-[(1,3,4-trioxo-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]ethyl acetateDB08497
(1S)-1-(3-chlorophenyl)-2-oxo-2-[(1,3,4-trioxo-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]ethyl acetateDB08498
N-[3-(2-fluoroethoxy)phenyl]-N′-(1,3,4-trioxo-1,2,3,4-tetrahydroisoquinolin-6-yl) butanediamideDB08499
Glycyrrhizic acidAntagonistDB13751[145, 146, 147]
Acetylsalicylic acidInhibitor/DownregulatorDB00945
Incadronic acidActivatorDB06255
Pamidronic acidActivatorDB00282
PAC-1ActivatorDB13048
TributyrinActivatorDB12709
OleandrinRegulatorDB12843
Caspase-4Incadronic acidActivatorDB06255
Caspase-7FicaDB03384
EmricasanDB05408
Incadronic acidActivatorDB06255
Caspase-8BardoxoloneDB12651[148]
Bryostatin 1InhibitorDB11752
AN-9RegulatorDB05103
Trichostatin AActivatorDB04297
OleandrinRegulatorDB12843
Caspase-9Pamidronic acidActivatorDB00282
Cathepsin DS-MethylcysteineDB02216
1H-Benzimidazole-2-carboxylic acidDB03028
2-MorpholinoethylamineDB03096
5-Amino-6-cyclohexyl-4-hydroxy-2-isobutyl-hexanoic acidDB07542
Cyclohexylmethyl-2,3-Dihydroxy-5-Methyl-HexylamideDB08740
DaxxCalcium citrateAgonistDB11093
Calcium phosphateAgonistDB11348
ZincDB01593[110, 111, 112]
HMGB1Ethyl pyruvateDB05869[149, 150]
ChloroquineInhibitorDB00608[151, 152, 153, 154]
MIF7-Hydroxy-2-Oxo-Chromene-3-Carboxylic Acid Ethyl EsterDB02728
3,4-Dihydroxycinnamic acid (caffeic acid)DB01880[155, 156]
4-Hydroxyphenylpyruvic acidDB07718
3-(4-Hydroxyphenyl)-4,5-Dihydro-5-Isoxazole-acetic acid methyl esterDB07888
4-Hydroxybenzaldehyde O-(cyclohexylcarbonyl)oximeDB08333
3-Fluoro-4-hydroxybenzaldehyde O-(cyclohexylcarbonyl)oximeDB08334
4-Hydroxybenzaldehyde O-(3,3-dimethylbutanoyl) oximeDB08335
6-Hydroxy-1,3-benzothiazole-2-sulfonamideDB08765
mTORCSF1126DB05210
RimiducidLigandDB04974
XL765DB05241
EverolimusInhibitorDB01590
RidaforolimusInhibitorDB06233
TemsirolimusInhibitorDB06287
SirolimusInhibitorDB00877[157]
PimecrolimusPotentiatorDB00337
FostamatinibInhibitorDB12010
GSK-1059615InhibitorDB11962
OleandrinDownregulatorDB12843
RIPK2FostamatinibInhibitorDB12010
SPHK2OpaganibInhibitorDB12764
TBK1FostamatinibInhibitorDB12010
TNFRUrelumabDB12077
TasonerminAgonistDB11626
Brentuximab vedotinBinder; Antibody; RegulatorDB08870
IratumumabAntibody; RegulatorDB05550
Belantamab mafodotinBinderDB15719
Idecabtagene vicleucelBinderDB16665
Ciltacabtagene autoleucelBinderDB16738
TeclistamabAntibody; RegulatorDB16655
ENMD-1198DB05959
ULK1/2FostamatinibInhibitorDB12010

Table 2.

Molecules targeting host proteins involved in autophagy or apoptosis process.

The table represents existing molecules that target the described host factors involved in the autophagy or apoptosis process, as identified through a search in the DrugBank Database [103]. The table also displays the specific action of each molecule on the targeted host factor, the corresponding DrugBank ID, and references to studies related to the molecule’s interaction with the Dengue virus or their role during the infection.

Glycyrrhizic acid also suppresses autophagy in cells infected with DENV-2. This suppression is attributed to the inhibition of HMGB1 and leads to a reduction in DENV-2 replication [123, 159]. Furthermore, glycyrrhizic acid derivatives exhibit antiviral activity against both DENV-1 and DENV-2, decreasing virus yield [160]. Consequently, glycyrrhizic acid has garnered attention as a potent inhibitor of dengue fever, holding promise for potential therapeutic applications. Bardoxolone has demonstrated a decrease in virus infectivity in cells infected with the DENV [160]. Conversely, inhibiting HMGB1 with ethyl pyruvate has been shown to enhance DENV-2 propagation [161, 162].

Additionally, caffeic acid has displayed inhibitory effects on DENV-1 infection [163, 164]. Sirolimus has been proposed as a potential candidate for repurposing in the treatment of dengue [20].

Although tested for their effects on dengue fever, a significant portion of these molecules that target host factors involved in autophagy and apoptosis have not been adequately studied regarding their specific impact on dengue infection. This highlights the necessity for further research to comprehensively evaluate their potential antiviral effects in the context of DENV infection.

Advertisement

5. The interaction between Dengue virus and the human immune system: exploring old and new putative targets

The immune system assumes the initial responsibility for viral clearance. Upon DENV inoculation in the skin by the mosquito vector, Langerhans cells and skin macrophages are the primary targets of viral infection. These infected cells subsequently undergo migration to lymph nodes, facilitating the dissemination of the virus throughout the bloodstream and to various tissues when more cells like monocytes, macrophages, and dendritic cells can also be infected [165, 166].

5.1 Pattern-recognition receptors (PRRs)

The primarily DENV-infected cells express specific pattern-recognition receptors (PRRs) including the antiviral innate immune response receptor RIG-I (retinoic acid-inducible gene I), melanoma differentiation-associated protein 5 (MDA5), and endosomal Toll-like receptor 3 (TLR3). These PRRs play a role in recognizing pathogen-associated molecular patterns (PAMPs) and initiating an immune response against the viral infection. The recognition of viral factors triggers a cascade of events, starting with the activation of mitochondrial antiviral signaling (MAVS), followed by the activation of TANK-binding kinase 1 (TBK1) and IκB kinase-ϵ (IKKϵ). These kinases phosphorylate interferon regulatory factor 3 (IRF3) and interferon regulatory factor 7 (IRF7), leading to the production of type I/III interferons (IFNs), proteins with known antiviral properties, during the initial stages of infection [167, 168]. TLR3 and Toll-like receptor 7 (TLR7) activate the recruitment of TIR-domain-containing adapter-inducing IFNβ (TRIF) and myeloid differentiation primary response gene 88 (MyD88). This recruitment leads to the activation of nuclear factor-kappa B (NF-κB) and IRF3 and IRF7, ultimately resulting in the production of IFNs. Elevated levels of IFNs stimulate the complement system, activate the Janus kinase (Jak)/signal transducer and activator of the transcription (STAT) pathway, and elicit an adaptive immune response, all working in synergy to eliminate the viral infection [168, 169].

The PRRs RIG-I and MDA5 belong to the RIG-I-like receptor (RLR) family, which constitutes a subset of DEAD/H-box helicases [170]. DEAD/H-box helicases serve as viral RNA sensors or co-sensors and play a significant role in immune response modulation. The RLR family consists of multiple members that can act as either pro-viral or antiviral factors, highlighting their diverse functions in viral infections [144, 145].

The knockdown of rig-i and mda5 resulted in a decrease in the production of IFNβ, interferon lambda 1 (IFNL1), interferon lambda 2 (IFNL2), interleukin-27 (IL-27), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor (TNF) during DENV-2 infection [146, 147]. Additionally, the knockdown of rig-i, mda5, and mavs inhibited the expression of CCL2 (macrophage inflammatory protein-1, MCP-1), CCL3 (macrophage inflammatory protein-1α, MIP-1α), and CCL4 (macrophage inflammatory protein-1α, MIP-1β) [147]. Cells infected with DENV-2 and exposed to a RIG-I agonist showed impaired viral infection, while depletion of rig-i increased the infection [148]. Moreover, activation of RIG-I by designed immune-modulating RNAs inhibited DENV-2 infection [149]. Both structural and nonstructural proteins of DENV are recognized for their role in modulating viral infection through the regulation of RLRs. The NS2A and NS2B proteins of DENV-4 were found to negatively regulate rig-i, mda5, mavs, and tbk1, resulting in reduced transcription of ifnβ [150]. Besides, the expression of DENV-2 NS2A led to a decrease in interferon beta (IFN-β) levels by inhibiting RIG-I, whereas the presence of DENV-2 NS2B, NS4A, and NS4B resulted in a reduction of IFN-β expression through inhibition of both RIG-I and MAVS [155]. Regarding structural proteins, studies have demonstrated that DENV-2 prM interacts with MDA5, resulting in the inhibition of IFN-β production [156].

Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) represents another crucial PRR and holds promise as a potential target for dengue therapeutics. It serves as both an attachment factor for DENV and facilitates viral cell entry [157]. The use of DC-SIGN antagonists inhibited viral infection in DENV-2-infected cells [151]. Additionally, blocking DC-SIGN and LDLR has been shown to reduce DENV-2 infection and protect mice from developing illness [152]. Therefore, targeting DC-SIGN holds promise as an effective approach for inhibiting viral infection in dengue therapeutics.

Tripartite motif (TRIM) proteins act as crucial regulators of PRR signaling pathways, modulating RLRs, toll-like receptors (TLRs), DNA sensor cGAS (cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) synthase), and the interferon alpha/beta (IFNα/β) receptors. Consequently, they play a vital role in the host defense against viral infections. TRIM proteins can form complexes with TRIF and stimulator of interferon genes (STING) to potentiate TLR3 and STING antiviral signaling pathways [153]. Notably, overexpression of trim56 and trim69 suppresses DENV-2 propagation in vitro by impairing genome replication, while their knockdown increases viral load [154, 171]. In vivo suppression of trim69 also elevates virus RNA levels and virus titer. Furthermore, TRIM69 colocalizes with DENV-2 NS3 in both in vitro and in vivo assays and with NS2B [171, 172]. Additionally, the knockdown of trim25 increases DENV-2 replication [173, 174]. Therefore, TRIM proteins represent potential antiviral targets for treating dengue fever. Exploring agonists, activators, or their development could be a promising strategy to discover potential drugs. Additionally, the characterization of other proteins from the TRIM family should be conducted to expand our knowledge about these proteins during DENV infections.

The immune response against DENV infection is orchestrated through PRRs, such as RIG-I, MDA5, TLR3, TLR7, DC-SIGN, TRIM, and others. These receptors recognize viral factors and trigger a sequence of events, ultimately leading to the production of interferons and the activation of various immune pathways. The modulation of RLRs by DENV structural and nonstructural proteins further highlights the virus’s strategies to evade host defenses. These multifaceted immune responses to DENV infection present potential targets for antiviral interventions.

5.2 RNA helicases of the DEAD box and related DExD/H proteins

Many studies have elucidated the role of other DEAD/H-box RNA helicases for DENV replication. During DENV-2 infection, ddx3x is upregulated, and its knockdown results in increased virus replication and a decrease in IFN-β transcript levels during initial infection, followed by an increase in late infection. When ddx3x is overexpressed, DENV production is decreased, and the IRF3-responsive PRD (III–I) promoter is activated. Conversely, the suppression of ddx50 and Integrator complex subunit 6 (ints6) leads to a decrease in viral production. These findings suggest that DEAD (Asp-Glu-Ala-Asp) box helicase 3, X-linked (DDX3X) could inhibit DENV replication, while DEAD (Asp-Glu-Ala-Asp) box polypeptide 50 (DDX50) and integrator complex subunit 6 (INTS6) act as pro-viral factors [175].

Like DDX3X, another DEAD-box RNA helicase, DDX25, is also upregulated during DENV-2 infection. The knockdown of ddx25 resulted in impaired DENV replication and an increase in IFN-β transcript levels after DENV-2 infection. As expected, the overexpression of ddx25 led to an increase in viral load and a decrease in ifn-β transcript levels after DENV-2 infection during initial infection, followed by an increase in ifn-β transcript levels in late infection [176]. Therefore, DDX25 appears to have a similar role as DDX3X during DENV-2 infection, acting as an antiviral molecule and playing a crucial role in the regulation of IFN-β pathways during the infection.

Considering the exposed, investigations using agonists of DDX3X and DDX25, as well as antagonists or inhibitors of DDX50 and INTS6, would be useful in identifying potential drug candidates.

5.3 Interferon type I pathways and ubiquitination

Interferon type I pathways are also regulated by ubiquitination, a process catalyzed by ubiquitin-related enzymes such as ubiquitin-conjugating enzyme E2 (UBE2). When the transcript levels of ube2b and ube2j1 are diminished, DENV-2 replication also exhibits a decline. On the other hand, the knockdown of ube2s, ube2u, ube2v1, and ube2i resulted in an increase in DENV-2 replication. During a DENV-2 in vitro infection, an upregulation of ube2j1 expression was observed, and its overexpression resulted in an augmentation of viral replication. Additionally, suppression of ube2j1 resulted in a reduction in ifn-β expression induced by RIG-I-N, tumor necrosis factor receptor-associated factor 3 (TRAF3), and IRF3-5D in DENV-2-infected cells [177]. Collectively, these data suggest that UBE2B and particularly UBE2J1 play a pro-viral role, while ubiquitin-conjugating enzyme 2S (UBE2S), ubiquitin-conjugating enzyme E2U (UBE2U), ubiquitin-conjugating enzyme E2 V1 (UBE2V1), and ubiquitin-conjugating enzyme E2 I (UBE2I) seem to have antiviral roles. However, further studies are required to substantiate these findings.

Throughout DENV-2 infection in vitro, the expression of E3 ubiquitin ligases seven in absentia homolog (SIAH1), SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1), and SMAD-specific E3 ubiquitin protein ligase 2 (SMURF2) is increased, and the knockdown of siah1 and smurf2 resulted in a decrease in DENV-2 titer. Furthermore, during DENV-2 infection in vitro, myd88 expression was diminished, while the knockdown of siah1 induced upregulation of myd88 expression, showing that SIAH1 negatively regulated this TLR signaling adaptor during the infection. Also, the depletion of myd88 did not lead to a reduction in viral titer [178]. So, the inhibition of SIAH1 could maintain or increase MyD88 levels, resulting in a decrease in infection. However, a separate study demonstrated that inhibiting MyD88 using a small molecule resulted in impaired DENV replication [179]. Therefore, investigating whether MyD88 and its modulators, such as SIAH1, act as pro-viral or antiviral factors and validating them as potential targets for antiviral strategies aimed at treating dengue is necessary.

Interferon-regulatory factors (IRFs) also hold promise as potential antiviral targets. In DENV-infected mice, the absence of irf3, irf5, and irf7 prevented severe dengue and promoted survival, whereas irf1-deficient mice succumbed to the infection. Furthermore, the simultaneous depletion of these four genes led to the development of severe disease [180]. These findings suggest that IRF-3, -5, and -7 may contribute to dengue severity. Thus, investigations focused on reducing these IRFs and increasing IRF-1 expressions could offer a promising approach to preventing disease progression to severe cases. Besides, IF16 has been reported to colocalize with DENV-2 NS4B, and the reduction of IF16 activity led to the suppression of genome replication, highlighting its critical role in this process [181].

Individuals with DENV infection exhibit heightened levels of IFN-λ. Additionally, epithelial cells infected with DENV-2 showed increased expression of interferon lambda 1 (IFN-λ1) and interferon lambda (IFN-λ2). The administration of IFN-λ1 and IFN-λ2 induces the inhibition of DENV-2 replication. Notably, this reduction in viral replication is further amplified in the presence of interferon alpha (IFN-α) [182]. Investigations using the bioactive component celastrol have revealed that hepatoma cells infected with all DENV serotypes and exposed to celastrol exhibited an increase in IFN-α production through the JAK-STAT pathway. This induction of the antiviral response was evidenced by the reduction of DENV replication and protein synthesis in hepatoma cells. Celastrol also induced IFN-α production in mice infected with DENV-2, resulting in reduced disease outcome and lethality in these animals [183]. Hence, upregulating interferon alpha (IFN-α) and interferon lambda (IFN-λ) could be a promising strategy to both reduce viral loads and hamper disease progression.

5.4 Complement system and other immune system factors

The complement system comprises diverse proteins that constitute the innate immune response, including the classic, lectin, and alternative pathways. The classical and lectin pathways are activated by pathogen recognition, while the alternative pathway is constitutively active but regulated by the protein factor H (complement factor H (CFH)). Once activated, the complement system initiates an enzymatic cascade that facilitates phagocytosis, lysis of infected cells, and regulation of the inflammatory response. Notably, during viral infections, the complement system plays a significant role in viral clearance, rendering it a promising target for therapy strategies [184, 185].

In individuals infected with DENV, monocytes present in peripheral blood mononuclear cells (PBMCs) exhibit downregulation of complement receptors (CRs) such as cr3 (cd11b) and cr4 (cd11c). Additionally, cd59 is downregulated in monocytes from DENV-infected patients, with cr4 (cd11c) showing more pronounced downregulation in patients with severe disease compared to non-severe cases. In vitro assays have demonstrated that suppressing cr3 in monocytes infected with DENV-2 reduces virus infection, a process mediated by IFN-α and TNF-α [186]. In addition, studies indicated that DENV-2 infection induces an increase in transcript levels of cfh in endothelial cells and monocytes [187].

Both DENV nonstructural and structural proteins also engage with complement system factors. C1q, a pattern-recognition molecule of the classical pathway, binds to recombinant DENV-1 and -2 NS1 and E proteins from all serotypes. Monocytes pre-exposed to C1q exhibited reduced DENV-2 infection, possibly impeding cell entry, as DC-SIGN transcript levels were also decreased [188, 189]. Moreover, NS1 from DENV-2 binds to mannose-binding lectin (MBL), a pattern-recognition molecule of the lectin pathway, and inhibits DENV-2 neutralization mediated by MBL. This suggests that MBL plays a role in impairing DENV virus entry and inhibiting infection, while NS1 binds to MBL to facilitate cell entry [190]. Considering these findings, investigations into compounds that suppress CRs and induce C1q and MBL should be conducted to explore their potential as antiviral drugs.

Regarding the cytokine TGF-β, its levels are elevated during DENV infection. Inhibition of transforming growth factor beta 1 (TGF-β1) in macrophages infected with DENV-2 has been shown to reduce viral load [191, 192]. Accordingly, exploring TGF-β inhibition could provide valuable insights to validate this cytokine as a potential drug target for decreasing viral load and/or assisting in the treatment of the pathogenesis of the disease.

5.5 Molecules targeting host factors from the immune system

As depicted, there are numerous functional investigation studies concerning immune system host factors and DENV infection, but only a limited few explore molecules with affinity for these host factors.

Notably, zinc has been recognized to interact with factor H, and research has indicated that zinc chelation and zinc nanoparticles exhibit anti-DENV-2 activity [193, 194].

Investigations have demonstrated the anti-DENV-2 effects of IFN-λ inhibitors in vitro [195]. Promisingly, there exist inhibitors of TBK1 and TGF-β1 that warrant exploration as potential anti-DENV candidates (Table 3). Additionally, agonists of IFN-λ, TRIM13, and TGF-β1 hold the potential to enrich our understanding of the interactions between these proteins and DENV infection (Table 3).

Host targetMoleculeActionDrugbank IDStudies involving DENV
C1qEtanerceptLigandDB00005
Factor H (CFH)CopperDB09130
ZincDB01593[125, 144]
Zinc chlorideLigandDB14533
Zinc sulfateDB14548
IFN-αSifalimumabDB12773
IFN-λGlucosamineInhibitorDB01296[195]
VIR201DB05110
FontolizumabDB05111
OlsalazineDB01250
Foreskin fibroblastAgonistDB10770
Foreskin keratinocyteAgonistDB10772
EmapalumabNeutralizerDB14724
MBLO3-sulfonylgalactoseDB01818
Methyl alpha-D-mannosideDB01979
4-(hydrogen sulfate)-beta-D-galactopyranoseDB02837
Methyl beta-L-fucopyranosideDB03194
N-acetyl-alpha-neuraminic acidDB03721
Alpha-L-methyl-fucoseDB03879
Alpha-Methyl-N-Acetyl-D-GlucosamineDB04426
TBK1FostamatinibInhibitorDB12010
TGF-β1HyaluronidaseInhibitorDB14740
Foreskin fibroblast (neonatal)AgonistDB10770
Foreskin keratinocyte (neonatal)AgonistDB10772
TerazosinInducerDB01162
TRIM13EtorphineAgonistDB01497
DiprenorphineDB01548
DihydromorphineAgonistDB01565
UBE2ArtenimolLigandDB11638

Table 3.

Molecules targeting the human immune system.

The table represents existing molecules that target the described host factors from the immune system, as identified through a search in the DrugBank Database [103]. The table also displays the specific action of each molecule on the targeted host factor, the corresponding DrugBank ID, and references to studies related to the molecule’s interaction with the Dengue virus or their role during the infection.

These prospects, along with other molecules that engage with distinct host factors within immune pathways, suggest promising avenues for further exploration. Therefore, in-depth studies employing these molecules should be actively pursued.

Advertisement

6. Final remarks

It is well established that the Dengue virus interacts with and modulates a wide range of host factors involved in several metabolic processes. This chapter explores human factors associated with autophagy, apoptosis, lipid metabolism, and the immune system. We have identified 99 host factors that, based on various studies, show promise as potential therapeutic targets for dengue treatment and warrant further exploration within this context (see Figure 1).

Figure 1.

Schematic representation of potential host factors as drug targets for dengue fever treatment. The host factors represented in the scheme are associated with various metabolic processes, including autophagy, apoptosis, lipid metabolism, and the immune system. In gray: Host factors with evidence of influencing DENV infection, with unclear mechanisms of interaction and infection suppression. In green: Host factors for which there is evidence suggesting that their activation suppresses DENV infection. In red: Host factors for which there is evidence suggesting that their inhibition suppresses DENV infection. Above is a brief mention of other processes and pathways containing host factors with potential as drug targets for dengue fever treatment, although these were not addressed in this chapter.

Some of these potential targets are activated or upregulated during the DENV infection process. Blocking or inhibiting these factors would block the virus at different stages, such as entry, genome replication, assembly, or production. Conversely, some other host factors are negatively regulated or inhibited during infection, suggesting that their activation may help suppress the infection (see Figure 1).

Lipid metabolism, key molecules, and pathways, such as de novo fatty acid synthesis, lipid droplet formation, cholesterol biosynthesis, and lipoprotein metabolism, play crucial roles in DENV infection, offering potential therapeutic targets for combating dengue fever. Autophagy and apoptosis also play unique roles in DENV infection. The virus modulates autophagy, enhancing its replication through ATGs and the mTOR signaling pathway modulation. Indeed, this process intersects with apoptosis during DENV infection, where the virus initially induces autophagy but later promotes apoptosis, involving various host factors such as ALX and BCL-2 family proteins in this complex interaction. The immune system serves as an antiviral defense but becomes a target during DENV infection. Receptors like RIG-I, MDA5, and DEAD/H-box RNA helicases, including DDX50 and DDX25, modulate DENV replication, while ubiquitination can have pro-viral and antiviral roles. TRIM proteins are pivotal regulators of PRR signaling pathways, with some, such as TRIM56 and TRIM69, capable of suppressing DENV propagation.

In Figure 1, it is possible to note that—focusing on the autophagy/apoptosis processes but mainly the lipid metabolism—most host factors are positively regulated (red) during dengue and act as pro-viral agents (red) during DENV infection and act as pro-viral agents. This is not a surprise since autophagy and apoptosis share certain host factors, as these processes are interconnected during infection. Moreover, autophagy is intricately linked to lipid metabolism and is modulated during DENV infection, primarily to facilitate ATP production that is essential for virus replication. Hence, a positive regulation of host factors involved in these processes is expected. On the other hand, major host factors associated with the immune system are typically downregulated or inhibited (green) during DENV infection. This aligns with the immune system’s role in viral clearance.

Noteworthy, there are other metabolic pathways such as signal transduction pathways, energy and carbohydrate metabolism, and genetic information processing, where studies have identified host factors as potential therapy targets for dengue treatment. Therefore, it is essential to explore these processes in future studies with the aim of discovering and validating host factors as drug targets for dengue therapy.

Despite the multitude of studies exploring host factors and their essential role in successful DENV infection, there is a lack of research dedicated to developing and screening ligands for these host factors to find drugs for dengue therapy. Given this, the chapter provides a comprehensive list of various ligands for these host factors to support the search for specific ligands in future studies (see Tables 13). Various molecules targeting host factors within lipid metabolism, autophagy, apoptosis, and the immune system have undergone extensive investigation as potential antiviral agents against DENV. These include statins, soraphen-A, hesperetin, minocycline, glycyrrhizic acid, orlistat, bardoxolone, sirolimus, caffeic acid, zinc, interferon lambda (IFN-λ) inhibitors, IFN-λ agonists, TBK1 inhibitors, and TGF-β1 inhibitors, representing potential alternatives for antiviral intervention.

In conclusion, this chapter emphasizes the need for continued research into host factors as drug targets to develop therapeutic strategies to combat DENV infection.

Advertisement

7. Future directions

Efforts directed at identifying potential drug targets, comprehending their interactions, and investigating existing molecules for therapeutic applications can represent a viable approach to discovering effective antiviral strategies against dengue. Some approved drugs are already designed to impact many of the host factors mentioned in this chapter and warrant exploration in studies seeking potential candidates for drug repurposing. Nevertheless, challenges like clinical validation, safety assessments, and optimization of drug dosages must be addressed for successful drug repurposing.

Additionally, this comprehensive chapter underscores the importance of host-virus interactions in DENV infection and emphasizes the potential of host factors as a strategy for dengue fever therapy. Furthermore, exploring existing drugs or compounds that interact with molecules identified as potential targets for dengue therapy can have broader applications in the context of flaviviruses and other arboviruses. For instance, a validated target for treating dengue fever may also prove effective in treating Zika fever. Similarly, a drug repurposing candidate initially intended for dengue fever treatment can also be considered for repurposing in the context of yellow fever therapy. The drug targets and molecules described here would provide ample prospects for further exploration and investigation.

Advertisement

Acknowledgments

The authors thank the René Rachou Institute - Fiocruz Minas, and the Vice Presidency of Research and Biological Collections (VPPCB) of Fiocruz for their support. We also extend our gratitude to Minas Gerais State Agency for Research and Development (FAPEMIG) for funding (PPE-00038-22, BPD-00023-22, and APQ-01688-23).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Goh VSL, Mok C-K, Chu JJH. Antiviral natural products for arbovirus infections. Molecules. 2020;25:2796
  2. 2. Kenney JL, Brault AC. The role of environmental, virological and vector interactions in dictating biological transmission of arthropod-borne viruses by mosquitoes. Advances in Virus Research. 2014;89:39-83
  3. 3. Cheng G, Liu Y, Wang P, et al. Mosquito defense strategies against viral infection. Trends in Parasitology. 2016;32:177-186
  4. 4. Wu P, Yu X, Wang P, et al. Arbovirus lifecycle in mosquito: Acquisition, propagation and transmission. Expert Reviews in Molecular Medicine. 2019;21:e1
  5. 5. PAHO. Guidelines for the Clinical Diagnosis and Treatment of Dengue, Chikungunya, and Zika. Washington, D.C: Pan American Health Organization. Epub ahead of print; 2022. DOI: 10.37774/9789275124871
  6. 6. Adelman ZN, Sanchez-Vargas I, Travanty EA, et al. RNA silencing of dengue virus type 2 replication in transformed C6/36 mosquito cells transcribing an inverted-repeat RNA derived from the virus genome. Journal of Virology. 2002;76:12925-12933
  7. 7. Akhtar IN. Viral genetics and structure. In: Dengue Virus Disease. Cambridge, Massachusetts, EUA: Elsevier. 2019. pp. 85-113
  8. 8. Balakumar M, Vontela HR, Shinde VV, et al. Dengue outbreak and severity prediction: Current methods and the future scope. Virus. 2022;33:125-131
  9. 9. Yang X, Quam MBM, Zhang T, et al. Global burden for dengue and the evolving pattern in the past 30 years. Journal of Travel Medicine. 2021;28. Epub ahead of print 2021:taab146. DOI: 10.1093/jtm/taab146
  10. 10. Bhatt S, Gething PW, Brady OJ, et al. The global distribution and burden of dengue. Nature. 2013;496:504-507
  11. 11. Salles TS, da Encarnação S-GT, de Alvarenga ESL, et al. History, epidemiology and diagnostics of dengue in the American and Brazilian contexts: A review. Parasites & Vectors. 2018;11:264
  12. 12. WHO. Dengue Guidelines for Diagnosis, Treatment, Prevention and Control. Geneva, Switzerland: World Health Organization; 2009; Available from: https://www.ncbi.nlm.nih.gov/pubmed/23762963
  13. 13. Tayal A, Kabra SK, Lodha R. Management of dengue: An updated review. Indian Journal of Pediatrics. 2023;90:168-177
  14. 14. Deen J, von Seidlein L. Paracetamol for dengue fever: No benefit and potential harm? The Lancet Global Health. 2019;7:e552-e553
  15. 15. Niranjan R, Sumitha MK, Sankari T, et al. Nonstructural protein-1 (NS1) of dengue virus type-2 differentially stimulate expressions of matrix metalloproteinases in monocytes: Protective effect of paracetamol. International Immunopharmacology. 2019;73:270-279
  16. 16. Swamy AM, Mahesh PY, Tumkur SR. Liver function in dengue and it’s correlation with disease severity: An retrospective cross-sectional observational study in a tertiary care center in coastal India. Pan African Medical Journal. 2021;40:261
  17. 17. Fink J, Gu F, Ling L, et al. Host gene expression profiling of dengue virus infection in cell lines and patients. PLoS Neglected Tropical Diseases. 2007;1:e86
  18. 18. Herrero LJ, Zakhary A, Gahan ME, et al. Dengue virus therapeutic intervention strategies based on viral, vector and host factors involved in disease pathogenesis. Pharmacology & Therapeutics. 2013;137:266-282
  19. 19. Chen J, Luo X, Qiu H, et al. Drug discovery and drug marketing with the critical roles of modern administration. American Journal of Translational Research. 2018;10:4302-4312
  20. 20. Amemiya T, Gromiha MM, Horimoto K, et al. Drug repositioning for dengue haemorrhagic fever by integrating multiple omics analyses. Scientific Reports. 2019;9:523
  21. 21. Obi J, Gutiérrez-Barbosa H, Chua J, et al. Current trends and limitations in dengue antiviral research. Tropical Medicine and Infectious Disease. 2021;6:180
  22. 22. Kwon Y-J, Heo J, Wong HEE, et al. Kinome siRNA screen identifies novel cell-type specific dengue host target genes. Antiviral Research. 2014;110:20-30
  23. 23. Nanaware N, Banerjee A, Mullick Bagchi S, et al. Dengue virus infection: A tale of viral exploitations and host responses. Viruses. 2021;13:1967
  24. 24. Urcuqui-Inchima S, Patiño C, Torres S, et al. Recent developments in understanding dengue virus replication. In: Advances in Virus Research. United States: Elsevier; 2010. pp. 1-39
  25. 25. Clyde K, Kyle JL, Harris E. Recent advances in deciphering viral and host determinants of dengue virus replication and pathogenesis. Journal of Virology. 2006;80:11418-11431
  26. 26. Cruz-Oliveira C, Freire JM, Conceição TM, et al. Receptors and routes of dengue virus entry into the host cells. FEMS Microbiology Reviews. 2015;39:155-170
  27. 27. Krishnan MN, Sukumaran B, Pal U, et al. Rab 5 is required for the cellular entry of dengue and West Nile viruses. Journal of Virology. 2007;81:4881-4885
  28. 28. Lescar J, Soh S, Lee LT, et al. The dengue virus replication complex: From RNA replication to protein-protein interactions to evasion of innate immunity. Advances in Experimental Medicine and Biology. 2018;1062:115-129
  29. 29. Vaughan G, Olivera H, Santos-Argumedo L, et al. Dengue virus replicative intermediate RNA detection by reverse transcription-PCR. Clinical and Vaccine Immunology. 2002;9:198-200
  30. 30. van den Elsen K, Quek JP, Luo D. Molecular insights into the flavivirus replication complex. Viruses. 2021;13:956
  31. 31. Neufeldt CJ, Cortese M, Acosta EG, et al. Rewiring cellular networks by members of the flaviviridae family. Nature Reviews. Microbiology. 2018;16:125-142
  32. 32. Iglesias NG, Gamarnik AV. Dynamic RNA structures in the dengue virus genome. RNA Biology. 2011;8:249-257
  33. 33. Gebhard LG, Filomatori CV, Gamarnik AV. Functional RNA elements in the dengue virus genome. Viruses. 2011;3:1739-1756
  34. 34. Yu I-M, Zhang W, Holdaway HA, et al. Structure of the immature dengue virus at low pH primes proteolytic maturation. Science. 2008;319:1834-1837
  35. 35. Zitzmann C, Schmid B, Ruggieri A, et al. A coupled mathematical model of the intracellular replication of dengue virus and the host cell immune response to infection. Frontiers in Microbiology. 2020;11:725
  36. 36. Zhang J, Lan Y, Sanyal S. Modulation of lipid droplet metabolism—A potential target for therapeutic intervention in flaviviridae infections. Frontiers in Microbiology. 2017;8. Epub ahead of print 28 November:2286. DOI: 10.3389/fmicb.2017.02286
  37. 37. Martín-Acebes MA, Vázquez-Calvo Á, Saiz J-C. Lipids and flaviviruses, present and future perspectives for the control of dengue, Zika, and West Nile viruses. Progress in Lipid Research. 2016;64:123-137
  38. 38. Wu W, Chen R, Wan Y, et al. Acetyl-CoA carboxylase (ACC) inhibitor, CP640186, effectively inhibited dengue virus (DENV) infection via regulating ACC phosphorylation. Molecules. Epub ahead of print 2022. 2022;27(23):8583. DOI: 10.3390/molecules27238583
  39. 39. Jiménez de Oya N, Esler WP, Huard K, et al. Targeting host metabolism by inhibition of acetyl-coenzyme a carboxylase reduces flavivirus infection in mouse models. Emerging Microbes and Infection. 2019;8:624-636
  40. 40. Heaton NS, Perera R, Berger KL, et al. Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis. Proceedings of the National Academy of Sciences. 2010;107:17345-17350
  41. 41. Tongluan N, Ramphan S, Wintachai P, et al. Involvement of fatty acid synthase in dengue virus infection. Virology Journal. 2017;14:28
  42. 42. Olzmann JA, Carvalho P. Dynamics and functions of lipid droplets. Nature Reviews. Molecular Cell Biology. 2019;20:137-155
  43. 43. Gaspar ML, Hofbauer HF, Kohlwein SD, et al. Coordination of storage lipid synthesis and membrane biogenesis. Journal of Biological Chemistry. 2011;286:1696-1708
  44. 44. Heaton NS, Randall G. Dengue virus-induced autophagy regulates lipid metabolism. Cell Host & Microbe. 2010;8:422-432
  45. 45. Zhang J, Lan Y, Li MY, et al. Flaviviruses exploit the lipid droplet protein AUP1 to trigger lipophagy and drive virus production. Cell Host & Microbe. 2018;23:819-831.e5
  46. 46. Ferreira JGG, Gava SG, Oliveira ES, et al. Gene expression signatures in AML-12 hepatocyte cells upon dengue virus infection and acetaminophen treatment. Viruses. 2020;12:1284
  47. 47. Jackson CL. Lipid droplet biogenesis. Current Opinion in Cell Biology. 2019;59:88-96
  48. 48. Carvalho FA, Carneiro FA, Martins IC, et al. Dengue virus capsid protein binding to hepatic lipid droplets (LD) is potassium ion dependent and is mediated by LD surface proteins. Journal of Virology. 2012;86:2096-2108
  49. 49. Moretti F, Bergman P, Dodgson S, et al. TMEM 41B is a novel regulator of autophagy and lipid mobilization. EMBO Reports. 2018;19. Epub ahead of print 20 September:e45889. DOI: 10.15252/embr.201845889
  50. 50. Yousefi M, Lee WS, Yan B, et al. TMEM41B and VMP1 modulate cellular lipid and energy metabolism for facilitating dengue virus infection. PLoS Pathogens. 2022;18:1-26
  51. 51. Hyrina A, Meng F, McArthur SJ, et al. Human subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P) regulates cytoplasmic lipid droplet abundance: A potential target for indirect-acting anti-dengue virus agents. PLoS One. 2017;12:1-22
  52. 52. Samsa MM, Mondotte JA, Iglesias NG, et al. Dengue virus capsid protein usurps lipid droplets for viral particle formation. PLoS Pathogens. 2009;5:e1000632
  53. 53. Marceau CD, Puschnik AS, Majzoub K, et al. Genetic dissection of flaviviridae host factors through genome-scale CRISPR screens. Nature. 2016;535:159-163
  54. 54. Martins IC, Gomes-Neto F, Faustino AF, et al. The disordered N-terminal region of dengue virus capsid protein contains a lipid-droplet-binding motif. Biochemical Journal. 2012;444:405-415
  55. 55. Oliveira ES, Colombarolli SG, Nascimento CS, et al. Increased levels of Txa₂ induced by dengue virus infection in IgM positive individuals is related to the mild symptoms of dengue. Viruses. 2018;10. Epub ahead of print 2018:104. DOI: 10.3390/v10030104
  56. 56. Nes WD. Biosynthesis of cholesterol and other sterols. Chemical Reviews. 2011;111:6423-6451
  57. 57. Cerqueira NMFSA, Oliveira EF, Gesto DS, et al. Cholesterol biosynthesis: A mechanistic overview. Biochemistry. 2016;55:5483-5506
  58. 58. Soto-Acosta R, Mosso C, Cervantes-Salazar M, et al. The increase in cholesterol levels at early stages after dengue virus infection correlates with an augment in LDL particle uptake and HMG-CoA reductase activity. Virology. 2013;442:132-147
  59. 59. Osuna-Ramos JF, Reyes-Ruiz JM, Bautista-Carbajal P, et al. Ezetimibe inhibits dengue virus infection in Huh-7 cells by blocking the cholesterol transporter Niemann–pick C1-like 1 receptor. Antiviral Research. 2018;160:151-164
  60. 60. Biswas HH, Gordon A, Nuñez A, et al. Lower low-density lipoprotein cholesterol levels are associated with severe dengue outcome. PLoS Neglected Tropical Diseases. 2015;9:e0003904
  61. 61. Poh MK, Shui G, Xie X, et al. U18666A, an intra-cellular cholesterol transport inhibitor, inhibits dengue virus entry and replication. Antiviral Research. 2012;93:191-198
  62. 62. Sreelatha L, Malakar S, Songprakhon P, et al. Serine protease inhibitor AEBSF reduces dengue virus infection via decreased cholesterol synthesis. Virus Research. 2019;271:197672
  63. 63. Rothwell C, LeBreton A, Young Ng C, et al. Cholesterol biosynthesis modulation regulates dengue viral replication. Virology. 2009;389:8-19
  64. 64. Mohamed B, Mazeaud C, Baril M, et al. Very-long-chain fatty acid metabolic capacity of 17-beta-hydroxysteroid dehydrogenase type 12 (HSD17B12) promotes replication of hepatitis C virus and related flaviviruses. Scientific Reports. 2020;10:1-15
  65. 65. Noels H, Lehrke M, Vanholder R, et al. Lipoproteins and fatty acids in chronic kidney disease: Molecular and metabolic alterations. Nature Reviews. Nephrology. 2021;17:528-542
  66. 66. Feingold KR. Lipid and lipoprotein metabolism. Endocrinology and Metabolism Clinics of North America. 2022;51:437-458
  67. 67. Feingold KR, Grunfeld C. Lipids: A key player in the battle between the host and microorganisms. Journal of Lipid Research. 2012;53:2487-2489
  68. 68. Asztalos BF, Niisuke K, Horvath KV. High-density lipoprotein: Our elusive friend. Current Opinion in Lipidology. 2019;30:314-319
  69. 69. Kostner KM, Kostner GM. Lipoprotein (a): A historical appraisal. Journal of Lipid Research. 2017;58:1-14
  70. 70. Julve J, Martín-Campos JM, Escolà-Gil JC, et al. Chylomicrons: Advances in biology, pathology, laboratory testing, and therapeutics. Clinica Chimica Acta. 2016;455:134-148
  71. 71. Tiwari S, Siddiqi SA. Intracellular trafficking and secretion of VLDL. Arteriosclerosis, Thrombosis, and Vascular Biology. 2012;32:1079-1086
  72. 72. El-Bacha T, Struchiner CJ, Cordeiro MT, et al. 1 H nuclear magnetic resonance metabolomics of plasma unveils liver dysfunction in dengue patients. Journal of Virology. 2016;90:7429-7443
  73. 73. Durán A, Carrero R, Parra B, et al. Association of lipid profile alterations with severe forms of dengue in humans. Archives of Virology. 2015;160:1687-1692
  74. 74. Faustino AF, Carvalho FA, Martins IC, et al. Dengue virus capsid protein interacts specifically with very low-density lipoproteins. Nanomedicine. 2014;10:247-255
  75. 75. Benfrid S, Park K, Dellarole M, et al. Dengue virus NS1 protein conveys pro‐inflammatory signals by docking onto high‐density lipoproteins. EMBO Reports. 2022;23:e53600. Epub ahead of print 5 July 2022. DOI: 10.15252/embr.202153600
  76. 76. Guevara J, Romo J, McWhorter T, et al. Analogs of LDL receptor ligand motifs in dengue envelope and capsid proteins as potential codes for cell entry. Journal of Viruses. 2015;2015:1-15
  77. 77. Zhou L, Li C, Gao L, et al. High-density lipoprotein synthesis and metabolism (review). Molecular Medicine Reports. 2015;12:4015-4021
  78. 78. Mendez AJ, Lin G, Wade DP, et al. Membrane lipid domains distinct from cholesterol/sphingomyelin-rich rafts are involved in the ABCA1-mediated lipid secretory pathway. Journal of Biological Chemistry. 2001;276:3158-3166
  79. 79. Khadka S, Vangeloff AD, Zhang C, et al. A physical interaction network of dengue virus and human proteins. Molecular & Cellular Proteomics. 2011;10:M111.012187
  80. 80. Coelho DR, Carneiro PH, Mendes-Monteiro L, et al. ApoA1 neutralizes proinflammatory effects of dengue virus NS1 protein and modulates viral immune evasion. Journal of Virology. 2021;95:1-12
  81. 81. Manchala NR, Dungdung R, Pilankatta R. Proteomic analysis reveals the enhancement of human serum apolipoprotein A-1(APO A-1) in individuals infected with multiple dengue virus serotypes. Tropical Medicine and International Health. 2017;22:1334-1342
  82. 82. Sposito AC, de Lima-Junior JC, Moura FA, et al. Reciprocal multifaceted interaction between HDL (high-density lipoprotein) and myocardial infarction. Arteriosclerosis, Thrombosis, and Vascular Biology. 2019;39:1550-1564
  83. 83. Fitzgerald ML, Mujawar Z, Tamehiro N. ABC transporters, atherosclerosis and inflammation. Atherosclerosis. 2010;211:361-370
  84. 84. Zahid MN, Turek M, Xiao F, et al. The postbinding activity of scavenger receptor class B type I mediates initiation of hepatitis C virus infection and viral dissemination. Hepatology. 2013;57:492-504
  85. 85. Mujawar Z, Tamehiro N, Grant A, et al. Mutation of the ATP cassette binding transporter A1 (ABCA1) C-terminus disrupts HIV-1 Nef binding but does not block the Nef enhancement of ABCA1 protein degradation. Biochemistry. 2010;49:8338-8349
  86. 86. Syder AJ, Lee H, Zeisel MB, et al. Small molecule scavenger receptor BI antagonists are potent HCV entry inhibitors. Journal of Hepatology. 2011;54:48-55
  87. 87. Li Y, Kakinami C, Li Q , et al. Human apolipoprotein A-I is associated with dengue virus and enhances virus infection through SR-BI. PLoS One. 2013;8. Epub ahead of print 2013:e70390. DOI: 10.1371/journal.pone.0070390
  88. 88. Alcalá AC, Maravillas JL, Meza D, et al. Dengue virus NS1 uses scavenger receptor B1 as a cell receptor in cultured cells. Journal of Virology. 2022. . Epub ahead of print 9 March 2022;96:e0166421. DOI: 10.1128/jvi.01664-21
  89. 89. Folly BB, Weffort-Santos AM, Fathman C, et al. Dengue-2 structural proteins associate with human proteins to produce a coagulation and innate immune response biased interactome. BMC Infectious Diseases. 2011;11:34
  90. 90. Faustino AF, Martins IC, Carvalho FA, et al. Understanding dengue virus capsid protein interaction with key biological targets. Scientific Reports. 2015;5:10592
  91. 91. Lee J-Y, Acosta EG, Stoeck IK, et al. Apolipoprotein E likely contributes to a maturation step of infectious hepatitis C virus particles and interacts with viral envelope glycoproteins. Journal of Virology. 2014;88:12422-12437
  92. 92. Lingwood D, Simons K. Lipid rafts as a membrane-organizing principle. Science. 1979;2010(327):46-50
  93. 93. Sezgin E, Levental I, Mayor S, et al. The mystery of membrane organization: Composition, regulation and roles of lipid rafts. Nature Reviews. Molecular Cell Biology. 2017;18:361-374
  94. 94. García Cordero J, León Juárez M, González-Y-Merchand JA, et al. Caveolin-1 in lipid rafts interacts with dengue virus NS3 during polyprotein processing and replication in HMEC-1 cells. PLoS One. 2014;9:e90704
  95. 95. Diwaker D, Mishra KP, Ganju L, et al. Protein disulfide isomerase mediates dengue virus entry in association with lipid rafts. Viral Immunology. 2015;28:153-160
  96. 96. Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, et al. DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target. PLoS Pathogens. 2017;13. Epub ahead of print 2017:e1006257. DOI: 10.1371/journal.ppat.1006257
  97. 97. Tillander V, Alexson SEH, Cohen DE. Deactivating fatty acids: Acyl-CoA thioesterase-mediated control of lipid metabolism. Trends in Endocrinology & Metabolism. 2017;28:473-484
  98. 98. Hunt MC, Alexson SEH. The role acyl-CoA thioesterases play in mediating intracellular lipid metabolism. Progress in Lipid Research. 2002;41:99-130
  99. 99. St Clair LA, Mills SA, Lian E, et al. Acyl-CoA thioesterases: A rheostat that controls activated fatty acids modulates dengue virus serotype 2 replication. Viruses. 2022;14. Epub ahead of print 2022:240. DOI: 10.3390/v14020240
  100. 100. Gullberg RC, Steel JJ, Pujari V, et al. Stearoly-CoA desaturase 1 differentiates early and advanced dengue virus infections and determines virus particle infectivity. PLoS Pathogens. 2018;14:1-28
  101. 101. Kikuchi K, Tsukamoto H. Stearoyl-CoA desaturase and tumorigenesis. Chemico-Biological Interactions. 2020;316:108917
  102. 102. Álvarez-Fernández H, Mingo-Casas P, Blázquez A-B, et al. Allosteric inhibition of neutral sphingomyelinase 2 (nSMase2) by DPTIP: From antiflaviviral activity to deciphering its binding site through in silico studies and experimental validation. International Journal of Molecular Sciences. 2022;23:13935
  103. 103. Wishart DS, Feunang YD, Guo AC, et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Research. 2018;46:D1074-D1082
  104. 104. Eberle RJ, Olivier DS, Amaral MS, et al. Promising natural compounds against flavivirus proteases: Citrus flavonoids hesperetin and hesperidin. Plants. 2021;10:2183
  105. 105. Mao J, Lin E, He L, et al. Autophagy and viral infection. Advances in Experimental Medicine and Biology. 2019;1209:55-78
  106. 106. Parzych KR, Klionsky DJ. An overview of autophagy: Morphology, mechanism, and regulation. Antioxidants & Redox Signaling. 2014;20:460-473
  107. 107. Hitakarun A, Khongwichit S, Wikan N, et al. Evaluation of the antiviral activity of orlistat (tetrahydrolipstatin) against dengue virus, Japanese encephalitis virus, Zika virus and chikungunya virus. Scientific Reports. 2020;10:1499
  108. 108. Yu L, Chen Y, Tooze SA. Autophagy pathway: Cellular and molecular mechanisms. Autophagy. 2018;14:207-215
  109. 109. Mizushima N. A brief history of autophagy from cell biology to physiology and disease. Nature Cell Biology. 2018;20:521-527
  110. 110. Lai Y-C, Chuang Y-C, Chang C-P, et al. Minocycline suppresses dengue virus replication by down-regulation of macrophage migration inhibitory factor-induced autophagy. Antiviral Research. 2018;155:28-38
  111. 111. Wu YW, Mettling C, Wu SR, et al. Autophagy-associated dengue vesicles promote viral transmission avoiding antibody neutralization. Scientific Reports. 2016;6:1-10
  112. 112. Manchala NR, Dungdung R, Trivedi P, et al. Mycophenolic acid (MPA) modulates host cellular autophagy progression in sub genomic dengue virus-2 replicon cells. Microbial Pathogenesis. 2019;137:103762
  113. 113. Komatsu M, Waguri S, Koike M, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell. 2007;131:1149-1163
  114. 114. Deretic V. Autophagy as an innate immunity paradigm: Expanding the scope and repertoire of pattern recognition receptors. Current Opinion in Immunology. 2012;24:21-31
  115. 115. Wu Y, Zhou T, Hu J, et al. Autophagy activation induces p62-dependent autophagic degradation of dengue virus capsid protein during infection. Frontiers in Microbiology. 2022;13. Epub ahead of print 5 July 2022:889693. DOI: 10.3389/fmicb.2022.889693
  116. 116. Lampada A, O’Prey J, Szabadkai G, et al. mTORC1-independent autophagy regulates receptor tyrosine kinase phosphorylation in colorectal cancer cells via an mTORC2-mediated mechanism. Cell Death and Differentiation. 2017;24:1045-1062
  117. 117. Kong W, Mao J, Yang Y, et al. Mechanisms of mTOR and autophagy in human endothelial cell infected with dengue Virus-2. Viral Immunology. 2020;33:61-70
  118. 118. Lahon A, Arya RP, Banerjea AC. Dengue virus dysregulates master transcription factors and PI3K/AKT/mTOR signaling pathway in megakaryocytes. Frontiers in Cellular and Infection Microbiology. 2021;11. Epub ahead of print 26 August:715208. DOI: 10.3389/fcimb.2021.715208
  119. 119. Rox K, Heyner M, Krull J, et al. Physiologically based pharmacokinetic/pharmacodynamic model for the treatment of dengue infections applied to the broad spectrum antiviral soraphen A. ACS Pharmacology & Translational Science. 2021;4:1499-1513
  120. 120. Jordan TX, Randall G. Dengue virus activates the AMP kinase-mTOR axis to stimulate a proviral lipophagy. Journal of Virology. 2017;91. Epub ahead of print June 2017:e02020-e02016. DOI: 10.1128/JVI.02020-16
  121. 121. Carter CC, Mast FD, Olivier JP, et al. Dengue activates mTORC2 signaling to counteract apoptosis and maximize viral replication. Frontiers in Cellular and Infection Microbiology. 2022;12. Epub ahead of print 12 September 2022:979996. DOI: 10.3389/fcimb.2022.979996
  122. 122. Zhu X, Messer JS, Wang Y, et al. Cytosolic HMGB1 controls the cellular autophagy/apoptosis checkpoint during inflammation. Journal of Clinical Investigation. 2015;125:1098-1110
  123. 123. Chaudhary N, Srivastava S, Gupta S, et al. Dengue virus induced autophagy is mediated by HMGB1 and promotes viral propagation. International Journal of Biological Macromolecules. 2023;229:624-635
  124. 124. Ghavami S, Hashemi M, Ande SR, et al. Apoptosis and cancer: Mutations within caspase genes. Journal of Medical Genetics. 2009;46:497-510
  125. 125. Wilson NS, Dixit V, Ashkenazi A. Death receptor signal transducers: Nodes of coordination in immune signaling networks. Nature Immunology. 2009;10:348-355
  126. 126. Pobezinskaya YL, Liu Z. The role of TRADD in death receptor signaling. Cell Cycle. 2012;11:871-876
  127. 127. Strasser A, Vaux DL. Viewing BCL2 and cell death control from an evolutionary perspective. Cell Death and Differentiation. 2018;25:13-20
  128. 128. Ashkenazi A, Salvesen G. Regulated cell death: Signaling and mechanisms. Annual Review of Cell and Developmental Biology. 2014;30:337-356
  129. 129. Okamoto T, Suzuki T, Kusakabe S, et al. Regulation of apoptosis during flavivirus infection. Viruses. 2017;9:243
  130. 130. Pan Y, Cheng A, Wang M, et al. The dual regulation of apoptosis by flavivirus. Frontiers in Microbiology. 2021;12. Epub ahead of print 24 March 2021:654494. DOI: 10.3389/fmicb.2021.654494
  131. 131. Strappazzon F, Torch S, Chatellard-Causse C, et al. Alix is involved in caspase 9 activation during calcium-induced apoptosis. Biochemical and Biophysical Research Communications. 2010;397:64-69
  132. 132. Thepparit C, Khongwichit S, Ketsuwan K, et al. Dengue virus requires apoptosis linked gene-2-interacting protein X (ALIX) for viral propagation. Virus Research. 2019;261:65-71
  133. 133. Czabotar PE, Lessene G, Strasser A, et al. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nature Reviews. Molecular Cell Biology. 2014;15:49-63
  134. 134. Suzuki T, Okamoto T, Katoh H, et al. Infection with flaviviruses requires BCLXL for cell survival. PLoS Pathogens. 2018;14:e1007299
  135. 135. Huang J, Li Y, Qi Y, et al. Coordinated regulation of autophagy and apoptosis determines endothelial cell fate during dengue virus type 2 infection. Molecular and Cellular Biochemistry. 2014;397:157-165
  136. 136. Lu ZY, Cheng MH, Yu CY, et al. Dengue nonstructural protein 1 maintains autophagy through retarding caspase-mediated cleavage of beclin-1. International Journal of Molecular Sciences. 2020;21:1-19
  137. 137. Long X, Li Y, Qi Y, et al. XAF1 contributes to dengue virus‐induced apoptosis in vascular endothelial cells. The FASEB Journal. 2013;27:1062-1073
  138. 138. Mura M, Hopkins TG, Michael T, et al. LARP1 post-transcriptionally regulates mTOR and contributes to cancer progression. Oncogene. 2015;34:5025-5036
  139. 139. Mehto S, Kundu S, Chauhan S, et al. RIPosomes are targets of IRGM-SQSTM1-dependent autophagy. Autophagy. 2023;19:1045-1047
  140. 140. Takahashi Y, Karbowski M, Yamaguchi H, et al. Loss of Bif-1 suppresses Bax/Bak conformational change and mitochondrial apoptosis. Molecular and Cellular Biology. 2005;25:9369-9382
  141. 141. Dalrymple NA, Cimica V, Mackow ER. Dengue virus NS proteins inhibit RIG-I/MAVS signaling by blocking TBK1/IRF3 phosphorylation: Dengue virus serotype 1 NS4A is a unique interferon-regulating virulence determinant. MBio. 2015;6. Epub ahead of print July 2015:e00553-e00515. DOI: 10.1128/mBio.00553-15
  142. 142. Rasmussen NL, Zhou J, Olsvik H, et al. The inflammation repressor TNIP1/ABIN-1 is degraded by autophagy following TBK1 phosphorylation of its LIR. Autophagy. 2023;19(10):1-3
  143. 143. Hubert V, Peschel A, Langer B, et al. LAMP-2 is required for incorporating syntaxin-17 into autophagosomes and for their fusion with lysosomes. Biology Open. 2016;5:1516-1529
  144. 144. Ullah R, Li J, Fang P, et al. DEAD/H-box helicases: Anti-viral and pro-viral roles during infections. Virus Research. 2022;309:198658
  145. 145. Samir P, Kanneganti T-D. DEAD/H-box helicases in immunity, inflammation, cell differentiation, and cell death and disease. Cell. 2022;11:1608
  146. 146. Sprokholt JK, Kaptein TM, van Hamme JL, et al. RIG-I-like receptor activation by dengue virus drives follicular T helper cell formation and antibody production. PLoS Pathogens. 2017;13:1-19
  147. 147. Sprokholt JK, Kaptein TM, van Hamme JL, et al. RIG-I–like receptor triggering by dengue virus drives dendritic cell immune activation and TH1 differentiation. The Journal of Immunology. 2017;198:4764-4771
  148. 148. Olagnier D, Scholte FEM, Chiang C, et al. Inhibition of dengue and chikungunya virus infections by RIG-I-mediated type I interferon-independent stimulation of the innate antiviral response. Journal of Virology. 2014;88:4180-4194
  149. 149. Ho V, Yong HY, Chevrier M, et al. RIG-I activation by a designer short RNA ligand protects human immune cells against dengue virus infection without causing cytotoxicity. Journal of Virology. 2019;93:1-18
  150. 150. Dalrymple NA, Cimica V, Mackow ER. Dengue virus NS proteins inhibit RIG-I/MAVS signaling by blocking TBK1/IRF3 phosphorylation: Dengue virus serotype 1 NS4A is a unique interferon-regulating virulence determinant. MBio. 2015;6:1-12
  151. 151. Varga N, Sutkeviciute I, Ribeiro-Viana R, et al. A multivalent inhibitor of the DC-SIGN dependent uptake of HIV-1 and dengue virus. Biomaterials. 2014;35:4175-4184
  152. 152. Chen J-M, Fan Y-C, Lin J-W, et al. Bovine lactoferrin inhibits dengue virus infectivity by interacting with heparan sulfate, low-density lipoprotein receptor, and DC-SIGN. International Journal of Molecular Sciences. 2017;18:1957
  153. 153. van Gent M, Sparrer KMJ, Gack MU. TRIM proteins and their roles in antiviral host defenses. Annual Review of Virology. 2018;5:385-405
  154. 154. Liu B, Li NL, Wang J, et al. Overlapping and distinct molecular determinants dictating the antiviral activities of TRIM56 against flaviviruses and coronavirus. Journal of Virology. 2014;88:13821-13835
  155. 155. Nie Y, Deng D, Mou L, et al. Dengue virus 2 NS2B targets MAVS and IKKε to evade the antiviral innate immune response. Journal of Microbiology and Biotechnology. 2023;33:600-606
  156. 156. Sui L, Zhao Y, Wang W, et al. Flavivirus prM interacts with MDA5 and MAVS to inhibit RLR antiviral signaling. Cell & Bioscience. 2023;13:9
  157. 157. Liu P, Ridilla M, Patel P, et al. Beyond attachment: Roles of DC-SIGN in dengue virus infection. Traffic. 2017;18:218-231
  158. 158. Leela SL, Srisawat C, Sreekanth GP, et al. Drug repurposing of minocycline against dengue virus infection. Biochemical and Biophysical Research Communications. 2016;478:410-416
  159. 159. Hour M-J, Chen Y, Lin C-S, et al. Glycyrrhizic acid derivatives bearing amino acid residues in the carbohydrate part as dengue virus E protein inhibitors: Synthesis and antiviral activity. International Journal of Molecular Sciences. 2022;23:10309
  160. 160. Baltina LA, Tasi Y-T, Huang S-H, et al. Glycyrrhizic acid derivatives as dengue virus inhibitors. Bioorganic & Medicinal Chemistry Letters. 2019;29:126645
  161. 161. Ong SP, Lee LM, Leong YFI, et al. Dengue virus infection mediates HMGB1 release from monocytes involving PCAF acetylase complex and induces vascular leakage in endothelial cells. PLoS One. 2012;7:e41932
  162. 162. Chaudhary N, Srivastava S, Dave U, et al. High-mobility group box 1 protein promotes dengue virus replication by interacting with untranslated regions of viral genome. Virus Research. 2022;309:198668
  163. 163. Martinez F, Ghietto LM, Lingua G, et al. New insights into the antiviral activity of nordihydroguaiaretic acid: Inhibition of dengue virus serotype 1 replication. Phytomedicine. 2022;106:154424
  164. 164. Sinha M, Chakraborty U, Kool A, et al. In-vitro antiviral action of Eupatorium perfoliatum against dengue virus infection: Modulation of mTOR signaling and autophagy. Journal of Ethnopharmacology. 2022;282:114627
  165. 165. King CA, Wegman AD, Endy TP. Mobilization and activation of the innate immune response to dengue virus. Frontiers in Cellular and Infection Microbiology. 2020;10. Epub ahead of print 3 November 2020:574417. DOI: 10.3389/fcimb.2020.574417
  166. 166. Kao Y-T, Lai MMC, Yu C-Y. How dengue virus circumvents innate immunity. Frontiers in Immunology. 2018;9:2860. Epub ahead of print 4 December 2018. DOI: 10.3389/fimmu.2018.02860
  167. 167. Lee MF, Voon GZ, Lim HX, et al. Innate and adaptive immune evasion by dengue virus. Frontiers in Cellular and Infection Microbiology. 2022;12. Epub ahead of print 16 September 2022:1004608. DOI: 10.3389/fcimb.2022.1004608
  168. 168. Ashley C, Abendroth A, McSharry B, et al. Interferon-independent upregulation of interferon-stimulated genes during human cytomegalovirus infection is dependent on IRF3 expression. Viruses. 2019;11:246
  169. 169. Fernandes-Santos C, Azeredo EL de. Innate immune response to dengue virus: Toll-like receptors and antiviral response. Viruses 2022; 14: 992.
  170. 170. Yoneyama M, Onomoto K, Jogi M, et al. Viral RNA detection by RIG-I-like receptors. Current Opinion in Immunology. 2015;32:48-53
  171. 171. Wang K, Zou C, Wang X, et al. Interferon-stimulated TRIM69 interrupts dengue virus replication by ubiquitinating viral nonstructural protein 3. PLoS Pathogens. 2018;14:1-24
  172. 172. Bagga T, Tulsian NK, Mok YK, et al. Mapping of molecular interactions between human E3 ligase TRIM69 and dengue virus NS3 protease using hydrogen–deuterium exchange mass spectrometry. Cellular and Molecular Life Sciences. 2022;79:233
  173. 173. Manokaran G, Finol E, Wang C, et al. Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness. Science. 1979;2015(350):217-221
  174. 174. Sanchez JG, Sparrer KMJ, Chiang C, et al. TRIM25 binds RNA to modulate cellular anti-viral defense. Journal of Molecular Biology. 2018;430:5280-5293
  175. 175. Li G, Feng T, Pan W, et al. DEAD-box RNA helicase DDX3X inhibits DENV replication via regulating type one interferon pathway. Biochemical and Biophysical Research Communications. 2015;456:327-332
  176. 176. Feng T, Sun T, Li G, et al. DEAD-box helicase DDX25 is a negative regulator of type I interferon pathway and facilitates RNA virus infection. Frontiers in Cellular and Infection Microbiology. 2017;7:1-11
  177. 177. Feng T, Deng L, Lu X, et al. Ubiquitin-conjugating enzyme UBE2J1 negatively modulates interferon pathway and promotes RNA virus infection. Virology Journal. 2018;15:1-9
  178. 178. Murphy Schafer AR, Smith JL, Pryke KM, et al. The E3 ubiquitin ligase SIAH1 targets MyD88 for proteasomal degradation during dengue virus infection. Frontiers in Microbiology. 2020;11:1-13
  179. 179. Saikh KU, Morazzani EM, Piper AE, et al. A small molecule inhibitor of MyD88 exhibits broad spectrum antiviral activity by up regulation of type I interferon. Antiviral Research. 2020;181:104854
  180. 180. Carlin AF, Plummer EM, Vizcarra EA, et al. An IRF-3-, IRF-5-, and IRF-7-independent pathway of dengue viral resistance utilizes IRF-1 to stimulate type I and II interferon responses. Cell Reports. 2017;21:1600-1612
  181. 181. Richardson RB, Ohlson MB, Eitson JL, et al. A CRISPR screen identifies IFI6 as an ER-resident interferon effector that blocks flavivirus replication. Nature Microbiology. 2018;3:1214-1223
  182. 182. Palma-Ocampo HK, Flores-Alonso JC, Vallejo-Ruiz V, et al. Interferon lambda inhibits dengue virus replication in epithelial cells. Virology Journal. 2015;12:1-14
  183. 183. Yu JS, Tseng CK, Lin CK, et al. Celastrol inhibits dengue virus replication via up-regulating type I interferon and downstream interferon-stimulated responses. Antiviral Research. 2017;137:49-57
  184. 184. Mellors J, Tipton T, Longet S, et al. Viral evasion of the complement system and its importance for vaccines and therapeutics. Frontiers in Immunology. 2020;11. Epub ahead of print 9 July 2020:1450. DOI: 10.3389/fimmu.2020.01450
  185. 185. Garred P, Tenner AJ, Mollnes TE. Therapeutic targeting of the complement system: From rare diseases to pandemics. Pharmacological Reviews. 2021;73:792-827
  186. 186. Marinho CF, Azeredo EL, Torrentes-Carvalho A, et al. Down-regulation of complement receptors on the surface of host monocyte even as in vitro complement pathway blocking interferes in dengue infection. PLoS One. 2014;9. Epub ahead of print 2014:e102014. DOI: 10.1371/journal.pone.0102014
  187. 187. Cabezas S, Bracho G, Aloia AL, et al. Dengue virus induces increased activity of the complement alternative pathway in infected cells. Journal of Virology. 2018;92:1-20
  188. 188. Douradinha B, McBurney SP, Soares de Melo KM, et al. C1q binding to dengue virus decreases levels of infection and inflammatory molecules transcription in THP-1 cells. Virus Research. 2014;179:231-234
  189. 189. Silva EM, Conde JN, Allonso D, et al. Mapping the interactions of dengue virus NS1 protein with human liver proteins using a yeast two-hybrid system: Identification of C1q as an interacting partner. PLoS One. 2013;8:e57514
  190. 190. Thiemmeca S, Tamdet C, Punyadee N, et al. Secreted NS1 protects dengue virus from mannose-binding lectin–mediated neutralization. The Journal of Immunology. 2016;197:4053-4065
  191. 191. Teixeira GS, Andrade AA, Torres LR, et al. Suppression of TGF-β/Smad2 signaling by GW788388 enhances DENV-2 clearance in macrophages. Journal of Medical Virology. 2022;94:4359-4368
  192. 192. Pandey N, Jain A, Garg RK, et al. Serum levels of IL-8, IFNγ, IL-10, and TGF β and their gene expression levels in severe and non-severe cases of dengue virus infection. Archives of Virology. 2015;160:1463-1475
  193. 193. Kar M, Khan NA, Panwar A, et al. Zinc chelation specifically inhibits early stages of dengue virus replication by activation of NF-κB and induction of antiviral response in epithelial cells. Frontiers in Immunology. 2019;10. Epub ahead of print 1 October 2019:2347. DOI: 10.3389/fimmu.2019.02347
  194. 194. Kothai R, Arul B, Anbazhagan V. Anti-dengue activity of ZnO nanoparticles of crude fucoidan from brown seaweed S. Marginatum. Applied Biochemistry and Biotechnology. Epub ahead of print 19 May 2022. 2022;195:3747-3763. DOI: 10.1007/s12010-022-03966-w
  195. 195. Gordts SC, Renders M, Férir G, et al. NICTABA and UDA, two GlcNAc-binding lectins with unique antiviral activity profiles. Journal of Antimicrobial Chemotherapy. 2015;70:1674-1685

Written By

Naiara Clemente Tavares, Camila Sales Nascimento, Jaquelline Germano de Oliveira and Carlos Eduardo Calzavara-Silva

Submitted: 09 October 2023 Reviewed: 07 November 2023 Published: 05 January 2024