Open access peer-reviewed chapter

Dengue Immunopathogenesis: A Crosstalk between Host and Viral Factors Leading to Disease: Part I - Dengue Virus Tropism, Host Innate Immune Responses, and Subversion of Antiviral Responses

Written By

Henry Puerta-Guardo, Scott B. Biering, Eva Harris, Norma Pavia-Ruz, Gonzalo Vázquez-Prokopec, Guadalupe Ayora-Talavera and Pablo Manrique-Saide

Submitted: 11 May 2020 Reviewed: 09 June 2020 Published: 12 September 2020

DOI: 10.5772/intechopen.93140

From the Edited Volume

Dengue Fever in a One Health Perspective

Edited by Márcia Aparecida Sperança

Chapter metrics overview

1,042 Chapter Downloads

View Full Metrics

Abstract

Dengue is the most prevalent emerging mosquito-borne viral disease, affecting more than 40% of the human population worldwide. Many symptomatic dengue virus (DENV) infections result in a relatively benign disease course known as dengue fever (DF). However, a small proportion of patients develop severe clinical manifestations, englobed in two main categories known as dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Secondary infection with any of the four dengue virus serotypes (DENV1, -2, -3, and -4) is a risk factor to develop severe forms of dengue disease. DSS is primarily characterized by sudden and abrupt endothelial dysfunction, resulting in vascular leak and organ impairment, which may progress to hypovolemic shock and death. Severe DENV disease (DHF/DSS) is thought to follow a complex relationship between distinct immunopathogenic processes involving host and viral factors, such as the serotype cross-reactive antibody-dependent enhancement (ADE), the activation of T cells and complement pathways, the phenomenon of the cytokine storm, and the newly described viral toxin activity of the nonstructural protein 1 (NS1), which together play critical roles in inducing vascular leak and virus pathogenesis. In this chapter that is divided in two parts, we will outline the recent advances in our understanding of DENV pathogenesis, highlighting key viral-host interactions and discussing how these interactions may contribute to DENV immunopathology and the development of vascular leak, a hallmark of severe dengue. Part I will address the general features of the DENV complex, including the virus structure and genome, epidemiology, and clinical outcomes, followed by an updated review of the literature describing the host innate immune strategies as well as the viral mechanisms acting against and in favor of the DENV replication cycle and infection.

Keywords

  • dengue
  • immunopathogenesis
  • dengue shock syndrome
  • severe dengue
  • virus replication
  • cell tropism
  • innate immune response
  • antiviral response
  • immune evasion
  • complement
  • endothelial dysfunction
  • vascular leak

1. Introduction

Dengue is still considered the most prevalent viral disease transmitted by arthropod mosquitoes (e.g., Aedes mosquitoes), with 50–100 million dengue infections occurring annually, and a global incidence of 30-fold increase observed over the past 50 years [1, 2, 3]. Most of the dengue infections with any of the four dengue virus (DENV) serotypes [1, 2, 3, 4] result in inapparent, subclinical illness, or mild disease symptoms known as dengue fever (DF). However, some DENV infections can potentially evolve into more severe and fatal disease outcomes known as dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) [2]. DHF and DSS are mainly characterized by low numbers of circulating platelets (thrombocytopenia) associated with hemorrhagic manifestations and increased vascular permeability associated with endothelium hyperpermeability, resulting in plasma leakage, low blood pressure, and shock that can lead to death [2]. DENV infections frequently occur in the context of preexisting immunity, where the immune responses to prior DENV infection play an important role in determining the outcome of dengue epidemics and disease severity via antibody-dependent enhancement (ADE) and potentially harmful T cell responses in an original antigenic sin-dependent manner [4, 5, 6, 7, 8]. Both mechanisms lead to increased activation of immune cells, resulting in exacerbated immune responses or cytokine storms that cause endothelium dysfunction and vascular leak [9, 10]. Collectively, these host immunological responses are thought to create a physiological environment that promotes vascular permeability. However, the exact mechanisms underlying the capillary leak are probably more complex than a cytokine storm, and the risk of severe disease upon DENV infection cannot be explained completely by a misdirected host immune response to a prior infecting serotype; rather, disease severity appears to be determined by a combination of multiple host and viral factors leading to favorable and unfavorable interactions that regulate viral pathogenesis.

Despite considerable advances in understanding the immunological mechanisms activated during DENV infection, the pathogenic mechanisms underlying the alterations in permeability of the microvasculature remain unclear. The absence of a good animal model faithful to human disease and the limited knowledge of the factors regulating the intrinsic microvascular permeability in health have seriously hampered the research progress in this area. However, in the last decades, significant progress has been made regarding viral and host cellular components involved in DENV infection and disease [8]. The nonstructural protein 1 (NS1) protein of DENV and other related flaviviruses has been described as an essential cofactor in virus replication and assembly [11, 12]. Interestingly, the secreted form of NS1 is also implicated in immune evasion strategies via interaction with several proteins of the complement pathways that protect the virus-infected cells from the immune system processing [12, 13, 14]. Contrary, NS1 and anti-NS1 antibodies can also mediate complement activation that may alter capillary permeability [15]. Additionally, the soluble NS1 from DENV can interact with the surface of endothelial cells, immune cells, and platelets to cause endothelial barrier dysfunction and vascular leakage, and potentially hampers the coagulation cascades leading to hemorrhagic manifestations during DENV infection. These phenomena occur via activation of endothelial-intrinsic mechanisms leading to the disruption of the EGL and the integrity of the cell-to-cell contacts and/or induction of pro-inflammatory cytokines, chemokines, and proteases via the TLR4 activation of monocytes/macrophages that may act also on endothelial cells leading to endothelial hyperpermeability and vascular leak [16, 17, 18, 19, 20, 21, 22]. Furthermore, NS1 is highly immunogenic and conserved between the Flavivirus genus; thus, NS1 from other flaviviruses have been also reported to activate endothelial-intrinsic mechanisms causing vascular leakage in a tissue-dependent manner that mimics each flavivirus disease pathophysiology [17, 23, 24, 25]. Additionally, NS1 immunization using mouse models and DENV vaccination or natural DENV infection in humans can elicit antibodies’ responses that have been implicated in the contradictory roles of protection and pathogenesis in the infected host [25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42]. Today, no specific and effective vaccine candidate, antiviral therapy, or anti-inflammatory therapeutics have been licensed to combat dengue disease. An effective dengue vaccine is surely needed to avert the millions of dengue cases that occur around the world, continuously threatening with fatal outcomes. For decades, numerous experts in infectious diseases including clinicians, epidemiologists, basic scientists, and vaccine and drug developers have been trying to elucidate the ultimate mechanism of DENV pathogenesis leading to severe dengue disease. The NS1 protein of flaviviruses constitutes a unique “viral toxin” that seems to connect many of the already described DENV immunopathogenic mechanisms leading to severe dengue disease; thus, NS1 might represent the corner piece that completes the elusive dengue pathogenesis puzzle. Therapeutic approaches and vaccine development targeting NS1 may provide different opportunities for the future defeating of the global dengue disease. A better understanding of DENV immunopathogenesis will assist not only in the development of therapeutic interventions but also in the understanding of dengue vaccine efficacy or vaccine adverse events. This chapter briefly summaries the key clinical, virological, and epidemiological facts about DENV innate and humoral responses and gives an extensive update of insights about the viral and host factors that contribute to DENV pathogenesis leading to the development of severe dengue manifestations during DENV infection.

Advertisement

2. Dengue virus features: genomic organization, structure, and life cycle

Dengue virus (DENV) belongs to the genus Flavivirus (family Flaviviridae), a group of small (50-nm virion diameter) viruses containing a single-positive-stranded RNA genome [5′ capped, not 3′ poly(A) tail] which encodes for three structural proteins: capsid (C), membrane (M), and envelope (E) and seven nonstructural (NS) proteins named NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5 (Figure 1A, B) [43]. The DENV group is comprised of four evolutionary distinct but antigenically and genetically related viruses better known as DENV serotypes −1, −2, −3, and −4 (DENV-1-4), [44, 45]. These four established serotypes (DENV 1–4) share a high degree of sequence similarity between the genomes (~65–70%) [46], with average sequence identity between proteomes of 39–79% [47]. DENV is transmitted to humans by Aedes mosquitoes, mainly Aedes aegypti. However, the global distribution of Aedes albopictus (the Asian tiger mosquito), considered a secondary vector for DENV transmission, is changing rapidly and it is now becoming an increasingly important vector and a common cause of epidemics in Aedes aegypti-free countries [48, 49, 50]. Human-to-mosquito transmission occurs once the mosquito takes a blood meal from DENV-infected people who are viremic, which is normally up to 2 days before someone shows symptoms of the illness or up to 2 days after the fever has resolved. High viremia and high fever in patients are positively associated with a high rate of DENV transmission from humans to mosquitoes [51]. After feeding on a DENV-infected person, depending mainly on temperature, the virus rapidly replicates in the mosquito’s midgut, and within an average of 5.9 days, it disseminates to secondary tissues, including the salivary glands where the virus can be transmitted to the new host (extrinsic incubation period) [52]. Once infectious, the mosquito is capable of transmitting the virus for the rest of its life [53].

Figure 1.

Dengue virus genome organization, NS1 structure, and dengue epidemiology and disease outcome. (A) Schematic representation of the DENV genome and polyprotein. Dengue virus (DENV; genus Flavivirus, family Flaviviridae) is a positive-sense, single-stranded (∼11-kb length), and RNA-enveloped virus with an icosahedral capsid protecting the virus genome, which is transmitted by mosquitoes of the Aedes genus (Aedes aegypti, Ae. albopictus) and affects more than 40% of the human population worldwide living in tropical and subtropical areas. The viral RNA genome poses one single open reading frame encoding for one single polyprotein, which after being processed by cellular and viral proteases generates three structural proteins known as the capsid (C), the membrane (M), and the envelope (E) and seven nonstructural (NS) proteins known as NS1, NS2A, NS2B, NS3, NS4A, NS4B, and N5. The viral RNA contains a cap in the 5′-end, and it has no poly-a tail in the 3′-end. Several secondary structures or UTRs (untranslated regions) are found in both ends which have been shown to participate in viral replication as well as host adaptation. Of the structural proteins, the envelope (E) is the major protein on the virion, which participates in cellular receptor recognition to infect the host cells and the main target for adaptive immune responses in humans. On the other hand, the NS proteins play critical roles in the virus replication cycle and the subversion of the host antiviral responses, particularly those triggered by the innate immune response against DENV infection. (B) Of these NS proteins, NS1 is the only viral protein secreted by DENV-infected cells in which the plasma circulating levels are increased during the acute phase of DENV-infected patients undergoing severe disease. The NS1 protein circulates as a lipoprotein-like particle with a hexameric conformational structure containing three domains termed as the wing domain (here in yellow), the β-ladder domain (in blue), and the β-roll domain (in red) (NS1 hexamer is depicted in this figure). NS1 has been demonstrated to play critical roles in the formation of new viral particles, the evasion of the immune system, and very recently, it was implicated in modulating the virus pathogenesis of DENV that is mainly associated to its potential role in acting as a pathogen-associated pattern molecule (PAMP) which activates the production of pro-inflammatory host soluble factors such as cytokines, chemokines, proteases, etc. from the immune cells and directly triggers the barrier dysfunction of endothelial cell cultures in vitro and vascular leak in the mouse models in vivo. Taking these together, NS1 is now considered as a viral toxin not only of the DENV complex but of many of the closely related flaviviruses, including ZIKV, WNV, and YFV among others. (C) The DENV complex is composed of four serologically but antigenically related types of viruses, known as DENV serotypes (1, 2, 3, and 4). The primary infection with any of the four serotypes often cause inapparent, asymptomatic, or mild diseases that prime the immune system for a long-life immunity against the infecting DENV serotype (here DENV-1, in yellow) which is dominated by homotypic immune responses at mostly all levels, including antibody-B cells’ and T cells’ responses. During secondary infections, things get more complicated as infection with a different DENV serotype as the one from the primary infection; here DENV-2 (blue), DENV-3 (red), and DENV-4 (green) results in cross-reactive and heterologous immune responses that are considered the main risk factor to develop severe manifestations of DENV infections, including dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), which may lead to life-threatening health complications and sometimes, death. In the course of this chapter, we will explain how these epidemiological factors may be associated with the development of severe dengue disease that may be a consequence of a combined interplay set of immunopathogenic mechanisms triggered by the host immune response having DENV infection as the main trigger. The model of NS1 hexamer was built based on the crystal structure deposited in the Protein Data Bank (PDB4O6B). Molecular graphics were performed using the PyMOL molecular graphics.

After a mosquito bites a human, DENV is delivered into the dermis where it can infect/replicate in dendritic cells (DCs) (Langerhans cells) and keratinocytes residing in the basal and suprabasal layers of the epidermis [54, 55, 56] (Figure 2). Virus dissemination to the local lymph nodes occurs in association with infected migratory dendritic cells or as free viruses of the lymphatic fluid leading to viremia [57]. At this stage, mosquito saliva has shown to enhance the replication and pathogenesis of numerous arthropod-borne viruses, including DENV [58, 59, 60, 61, 62]. DENV infection cycle initiates with the virus attachment to the target cells [63]. The current model suggests that DENV uses both attachment factors and primary receptor(s) that facilitate virus recruitment on the cell surface, and later, internalization inside host cells via receptor-mediated endocytosis including clathrin-mediated and nonclassical clathrin-independent endocytosis [64, 65]. Despite this, the single definitive receptor mediating this critical step in the DENV replication cycle continues to be elusive. So far, numerous candidates have been described in the mammalian and mosquito cells, including glycosaminoglycans such as heparan sulfate and lectins, the adhesion molecule of dendritic cells (DC-SIGN), the mannose receptor (MR) of macrophages, the lipopolysaccharide (LPS) receptor CD14, and stress-induced proteins such as the heat-shock proteins 70 and 90 and the endoplasmic reticulum (ER) chaperonin GRP78 [64, 65, 66, 67, 68]. This suggests that DENV may not use a unique, specific receptor to enter cells, but recognizes diverse molecules, both in the vertebrate and mosquito hosts, which can potentially explain the broad tissue range that defines DENV tropism and infection.

Figure 2.

Dengue virus infection, pathogenesis, and immune responses in the skin. The skin represents the first line of defense of the human body against pathogens such as viruses, as it generates early immune responses, aiming to protect humans against cutaneous and systemic infection (1). The human skin constitutes a complex organ nicely structured in three escalated layers known as the epidermis, which is composed of closely packed epithelial cells including keratinocytes, melanocytes, and Langerhans cells (LCs), a specialized type of dendritic cell (DC) that constantly probes for antigen in the most exposed, superficial layer of the skin; the dermis, which is made of dense, irregular connective tissue, blood vessels, and other structures; and the inner hypodermis, which is composed mainly of loose connective and fatty tissues (2). Upon disturbance of the epidermal barrier by mosquito blood-feeding, DENV-infected mosquitoes inoculate newly generated infectious virus particles along with mosquito saliva in which a complex mixture of proteins that exerts profound effects in the human immune system allows the acquisition of the mosquito blood meal from its host by circumventing vasoconstriction, platelet aggregation, coagulation, and inflammation or hemostasis (3). In the skin, major constituents of the innate immune system include phagocytic cells such as macrophages, neutrophils, and DCs as well as innate leukocytes such as natural killer (NK) cells, mast cells (MCs), basophils, and eosinophils. Also, epidermal keratinocytes act as active innate immune cells. In response to sensing pathogen-associated molecular patterns (PAMPs) expressed by microbes and host danger molecules, innate immune receptors present on keratinocytes and APCs become activated, causing the release of inflammatory cytokines and antimicrobial peptides (4). At the site of inoculation in the skin, the key targets of DENV infection are immune cells of the myeloid lineage, including various subsets of DCs, monocytes/macrophages, and MCs (5). Despite this, limited virus particles are thought to be deposited in the epidermis during mosquito blood-feeding (3), and in that location, Langerhans cells as well as keratinocytes are considered target cells (6). In the dermis, DCs and monocytes/macrophages are also prime infection targets (7). MCs are not substantially infected in the skin (8). However, exposure to DENV triggers an augmented activation of MCs leading to degranulation and release of de novo-synthesized inflammatory and vasoactive mediators, including proteases, leukotrienes, and histamine that, along with some vasoactive molecules and maybe the secreted NS1 originated from infected mosquito cells in the salivary glands, promote edema within the site of infection as a consequence of the increased microvascular permeability (9). Activation of MCs also induces the secretion of cytokines and chemokines that leads to the recruitment of NK cells, neutrophils, and monocyte-derived dendritic cells (mDCs) to the site of infection (10). Already in the skin, mDCs can serve as targets of infection, allowing the amplification of the virus in the skin, while natural killer (NK) cells, natural killer T (NKT) cells, and CD8+ T cells can kill DENV-infected cells and promote virus clearance in a cellular cytotoxic-dependent manner (11). Once human skin is infected, DENV-infected DCs take virus into the draining lymph nodes using afferent lymphatic vessels where they spread DENV infection and most importantly activate antigen-specific CD4+ and CD8+ T cells which initiates the adaptive immune response (12, 13). In the T cell zones, activated T cells become effectors cells to promote the development of DENV-specific memory B cells and plasma cells in the germinal center of LN. Activated T cells can reenter circulation and potentially return to the skin for virus clearance during subsequent DENV infections, playing an important role in protecting against DENV (14). In the skin, in addition to the mosquito saliva, DENV infection of target cells such as DCs, monocytes/macrophages, and MCs can be also modulated by the presence of preexisting antibody responses against previous infections with distinct DENV serotypes or other closely related flaviviruses, in an antibody-dependent enhancement (ADE) manner (See DENV-ADE in Part II for more details). For MCs and DCs, DENV-ADE is possible through Fc-γ-receptors (FcγRs). Besides, MCs degranulation can be enhanced through cross-linking of FcεRs when bound to DENV-specific IgE, leading to augmented MC activation and presumably immune-mediated vascular injury (15). After skin infection, DENV must achieve systemic infection to complete its transmission cycle by infecting new mosquito hosts. Infection of secondary LNs following infection of the draining LN are considered the amplification centers for DENV that contributes to the systemic infection and virus transmission (16).

After the internalization of the virion, a fusion between the viral E protein and the endosomal membrane mediates the access of the viral genome into the cytoplasm [43, 65]. The E protein is a glycosylated viral protein and a member of class II viral membrane fusion protein family [43, 69]. The crystal structure of E glycoprotein ectodomain revealed three domains contributing to the β-barrel central structure of the protein (domain I, DI), permitting the fusion of viral and cellular membranes during virus entry (domain II, DII, and fusion loop), and a structural basis for immune recognition and cellular receptor binding (domain III, DIII) [43, 70]. The low pH of the endosomal compartment induces conformational changes in the E glycoprotein, which allows the fusion of the viral and host membranes [43, 69]. This results in the release of the viral RNA genome into the cytoplasm. The single-stranded positive-sense RNA immediately acts as a messenger RNA, which can be subsequently translated by cellular machinery to generate viral polyproteins, subsequently processed by both cellular and viral proteases to generate mature viral proteins [69, 70]. In this stage, the nonstructural proteins have been shown to induce massive remodeling of ER membranes, manifesting as convoluted membranes and vesicle packets (VPs) to form a dynamic and membrane-bound multi-protein assembly, named the replication complex (RC) where the genome is replicated, and new viral RNA copies are incorporated into nascent particles [71, 72]. Viral RNA synthesis relies on NS5, the RNA-dependent RNA polymerase as well as on critical RNA secondary and tertiary structures [73, 74, 75]. NS3 is a protease-helicase which together with its cofactor NS2B, participates in the processing, efficient RNA synthesis, and capping of the viral polyprotein [72, 76, 77]. NS2A recruits nascent RNA as well as C-pre-M-E [78]. NS1 interacts with structural proteins and NS4A-2 K-4B to facilitate the production of infectious virus particles [11, 79]. Because of their critical roles in the DENV replication cycle, NS2B, NS3, and NS5 along with NS4B are the main focus to design new inhibitors for antiviral therapy against DENV and other related flaviviruses [80, 81, 82, 83]. Assembled viruses are transported through the trans-Golgi network (TGN) where under acidic conditions, a cellular protease, furin, cleaves pre-M, allowing full maturation of infectious virions that will be finally released via exocytosis [84].

Advertisement

3. Dengue virus infection, epidemiology, and clinical features

Dengue is the arboviral infection with the highest disease incidence worldwide, with 2.5 billion people living in dengue-endemic tropical and subtropical regions [1, 85, 86]. In the last four decades, the geographical spread and intensity of dengue have grown dramatically around the world accompanied by the wide distribution of the two main vector mosquitoes, Aedes aegypti and Aedes albopictus, which today are fully adapted to human dwellings creating new opportunities not only for DENV but also for other arthropod-transmitted viruses (arboviruses) transmission, such as Zika virus (ZIKV) and chikungunya virus (CHIKV), within human populations. These features along with the continuous growing of urbanization, globalization, and the lack of effective mosquito control represent some of the critical factors that have contributed to the emergence and reemergence of mosquito-transmitted viruses around the world [48, 87, 88].

Infection with any of the four DENV serotypes results in a diverse range of symptoms going from mild undifferentiated fever to life-threatening manifestations, which are characterized by increased vascular permeability, hemorrhage, and shock [89] (Figure 1C). In 1997, the World Health Organization (WHO) classified symptomatic DENV infections into three categories and subcategories known as dengue fever (DF), dengue hemorrhagic fever (DHF), and dengue shock syndrome (DSS). After an incubation period of 3–7 days, symptoms start suddenly and follow three phases: an initial febrile phase, a critical phase around the time of defervescence, and a spontaneous recovery phase [89]. Classical DF is an incapacitating disease that affects older children, adolescents, and adults, mainly characterized by the abrupt onset of fever (up to 40°C) and severe headache, accompanied by retro-orbital pain, myalgia, arthralgia, gastrointestinal discomfort, and transient rash [89]. In turn, DHF and DSS can rapidly deteriorate, progressing to hemorrhage with or without vascular leak after an early acute-onset febrile period, particularly during defervescence, where the symptoms are similar to those presented during classical DF. DHF and DSS are classified into four subcategories or grades (I–IV), where grades I and II (DHF) are represented by mild cases presenting some bleeding manifestations without shock (petechiae, purpura, ecchymosis, bruising, epistaxis, etc.), whereas III and IV (DSS) are more severe and accompanied by severe hemorrhagic manifestations and thrombocytopenia (platelets counts: <100,000 platelets/μL) and evidence of increased vascular permeability (ascites, pleural effusion, increased hematocrit concentrations, and severe abdominal pain) during a critical period, sometimes accompanied with a profound and prolonged shock that potentially leads to death [90]. In this critical stage, liver failure, myocarditis, and encephalopathy often occur with minimal associated plasma leakage [89]. In 2009, the WHO revised the classification system for dengue and established new guidelines that replaced the more complicated dengue fever/dengue hemorrhagic fever (DF/DHF) system to separate patients enduring severe disease from those with non-severe manifestations. This new guideline defined two new major entities—dengue and severe dengue—which encompasses a set of “warning signs” intended to help clinicians identify the patients likely to develop complications during the critical phase of the illness [89].

Currently, there is no effective and safe vaccine or FDA-approved specific antiviral drug options to combat dengue disease, with treatment being purely supportive [91]. Prevention or reduction of DENV transmission by implementing combined effective control strategies remains as the primary approach to be used to prevent DENV transmission within human populations [92]. With the majority of DENV infections being asymptomatic (70–80%), and most symptomatic infections not progressing to severe disease [3], the global distribution of dengue remains highly uncertain as the actual numbers of dengue cases are underreported and many cases are misclassified. One recent study estimate indicates that 390 million DENV infections occur annually with more than 500,000 cases of hospitalizations and more than 25,000 deaths (2.5% case fatality, annually) [1]. A different study estimated that 3.9 billion people living in 128 countries are at risk of being infected with dengue viruses [85]. These studies demonstrate the worldwide expansion of the dengue disease and the establishment of an increasingly important infectious disease of global public health significance.

Advertisement

4. Dengue immunopathogenesis and severe disease: host and viral factors

The hallmark of severe dengue is the transient perturbation in the integrity of the endothelium lining the inner side of blood vessels as well as the alteration in the coagulation cascade leading to shock and severe hemorrhage manifestations [9, 89]. Increased vascular permeability in severe dengue results in decreased circulating plasma volume, haemoconcentration, and pleural and peritoneal effusions that result in severe life-threatening shock [93, 94, 95, 96]. Numerous epidemiological pieces of evidence indicate that appearance of the life-threatening manifestations during severe dengue occurs shortly after the defervescence stage of dengue disease, when the peak of viremia passed, meaning that host innate and adaptive immune responses have cleared the virus from host tissues [97, 98]. At this time, a transient vascular leakage pathology is observed followed by a rapid recovery in association with the late febrile phase. This association led to the suggestion that the key biological mechanisms such as alterations on the vasculature that leads to the pathogenesis of clinical complications during DENV infection are rather functional than the structural changes in the endothelium and are primarily a consequence of short-lived biological mediators closely linked to the host immune responses [93, 94, 95, 96].

Although many severe infections occur upon secondary encounters with heterologous DENV serotypes [9, 99], suggesting an immune-mediated process is involved, the multifactorial immunopathogenic process of DENV infection implies a complex interaction between distinct viral and host processes that sometimes leads to increased virus infection, exacerbated immune responses, and the appearance of life-threatening severe manifestations such as severe plasma leakage, hemorrhage, and organ failure. Higher virus pathogenicity (virulence), preexisting serotype cross-reactive antibodies, activation of DENV-infected immune cells [e.g., monocytes and mast cells (MCs)], T cell responses, activation of complement pathways, the potential infection of endothelial cells, and the new pathogenic roles of the secreted NS1 of DENV may work synergistically to induce the release of vasoactive cytokines which results in increased endothelial permeability causing vascular leakage and pleural effusion, which are still considered pathognomonic features of severe dengue that leads occasionally to shock and death [8, 9, 35, , 96, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110]. In this section, we highlight in two parts I and II, the immunological events elicited by DENV infection, which have been suggested to play a key role in the development of severe dengue manifestations.

4.1 Dengue virus tropism and infection of immune cells

Numerous in vitro studies have shown that DENV is able to infect a variety of cell types including epithelial cells, endothelial cells, hepatocytes, muscle cells, dendritic cells, monocytes, B cells, and mast cells [65, 66, 111, 112, 113, 114, 115, 116, 117]. Several autopsies and ex vivo studies have found the presence of DENV antigens (e.g., envelope protein, NS3) in some tissues such as the skin, liver, spleen, lymph node, kidney, bone marrow, lung, thymus, and brain [56, 67, 68, 118, 119, 120, 121, 122]. However, infectious virus particles have not always been isolated from all these organs but only from the liver and peripheral blood mononuclear cells (PBMCs), suggesting that: (a) the presence of DENV antigens such as the structural proteins E, pre-M, and C in several organs may not always be associated with the evidence of productive viral infection and severe organ pathology and (b) the immune cells and liver may be the main targets for DENV replication during the dengue disease [67]. In animal models such as the alpha/beta (IFN-α/β)-deficient mice (Ifnar −/−) and nonhuman primates, DENV has been recovered from the spleen, liver, peripheral lymph nodes, and the central nervous system [123, 124, 125, 126, 127]. However, the absence of an appropriate animal disease model has largely hampered with the understanding of the role played by DENV tropism in vivo. Sustained viral replication and severe manifestations have been observed in Ifnar −/− mice after infection with DENV, which gives a clear advantage to study DENV pathogenesis in vivo, but the absence of intact IFN signaling is a limitation that must be considered when interpreting data [128].

The fact that DENV can infect many mammalian and insect cell types in vitro and in vivo suggest there are different molecules or cellular routes that might be controlling virus attachment and internalization, resulting in productive infection [63]. Numerous studies have shown that C-type lectins including DC-SIGN (CD209) and C-type lectin domain family 5, member A (CLEC5A) expressed on dendritic cells and macrophages act as cellular receptors for DENV [129, 130, 131]. Other extensively studied DC receptors are the mannose receptor (MR), Langerins, Fc-receptors, TIM3, TIM4, and AXL [63, 65, 132, 133]. Contrary to the DC-SIGN that may primarily function as a viral attachment factor, DENV binding to CLEC5A (C-type lectin domain family 5, member A), highly expressed by monocytes, macrophages, neutrophils, and dendritic cells, has been shown to induce the production of antiviral and pro-inflammatory cytokines suggesting that this C-type lectin may act as a cognate receptor for dengue virion [131, 134]. These cytokines include type I IFNs and chemotactic factors such as migration inhibition factor (MIF), monocyte chemotactic factor (MCP), and IL-8 [102, 134]. DENV infection of DCs also induces the production of matrix metalloproteinases (MPPs), MMP-2 and MMP-9, which induces migration of DCs to lymph nodes where virus further replicates before it disseminates into the blood circulation [135]. In the skin, DENV also infects mast cells that can be activated leading to degranulation and increased secretion of various inflammatory cytokines (IL-1, IL-6, TNF-α, and IFN-α), chemokines (CCL5, CXCL12, and CX3CL1), and chymase, the latter being a protease found circulating at high levels in the blood of dengue patients, suggesting a potential role in the development of severe dengue that contributes to vascular leakage [115, 136, 137, 138, 139, 140]. All these innate immune processes together lead to an antiviral state in nearby cells, generating an inflammatory response and recruitment of natural killer (NK) cells to combat DENV infection [54, 141].

Along with DCs, monocytes and macrophages are also the primary targets of DENV infection [142, 143]. In lymphoid and nonlymphoid tissues, macrophages are considered the primary reservoirs of DENV after its dissemination from the skin [144]. Macrophages susceptible to DENV have been found in different organs in the mouse models or human autopsies, namely, Kupfer cells in the liver, alveolar macrophages in the lungs, dermal macrophages, microglial cells (brain and spinal cord), and monocytes in the peripheral blood [118, 120, 122, 145, 146, 147]. Comparable to DENV infection of DCs, DENV can use an array of cell surface receptors to infect monocytes and macrophages, including mannose receptor (CD205), CD14-associated protein, heat shock proteins (HSP70/HSP90), DC-SIGN (CD209), CD300a, AXL, TIM4, PD1, and the Fc receptors, particularly FcγRI (CD64) and FcγRII (CD32, 63). These two Fc-Rs play major roles in enhancing DENV infection of monocytes and macrophages, particularly during secondary infections [148, 149, 150, 151].

Other populations of immune cells including NK cells can also be activated during DENV infection, particularly in patients with DHF compared to those with DF [141, 152, 153]. Additionally, B cells and T cells have been studied to test permissiveness to DENV, but these studies have resulted in contradictory results [154, 155, 156]. In vitro studies using B cell and T cell lines (e.g., Raji cells, Daudi, and Jurkat) and primary B cells derived from healthy human peripheral blood mononuclear cells (PBMCs) have revealed the potential role of these cells in DENV replication, both in presence and absence of heterologous antibodies [67, 155, 157, 158, 159]. Additional studies using a humanized mouse model found that DENV infected both B and T cells accompanied by an important production of pro-inflammatory cytokines such as IL-6 and TNF-α, like monocytes and macrophages [160]. Despite this evidence, the role of lymphoid cells such as B and T cells in DENV tropism and replication needs further exploration.

4.2 DENV infection and the host innate immune responses

Although plasma leakage in severe dengue occurs at the end of the acute illness, there is substantial evidence that the pathophysiologic processes start at the earliest stages of DENV infection [95, 96]. Introduction of DENV particles along with mosquito saliva triggers a variety of host innate immune responses leading to the production of antiviral and pro-inflammatory cytokines mostly from the immune cells exposed to DENV [57, 62, 138]. At this stage, innate immune cells are the first to respond to infection through stimulation of patterns recognition receptors (PRRs) recognizing pathogen-associated molecular patterns (PAMPs) as well as endogenous molecules released from damaged cells, termed damage-associated molecular patterns (DAMPs) [161, 162]. PRR recognition triggers the production of cytokines and chemokines, which induces a local antiviral state [54, 55]. This local innate response could potentially play an important role in modulating local viremia and virus dissemination by recruiting susceptible target cells for DENV infection at the inoculation site [57, 62, 144].

PRRs include transmembrane proteins such as the Toll-like receptors (TLRs) and C-type lectin receptors (CLRs) as well as cytoplasmic proteins such as the retinoic acid-inducible gene (RIG)-I-like receptors (RLRs) and NOD-like receptors (NLRs) [161]. These are an essential part of the innate immune response against the virus, sensing viral replication in the cytoplasm [161, 163]. The PRRs that are associated with DENV recognition after infecting target cells are the cytoplasmic retinoic acid-inducible gene I (RIG-I) and the melanoma differentiation-associated protein 5 (MDA5) and the endosomal Toll-like receptor 3 (TLR3) and TLR7 [164, 165, 166]. Recognition of DENV RNA by TLR-3 results in the production of type I IFN and chemokines such as IL-8 via sensing of phosphate-containing RNA and long double-stranded RNA (dsRNA) in the cytoplasm or inside endosomal compartments [167, 168]. DENV infection in nonhuman primates demonstrated that the administration of TLR-3 and TLR7 agonists resulted in significantly decreased viral replication and increased production of pro-inflammatory chemokines as well as increased production of antibodies targeting DENV [169], indicating a protective role for TLRs during DENV infection.

Additional pathways such as the cyclic GMP-AM synthase (cGAS), a DNA-sensor pathway which triggers the simulator of IFN genes (STING) pathway are also activated during DENV infection leading to the production of type I IFN and activation of TLRs (TLR9), an endosomal PRR that recognizes cytoplasmic DNA originated from mitochondrial damage [170, 171, 172]. In addition to type I IFN production, small RNAs such as micro RNAs (miRNA) and the complement system are important components of the innate immune response against viral infections [173, 174]. miRNAs are processed by and interact with the proteins in the RNA interference (RNAi) pathway, such as Dicer, Drosha, Argo1, and Argo2 [175]. RNA interference (RNAi) is an important antiviral defense response in plants and invertebrates [176]. In DENV infection, knockdown of these components resulted in increased DENV replication in mammalian cells, suggesting that the RNAi pathways may play important roles in the cellular anti-DENV responses [177, 178]. Additional evidence showed that DENV can interfere with RNAi pathways in human hepatocytes cells via NS4B and subgenomic flavivirus RNA (sfRNA) interactions with Dicer’s ability to process small RNA in vitro [179]. sfRANs are abundant noncoding RNA sequences derived from the stalling of the host 5′-3′ exoribonuclease XRN1/Pacman in the 3′-untranslated regions (3′UTRs) of the viral genomic RNA [180]. sfRNAs have been shown to block exonuclease XRN1, increasing the overall messenger RNA stability within the host cell which may also benefit the viral RNA [181, 182, 183]. However, evidence for RNAi contribution to mammalian antiviral defense are few and still controversial [179]. miRNAs have been shown to regulate TLRs and IL-1 signaling pathways in response to viral infection, which provides control of host innate immune responses [184]. So far, there have been reported several cellular miRNAs (miRNAome) that are modulated during DENV infection of mammalian cells, mainly related to the regulation of IFN-β signaling pathways [185, 186, 187, 188]. Some of these miRNAs have been proposed to be used as biomarkers in dengue-infected patients [189, 190]. Interestingly, modulation of microRNA expressions have been also described upon DENV infection of insect cells (e.g., C6/36 cells) as well as adult vector mosquitoes such as Aedes albopictus [191, 192, 193], suggesting that DENV might also regulate the activation of these antiviral RNAi pathways in vector mosquitoes, potentially avoiding viral clearance that promotes viral replication and transmission [194]. Despite RNAi constituting an evolutionarily conserved phenomenon of the mosquito-innate immune response to virus infections [195], viruses have found ways to subvert the RNAi-mediated antiviral responses in vector mosquitoes to manipulate miRNA profiles to their own benefit [196].

Regarding the complement system, this multifaceted pathway has been shown to limit DENV replication; however, excessively activated complement components have been also associated with disease severity [197]. The complement cascade constitutes an integral component of the immune system, composed of many plasma proteins that once activated can initiate a proteolytic cascade, resulting in the release of chemokines, facilitation of particle phagocytosis via opsonization, and deposition of the cell-killing membrane attack complex (MAC) designed to target and destroy foreign pathogens such as viruses [174]. Activation of the complement system occurs via three convergent pathways: the classical, the lectin, and the alternative pathways [174]. In vitro experiments showed that DENV replication enhances complement activation [197, 198, 199]. Additionally, clinical and in vivo studies have shown that excessive consumption of some complement components (e.g., C3, C4, and factor B) contributed to severe manifestations by increasing the levels of complement-activated products which enhance vascular permeability to cause severe dengue disease [9, 108, 109, 200]. In fact, increased circulation of anaphylatoxins (C3a, C4a, and C5a) in the blood of severe patients correlated with symptoms of vascular leakage [109, 200]. In autopsy studies from children who died of acute severe dengue manifestations (DHF/DSS), augmented deposition of complement components from both classical and alternative pathways were found on hepatocytes which results in severe liver damage and death [120]. Altogether, these data support the hypothesis that exacerbated complement activation influences dengue disease immunopathogenesis leading to disease severity [197].

4.3 DENV subversion of antiviral responses

The first barrier to overcome for successful viral infection is the rapid innate immune responses of the host, including type I IFNs, inflammatory cytokines, complement responses, NK cells, apoptosis, and autophagy [201, 202]. These innate immune responses are meant to defeat viral infections by engaging specific viral components (e.g., RNA and DNA) leading to activation of immediate protective defense mechanisms such as the rapid recognition of PAMP in nonimmune and innate immune cells [161]. IFN production is a key goal of PRR activation for viral pathogens, and DENV is highly susceptible to effective induction of both type I (IFN α/β) and type II (IFN γ) interferons [124, 203, 204]. Accordingly, in vivo DENV infection of wild-type mice causes little disease; in contrast, in mice lacking of type I IFN receptors (IFANR), DENV infection causes mortality [126].

Secreted type I IFNs trigger autocrine and paracrine induction of cellular antiviral responses and warning signals to noninfected adjacent cells, such as the expression of the interferon stimulated genes (ISGs) [205, 206]. ISGs have been shown to exert numerous antiviral effector functions, many of which are still not fully described [207]. Upon DENV infection, RLRs are activated to trigger antiviral responses based on the induction of type I IFN and pro-inflammatory cytokines [208]. The binding of type I IFN with its receptor activates multi-subsets of ISGs through JAK-STAT signaling which amplifies and sustains the initial antiviral responses [207, 209, 210]. However, ISGs can also be activated in IFN-independent pathways during DENV infection [211]. DENV infection has been shown to trigger the transcriptional activation of ISGs in vivo and in vitro [208, 212, 213, 214, 215]. For instance, a tripartite motif (TRIM) protein encoding gene, TRIM69, is induced during DENV infection as an ISG. TRIM69 restricts DENV replication by direct interaction with DENV NS3, which mediates its polyubiquitination and degradation in a process called ISGylation [216]. In addition to ISGs, activation of the transcription factors IRF-3, IRF-7, and NF-κB through either the TLR or RIG-I/MDA5 pathways results in the production of type I IFN which contributes to anti-DENV immunity [217, 218]. IRF-3 and IRF-7 are part of the interferon regulatory factors (IRFs) considered the master regulators of the type I IFN production that contribute to the suppression of viruses [219]. Due to the central importance in viral defense, many pathogenic viruses, including DENV, have evolved mechanisms to suppress IRF signaling. In the case of DENV, the nonstructural proteins restrict IRF3 and IFN response which facilitate DENV replication and virulence [220].

In recent years, considerable advances have been made toward understanding of the specific IFN antagonistic mechanisms evolved by DENV to subvert these intracellular antiviral mechanisms and directly inhibiting these cellular signaling cascades, which results in enhanced virus infection, pathogenesis, and disease [167, 221]. This is supported by the increased susceptibility of mice deficient in IFN-α/β and IFN-γ receptors (AG129) to DENV infection as compared to wild-type mice [124, 126, 127]. Although IFN response is antagonized in mouse, human cells still induce high levels of IFN production in response to DENV, so this pathway is not entirely abrogated in humans during infection [203, 222]. Accordingly, humans infected with DENV have high levels of circulating of type I and type II IFNs [223, 224, 225]. Strong IFN-α responses have shown to correlate with milder dengue clinical conditions [226]. Similarly, the levels of the dengue-related gene expression of ISGs have been reported to be lower in patients with more severe disease [227, 228, 229] suggesting that DENV may abrogate IFN responses to facilitate viral infection which results in severe manifestations.

From the viral perspective, DENV uses its nonstructural (NS) proteins to block and inhibit the antiviral sensing pathways in infected cells. NS2a, NS3, NS4a, NS4b, and NS5 prevent the virus from being sensed by RIG-I, inhibiting IFNβ induction [230, 231, 232, 233]. NS2a, NS4a, and NS4b complex inhibits STAT1 signaling after IFNAR activation in vitro [233, 234]. NS5 induces proteasomal degradation of STAT2 which inhibits IFN-mediated response [230]. NS2b induces degradation of cGAS, which prevents DNA sensing resulting from mitochondrial damage [170, 171, 235]. The NS2b/3 protease complex cleaves STING which inhibits IFN production [236]. This phenomenon has been shown to occur in human but not nonhuman primates, suggesting that DENV may have evolved to increase viral titers in human populations, while maintaining decreased titers and pathogenicity in rare animals would serve as a sustainable reservoir in nature [237].

In addition to NS proteins, flavivirus sfRNAs have been described to regulate the innate immune responses via binding and inactivating RNA-binding proteins which are crucial for innate immunity [180, 238]. DENV 3′UTRs possess RNA structures necessary for viral genome cyclization, viral RNA synthesis, translation, and replication [239]. sfRNAs regulate the pathogenicity in both mammalian and mosquito cells after interacting with proteins such as TRIM25 to inhibit RIG-I signaling and translation of ISGs [73, 240, 241]. Interestingly, reduced IFN responses have been found during DENV outbreaks where the infecting DENV serotype produced greater levels of sfRNA than the less pathogenic strains [100, 240]. Thus, high levels of sfRNAs may cause an epidemiological fitness of DENV, which results in lower stimulation of RIG-IMDA5 RNA sensors and reduced production of IFN, causing higher viremia levels that could be translated in more infections and severe diseases.

On the other hand, DENV utilizes the endoplasmic reticulum (ER) of host cells for replication and assembly. In this process, the ER undergoes extensive rearrangements and expansion that requires de novo synthesis of viral proteins [71]. Accumulation of unfolded proteins in the ER lumen leads to an unfolded protein response (UPR), a pro-survival cellular reaction induced in response to DENV-mediated ER stress [242, 243]. DENV has evolved to manipulate the UPR to cope with ER stress which hijacks the host cell machinery to evade the host immunity, facilitating viral replication [112]. Distinct in vitro and in vivo studies have shown that DENV induced ER stress and manipulates the host metabolism and protein production by increasing the autophagy (lipophagy) activity, viral replication, and pathogenesis through UPR signaling pathways [244, 245, 246, 247]. Autophagy is the lysosomal degradation of cytoplasmic contents, which results in the recycling of cellular macromolecules as well as the activation of cellular host responses to starvation or stress [248]. Autophagy has been implicated as an innate immune response that would engulf and destroy pathogens by degrading cytosolic contents [249, 250]. In DENV infection, functional autophagy components have been shown to either promote or restrict viral RNA replication and virus production [251, 252, 253]. However, DENV has found ways of preventing autophagic processing and degradation of viral components [254, 255]. Several studies have linked DENV induction of autophagy to the regulation of lipid metabolism, leading to increased degradation of lipid droplets that produces more fatty acid material important for viral replication [247]. In this process, the NS3, NS1, and C proteins of DENV have been found to increase fatty acid biosynthesis and recruitment of lipid droplets to the DENV replication complex, facilitating viral particle assembly [256, 257].

Furthermore, several studies have shown that lipids and lipoproteins play a role in modifying DENV infectivity in both mammal and insect cells in vitro [258, 259]. Modulation of cholesterol levels in the host cells facilitates viral entry, replication, virus assembly, and control type I IFN response [260, 261]. This modulation involves the regulation of cholesterol levels, expression of cholesterol receptors as well as changes in cholesterol synthesis related to important modifications in the cellular metabolism [114, 262, 263]. Interestingly, clinical studies have found that levels of total serum cholesterol and LDL-C levels are modulated over the course of dengue illness, with generally lower levels associated with increased dengue severity [264, 265, 266]. In general, low cholesterol levels have been associated with critical illness related to sepsis and vascular disorders [267]. Thus, the association of cholesterol with severe dengue outcome may be an important indicator of the pathophysiology of DHF/DSS.

About the complement pathway, DENV has evolved strategies to limit recognition and activation of the complement cascade [108, 165]. NS1 is the only flavivirus protein that is secreted by infected cells and has been shown to modulate the complement pathway [14, 268]. NS1 promotes efficient degradation of C4 to C4b to protect DENV from complement-dependent neutralization [13, 269]. The NS1 protein of DENV and other flaviviruses such as WNV NS1 interacts with some components of the alternative complement pathway such as the C3bBb convertase, which limits the formation of C5b-9 membrane attack complex (MAC) [268, 270]. Additional studies have found that NS1 proteins from DENV, WNV, and YFV all attenuate classical and lectin pathway activation by directly interacting with C4, which reduces C4b deposition and C3 convertase (C4b2a) activity [13, 271]. Also, anti-NS1 antibodies have been shown to induce complement consumption and C5b-9 generation [272]. Overall, through protein-to-protein interactions between the viral and host factors involved in antiviral responses and careful manipulation of cellular processes, such as ER expansion, autophagy and lipid metabolism, and complement pathways, DENV hijacks many host antiviral responses to facilitate virus replication leading to pathogenesis.

Advertisement

5. Concluding remarks (Part I)

Dengue is the most prevalent arboviral disease transmitted by mosquitoes, which poses an enormous burden to the public health systems worldwide as more than 40% of the world population is at risk of infection. The infection with any of the four DENV serotypes (DENV1–4) can lead to a wide spectrum of clinical manifestations that range from the asymptomatic or inapparent to moderate flu-like symptoms, known as dengue fever (DF), and life-threatening manifestations identified by the WHO, known as the dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), also known as severe dengue, with or without warning signs. In endemic areas where multiple DENV serotypes can seasonally circulate, distinct epidemiological studies have demonstrated that an individual human being can be exposed to sequential infections with distinct DENV serotypes, which poses a risk of developing severe manifestations such as DHF/DSS. This phenomenon has been attributed to the potential enhancement activity that the preexisting antibody response elicited from a previous infection with one serotype (e.g., DENV-1) may have on the infection with a different serotype (e.g., DENV-2). This process leads to an increased viral burden that triggers a series of immunological and cellular events (e.g., ADE, cytokine storm, skewed T cell responses, and complement pathways), which despite being intended to prevent the invasion and infection of the infecting viral pathogens, can induce host tissue damage leading to pathology and disease. The cellular and molecular mechanisms involved in this phenomenon will be explained in more detail in the Part II of this chapter entitled “Adaptive immune response and NS1 pathogenesis.”

As an arthropod-transmitted virus (arbovirus), DENV is initially transmitted by an infected vector mosquito in which the virus has already been amplified after replication in its distinct tissues, starting at the midgut to finalize in the salivary glands, where a new transmission cycle begins after blood feeding from a new host (Figure 2). Following inoculation from the bite of an infected mosquito, viruses undergo replication in the local tissues such as the skin. In the skin, infectious virus particles along with mosquito saliva components including proteases and immunomodulatory proteins among others are sown in the epidermis and dermis, leading to an activation of a cascade of events including the recruitment of skin resident cells (e.g., Langerhans cells, mast cells, and keratinocytes) and new cells (e.g., T cells and neutrophils) into the site of the infection that later serve as viral targets for viral replication. After infection of target cells, sensing of viral products (e.g., PAMPs and DAMPs) results in the activation of innate immune responses (e.g., type I IFN chemokines), the first line of defense, which establishes inflammatory and antiviral states intended to prevent the virus to colonize and to replicate in the skin; however, DENV has elaborated several pathogenic mechanisms to hijack these responses and escape from the normal immune system processing, which results in its dissemination and seeds into the lymph nodes. There, DENV further replicates in monocyte lineage cells, resulting in a primary viremia after its systemic dissemination through the circulatory bloodstream, which results in the subsequent infection of peripheral tissues such as the liver, spleen, and kidney. Overall, the skin represents not only the first line of defense against arboviruses but also the main place where viruses have learned to evade the host immune responses leading to invasion and dissemination toward the establishment of systemic host infection, which will potentially assure subsequent virus transmission into a new host. In this Part I of the chapter entitled “Dengue virus tropism, host innate immune responses, and subversion of antiviral responses,” we discussed the distinct features of DENV as well as the biological and molecular mechanisms that can tilt the balance to either a local viral infection and dissemination through the skin or to the control and prevention of viral infection by the innate and adaptive immune responses at the site of the infection. Thus, the immunopathogenesis of arboviruses such as DENV in the skin is a critical step and must be a focus of future studies intended to reduce/block arthropod-borne transmission into humans.

Advertisement

Acknowledgments

The authors would like to acknowledge to the Canadian Institutes of Health Research and the International Development Research Centre (Projects 108412 and 109071-002) and the Fondo Mixto CONACYT-Gobierno del Estado de Yucatan (Project YUC-2017-03-01-556).

References

  1. 1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al. The global distribution and burden of dengue. Nature. 2013;496(7446):504-507
  2. 2. World Health Organization. Global Strategy for Dengue Prevention and Control 2012-2020. Geneva: World Health Organization; 2012
  3. 3. Guo C, Zhou Z, Wen Z, Liu Y, Zeng C, Xiao D, et al. Global epidemiology of dengue outbreaks in 1990-2015: A systematic review and meta-analysis. Frontiers in Cellular and Infection Microbiology. 2017;7:317
  4. 4. Halstead SB. Dengue. Lancet. 2007;370(9599):1644-1652
  5. 5. Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, et al. Antibody-dependent enhancement of severe dengue disease in humans. Science (New York, N.Y.). 2017;358(6365):929-932
  6. 6. Rivino L. Understanding the human T cell response to dengue virus. Advances in Experimental Medicine and Biology. 2018;1062:241-250
  7. 7. Mongkolsapaya J, Dejnirattisai W, Xu XN, Vasanawathana S, Tangthawornchaikul N, Chairunsri A, et al. Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nature Medicine. 2003;9(7):921-927
  8. 8. Rothman AL. Immunity to dengue virus: A tale of original antigenic sin and tropical cytokine storms. Nature Reviews. Immunology. 2011;11(8):532-543
  9. 9. Green S, Rothman A. Immunopathological mechanisms in dengue and dengue hemorrhagic fever. Current Opinion in Infectious Diseases. 2006;19(5):429-436
  10. 10. Kuczera D, Assolini JP, Tomiotto-Pellissier F, Pavanelli WR, Silveira GF. Highlights for dengue Immunopathogenesis: Antibody-dependent enhancement, cytokine storm, and beyond. Journal of Interferon & Cytokine Research. 2018;38(2):69-80
  11. 11. Scaturro P, Cortese M, Chatel-Chaix L, Fischl W, Bartenschlager R. Dengue virus non-structural protein 1 modulates infectious particle production via interaction with the structural proteins. PLOS Pathogens. 2015;11(11):e1005277
  12. 12. Muller DA, Young PR. The flavivirus NS1 protein: Molecular and structural biology, immunology, role in pathogenesis and application as a diagnostic biomarker. Antiviral Research. 2013;98(2):192-208
  13. 13. Avirutnan P, Fuchs A, Hauhart RE, Somnuke P, Youn S, Diamond MS, et al. Antagonism of the complement component C4 by flavivirus nonstructural protein NS1. The Journal of Experimental Medicine. 2010;207(4):793-806
  14. 14. Avirutnan P, Hauhart RE, Somnuke P, Blom AM, Diamond MS, Atkinson JP. Binding of flavivirus nonstructural protein NS1 to C4b binding protein modulates complement activation. Journal of Immunology. 2011;187(1):424-433
  15. 15. Avirutnan P, Punyadee N, Noisakran S, Komoltri C, Thiemmeca S, Auethavornanan K, et al. Vascular leakage in severe dengue virus infections: A potential role for the nonstructural viral protein NS1 and complement. The Journal of Infectious Diseases. 2006;193(8):1078-1088
  16. 16. Glasner DR, Puerta-Guardo H, Beatty PR, Harris E. The good, the bad, and the shocking: The multiple roles of dengue virus nonstructural protein 1 in protection and pathogenesis. Annual Review of Virology. 2018;5(1):227-253
  17. 17. Puerta-Guardo H, Tabata T, Petitt M, Dimitrova M, Glasner DR, Pereira L, et al. Zika virus nonstructural protein 1 disrupts glycosaminoglycans and causes permeability in developing human placentas. The Journal of Infectious Diseases. 2020;221(2):313-324
  18. 18. Glasner DR, Ratnasiri K, Puerta-Guardo H, Espinosa DA, Beatty PR, Harris E. Dengue virus NS1 cytokine-independent vascular leak is dependent on endothelial glycocalyx components. PLOS Pathogens. 2017;13(11):e1006673
  19. 19. Chen HR, Chao CH, Liu CC, Ho TS, Tsai HP, Perng GC, et al. Macrophage migration inhibitory factor is critical for dengue NS1-induced endothelial glycocalyx degradation and hyperpermeability. PLOS Pathogens. 2018;14(4):e1007033
  20. 20. Chen HR, Chuang YC, Lin YS, Liu HS, Liu CC, Perng GC, et al. Dengue virus nonstructural protein 1 induces vascular leakage through macrophage migration inhibitory factor and autophagy. PLOS Neglected Tropical Diseases. 2016;10(7):e0004828
  21. 21. Avirutnan P, Zhang L, Punyadee N, Manuyakorn A, Puttikhunt C, Kasinrerk W, et al. Secreted NS1 of dengue virus attaches to the surface of cells via interactions with heparan sulfate and chondroitin sulfate E. PLOS Pathogens. 2007;3(11):e183
  22. 22. Chao CH, Wu WC, Lai YC, Tsai PJ, Perng GC, Lin YS, et al. Dengue virus nonstructural protein 1 activates platelets via Toll-like receptor 4, leading to thrombocytopenia and hemorrhage. PLOS Pathogens. 2019;15(4):e1007625
  23. 23. Puerta-Guardo H, Glasner DR, Espinosa DA, Biering SB, Patana M, Ratnasiri K, et al. Flavivirus NS1 triggers tissue-specific vascular endothelial dysfunction reflecting disease tropism. Cell Reports. 2019;26(6):1598, 613.e8
  24. 24. Wang C, Puerta-Guardo H, Biering SB, Glasner DR, Tran EB, Patana M, et al. Endocytosis of flavivirus NS1 is required for NS1-mediated endothelial hyperpermeability and is abolished by a single N-glycosylation site mutation. PLOS Pathogens. 2019;15(7):e1007938
  25. 25. Xu X, Vaughan K, Weiskopf D, Grifoni A, Diamond MS, Sette A, et al. Identifying candidate targets of immune responses in Zika virus based on homology to epitopes in other Flavivirus species. PLOS Currents. 2016;8
  26. 26. Li A, Yu J, Lu M, Ma Y, Attia Z, Shan C, et al. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nature Communications. 2018;9(1):3067
  27. 27. Wan SW, Chen PW, Chen CY, Lai YC, Chu YT, Hung CY, et al. Therapeutic effects of monoclonal antibody against dengue virus NS1 in a STAT1 knockout mouse model of dengue infection. Journal of Immunology. 2017;199(8):2834-2844
  28. 28. Henchal EA, Henchal LS, Schlesinger JJ. Synergistic interactions of anti-NS1 monoclonal antibodies protect passively immunized mice from lethal challenge with dengue 2 virus. The Journal of General Virology. 1988;69(Pt 8):2101-2107
  29. 29. Edeling MA, Diamond MS, Fremont DH. Structural basis of flavivirus NS1 assembly and antibody recognition. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(11):4285-4290
  30. 30. Shriver-Lake LC, Liu JL, Zabetakis D, Sugiharto VA, Lee CR, Defang GN, et al. Selection and characterization of anti-dengue NS1 single domain antibodies. Scientific Reports. 2018;8(1):18086
  31. 31. Jayathilaka D, Gomes L, Jeewandara C, Jayarathna GSB, Herath D, Perera PA, et al. Role of NS1 antibodies in the pathogenesis of acute secondary dengue infection. Nature Communications. 2018;9(1):5242
  32. 32. Amorim JH, Diniz MO, Cariri FA, Rodrigues JF, Bizerra RS, Goncalves AJ, et al. Protective immunity to DENV2 after immunization with a recombinant NS1 protein using a genetically detoxified heat-labile toxin as an adjuvant. Vaccine. 2012;30(5):837-845
  33. 33. Wan SW, Lu YT, Huang CH, Lin CF, Anderson R, Liu HS, et al. Protection against dengue virus infection in mice by administration of antibodies against modified nonstructural protein 1. PLOS One. 2014;9(3):e92495
  34. 34. Lin YL, Chen LK, Liao CL, Yeh CT, Ma SH, Chen JL, et al. DNA immunization with Japanese encephalitis virus nonstructural protein NS1 elicits protective immunity in mice. Journal of Virology. 1998;72(1):191-200
  35. 35. Beatty PR, Puerta-Guardo H, Killingbeck SS, Glasner DR, Hopkins K, Harris E. Dengue virus NS1 triggers endothelial permeability and vascular leak that is prevented by NS1 vaccination. Science Translational Medicine. 2015;7(304):304ra141
  36. 36. Falconar AK. The dengue virus nonstructural-1 protein (NS1) generates antibodies to common epitopes on human blood clotting, integrin/adhesin proteins and binds to human endothelial cells: Potential implications in haemorrhagic fever pathogenesis. Archives of Virology. 1997;142(5):897-916
  37. 37. Chen MC, Lin CF, Lei HY, Lin SC, Liu HS, Yeh TM, et al. Deletion of the C-terminal region of dengue virus nonstructural protein 1 (NS1) abolishes anti-NS1-mediated platelet dysfunction and bleeding tendency. Journal of Immunology. 2009;183(3):1797-1803
  38. 38. Espinosa DA, Beatty PR, Reiner GL, Sivick KE, Hix Glickman L, Dubensky TW Jr, et al. Cyclic dinucleotide-adjuvanted dengue virus nonstructural protein 1 induces protective antibody and T cell responses. Journal of Immunology. 2019;202(4):1153-1162
  39. 39. Lin CF, Lei HY, Shiau AL, Liu CC, Liu HS, Yeh TM, et al. Antibodies from dengue patient sera cross-react with endothelial cells and induce damage. Journal of Medical Virology. 2003;69(1):82-90
  40. 40. Bailey MJ, Broecker F, Duehr J, Arumemi F, Krammer F, Palese P, et al. Antibodies elicited by an NS1-based vaccine protect mice against Zika virus. mBio. 2019;10(2):e02861-18
  41. 41. Lai YC, Chuang YC, Liu CC, Ho TS, Lin YS, Anderson R, et al. Antibodies against modified NS1 wing domain peptide protect against dengue virus infection. Scientific Reports. 2017;7(1):6975
  42. 42. Reyes-Sandoval A, Ludert JE. The dual role of the antibody response against the flavivirus non-structural protein 1 (NS1) in protection and immuno-pathogenesis. Frontiers in Immunology. 2019;10:1651
  43. 43. Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, Lenches E, et al. Structure of dengue virus: Implications for flavivirus organization, maturation, and fusion. Cell. 2002;108(5):717-725
  44. 44. Katzelnick LC, Fonville JM, Gromowski GD, Bustos Arriaga J, Green A, James SL, et al. Dengue viruses cluster antigenically but not as discrete serotypes. Science. 2015;349(6254):1338-1343
  45. 45. Weaver SC, Vasilakis N. Molecular evolution of dengue viruses: Contributions of phylogenetics to understanding the history and epidemiology of the preeminent arboviral disease. Infection, Genetics and Evolution, Journal of Molecular Epidemiology and Evolutionary Genetics of Infectious Diseases. 2009;9(4):523-540
  46. 46. Holmes EC, Twiddy SS. The origin, emergence and evolutionary genetics of dengue virus. Infection, Genetics and Evolution. 2003;3(1):19-28
  47. 47. Khan AM, Heiny AT, Lee KX, Srinivasan KN, Tan TW, August JT, et al. Large-scale analysis of antigenic diversity of T-cell epitopes in dengue virus. BMC Bioinformatics. 2006;7(Suppl 5):S4
  48. 48. Lwande OW, Obanda V, Lindström A, Ahlm C, Evander M, Näslund J, et al. Globe-trotting Aedes aegypti and Aedes albopictus: Risk factors for arbovirus pandemics. Vector Borne and Zoonotic Diseases. 2020;20(2):71-81
  49. 49. Gubler DJ. Dengue, urbanization and globalization: The unholy trinity of the 21(st) century. Tropical Medicine and Health. 2011;39(4 Suppl):3-11
  50. 50. Paupy C, Delatte H, Bagny L, Corbel V, Fontenille D. Aedes albopictus, an arbovirus vector: From the darkness to the light. Microbes and Infection. 2009;11(14-15):1177-1185
  51. 51. Carrington LB, Simmons CP. Human to mosquito transmission of dengue viruses. Frontiers in Immunology. 2014;5:290
  52. 52. Chan M, Johansson MA. The incubation periods of dengue viruses. PLOS One. 2012;7(11):e50972
  53. 53. Kuno G, Chang GJ. Biological transmission of arboviruses: Reexamination of and new insights into components, mechanisms, and unique traits as well as their evolutionary trends. Clinical Microbiology Reviews. 2005;18(4):608-637
  54. 54. Rathore APS, St John AL. Immune responses to dengue virus in the skin. Open Biology. 2018;8(8):180087
  55. 55. Limon-Flores AY, Perez-Tapia M, Estrada-Garcia I, Vaughan G, Escobar-Gutierrez A, Calderon-Amador J, et al. Dengue virus inoculation to human skin explants: An effective approach to assess in situ the early infection and the effects on cutaneous dendritic cells. International Journal of Experimental Pathology. 2005;86(5):323-334
  56. 56. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, et al. Human skin Langerhans cells are targets of dengue virus infection. Nature Medicine. 2000;6(7):816-820
  57. 57. Schmid MA, Diamond MS, Harris E. Dendritic cells in dengue virus infection: Targets of virus replication and mediators of immunity. Frontiers in Immunology. 2014;5:647
  58. 58. Wichit S, Ferraris P, Choumet V, Misse D. The effects of mosquito saliva on dengue virus infectivity in humans. Current Opinion in Virology. 2016;21:139-145
  59. 59. Conway MJ, Watson AM, Colpitts TM, Dragovic SM, Li Z, Wang P, et al. Mosquito saliva serine protease enhances dissemination of dengue virus into the mammalian host. Journal of Virology. 2014;88(1):164
  60. 60. Schmid MA, Glasner DR, Shah S, Michlmayr D, Kramer LD, Harris E. Mosquito saliva increases endothelial permeability in the skin, immune cell migration, and dengue pathogenesis during antibody-dependent enhancement. PLOS Pathogens. 2016;12(6):e1005676-e
  61. 61. Cox J, Mota J, Sukupolvi-Petty S, Diamond MS, Rico-Hesse R. Mosquito bite delivery of dengue virus enhances immunogenicity and pathogenesis in humanized mice. Journal of Virology. 2012;86(14):7637-7649
  62. 62. Pingen M, Schmid MA, Harris E, McKimmie CS. Mosquito biting modulates skin response to virus infection. Trends in Parasitology. 2017;33(8):645-657
  63. 63. Reyes-del Valle J, Salas-Benito J, Soto-Acosta R, del Angel RM. Dengue virus cellular receptors and tropism. Current Tropical Medicine Reports. 2014;1(1):36-43
  64. 64. Perera-Lecoin M, Meertens L, Carnec X, Amara A. Flavivirus entry receptors: An update. Viruses. 2013;6(1):69-88
  65. 65. Cruz-Oliveira C, Freire JM, Conceicao TM, Higa LM, Castanho MA, Da Poian AT. Receptors and routes of dengue virus entry into the host cells. FEMS Microbiology Reviews. 2015;39(2):155-170
  66. 66. Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L. Flavivirus receptors: Diversity, identity, and cell entry. Frontiers in Immunology. 2018;9:2180
  67. 67. Begum F, Das S, Mukherjee D, Mal S, Ray U. Insight into the tropism of dengue virus in humans. Viruses. 2019;11(12):1136
  68. 68. 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(23):11418-11431
  69. 69. Stiasny K, Fritz R, Pangerl K, Heinz FX. Molecular mechanisms of flavivirus membrane fusion. Amino Acids. 2011;41(5):1159-1163
  70. 70. Heinz FX, Stiasny K. Flaviviruses and their antigenic structure. Journal of Clinical Virology. 2012;55(4):289-295
  71. 71. Welsch S, Miller S, Romero-Brey I, Merz A, Bleck CK, Walther P, et al. Composition and three-dimensional architecture of the dengue virus replication and assembly sites. Cell Host & Microbe. 2009;5(4):365-375
  72. 72. Lescar J, Soh S, Lee LT, Vasudevan SG, Kang C, Lim SP. 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
  73. 73. Villordo SM, Carballeda JM, Filomatori CV, Gamarnik AV. RNA structure duplications and flavivirus host adaptation. Trends in Microbiology. 2016;24(4):270-283
  74. 74. Villordo SM, Filomatori CV, Sanchez-Vargas I, Blair CD, Gamarnik AV. Dengue virus RNA structure specialization facilitates host adaptation. PLOS Pathogens. 2015;11(1):e1004604
  75. 75. El Sahili A, Lescar J. Dengue virus non-structural protein 5. Viruses. 2017;9(4):91
  76. 76. Gebhard LG, Incicco JJ, Smal C, Gallo M, Gamarnik AV, Kaufman SB. Monomeric nature of dengue virus NS3 helicase and thermodynamic analysis of the interaction with single-stranded RNA. Nucleic Acids Research. 2014;42(18):11668-11686
  77. 77. Liu L, Dong H, Chen H, Zhang J, Ling H, Li Z, et al. Flavivirus RNA cap methyltransferase: Structure, function, and inhibition. Frontiers in Biology (Beijing). 2010;5(4):286-303
  78. 78. Xie X, Zou J, Zhang X, Zhou Y, Routh AL, Kang C, et al. Dengue NS2A protein orchestrates virus assembly. Cell Host & Microbe. 2019;26(5):606-22.e8
  79. 79. Plaszczyca A, Scaturro P, Neufeldt CJ, Cortese M, Cerikan B, Ferla S, et al. A novel interaction between dengue virus nonstructural protein 1 and the NS4A-2K-4B precursor is required for viral RNA replication but not for formation of the membranous replication organelle. PLOS Pathogens. 2019;15(5):e1007736
  80. 80. Norazharuddin H, Lai NS. Roles and prospects of dengue virus non-structural proteins as antiviral targets: An easy digest. Malaysian Journal of Medical Sciences. 2018;25(5):6-15
  81. 81. Xie Q , Zhang B, Yu J, Wu Q , Yang F, Cao H, et al. Structure and function of the non-structural protein of dengue virus and its applications in antiviral therapy. Current Topics in Medicinal Chemistry. 2017;17(3):371-380
  82. 82. Wang Q-Y, Dong H, Zou B, Karuna R, Wan KF, Zou J, et al. Discovery of dengue virus NS4B inhibitors. Journal of Virology. 2015;89(16):8233
  83. 83. Wu H, Bock S, Snitko M, Berger T, Weidner T, Holloway S, et al. Novel dengue virus NS2B/NS3 protease inhibitors. Antimicrobial Agents and Chemotherapy. 2015;59(2):1100
  84. 84. Apte-Sengupta S, Sirohi D, Kuhn RJ. Coupling of replication and assembly in flaviviruses. Current Opinion in Virology. 2014;9:134-142
  85. 85. Brady OJ, Gething PW, Bhatt S, Messina JP, Brownstein JS, Hoen AG, et al. Refining the global spatial limits of dengue virus transmission by evidence-based consensus. PLOS Neglected Tropical Diseases. 2012;6(8):e1760
  86. 86. Messina JP, Brady OJ, Scott TW, Zou C, Pigott DM, Duda KA, et al. Global spread of dengue virus types: Mapping the 70 year history. Trends in Microbiology. 2014;22(3):138-146
  87. 87. Rezza G. Aedes albopictus and the reemergence of dengue. BMC Public Health. 2012;12:72
  88. 88. Gubler DJ. Dengue, urbanization and globalization: The unholy trinity of the 21(st) century. Tropical Medicine and Health. 2011;39(4 Suppl):3-11
  89. 89. World Health Organization. Dengue Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. Geneva: World Health Organization; 2009
  90. 90. World Health Organization. Dengue Haemorrhagic Fever: Diagnosis, Treatment, Prevention and Control. 2nd ed. Geneva: World Health Organization; 1997
  91. 91. Hung NT. Fluid management for dengue in children. Paediatrics and International Child Health. 2012;32(s1):39-42
  92. 92. Hladish TJ, Pearson CAB, Toh KB, Rojas DP, Manrique-Saide P, Vazquez-Prokopec GM, et al. Designing effective control of dengue with combined interventions. Proceedings of the National Academy of Sciences of the United States of America. 2020;117(6):3319-3325
  93. 93. Trung DT, Wills B. Systemic vascular leakage associated with dengue infections—The clinical perspective. Current Topics in Microbiology and Immunology. 2010;338:57-66
  94. 94. Yacoub S, Wertheim H, Simmons CP, Screaton G, Wills B. Cardiovascular manifestations of the emerging dengue pandemic. Nature Reviews. Cardiology. 2014;11(6):335-345
  95. 95. Yacoub S, Lam PK, Vu le HM, Le TL, Ha NT, Toan TT, et al. Association of microvascular function and endothelial biomarkers with clinical outcome in dengue: An observational study. The Journal of Infectious Diseases 2016;214(5):697-706
  96. 96. Srikiatkhachorn A. Plasma leakage in dengue haemorrhagic fever. Thrombosis and Haemostasis. 2009;102(6):1042-1049
  97. 97. Halstead SB. Controversies in dengue pathogenesis. Paediatrics and International Child Health. 2012;32(Suppl 1):5-9
  98. 98. Simmons CP, Farrar JJ, Nguyen VV, Wills B. Dengue. The New England Journal of Medicine. 2012;366(15):1423-1432
  99. 99. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, et al. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. The Journal of Infectious Diseases. 2000;181(1):2-9
  100. 100. OhAinle M, Balmaseda A, Macalalad AR, Tellez Y, Zody MC, Saborío S, et al. Dynamics of dengue disease severity determined by the interplay between viral genetics and serotype-specific immunity. Science Translational Medicine. 2011;3(114):114ra28-114ra28
  101. 101. Malavige GN, Ogg GS. T cell responses in dengue viral infections. Journal of Clinical Virology. 2013;58(4):605-611
  102. 102. Srikiatkhachorn A, Mathew A, Rothman AL. Immune-mediated cytokine storm and its role in severe dengue. Seminars in Immunopathology. 2017;39(5):563-574
  103. 103. Libraty DH, Young PR, Pickering D, Endy TP, Kalayanarooj S, Green S, et al. High circulating levels of the dengue virus nonstructural protein NS1 early in dengue illness correlate with the development of dengue hemorrhagic fever. The Journal of Infectious Diseases. 2002;186(8):1165-1168
  104. 104. Rothman AL. Dengue: Defining protective versus pathologic immunity. The Journal of Clinical Investigation. 2004;113(7):946-951
  105. 105. Mangada MM, Rothman AL. Altered cytokine responses of dengue-specific CD4+ T cells to heterologous serotypes. Journal of Immunology. 2005;175(4):2676-2683
  106. 106. Guzman MG, Alvarez M, Halstead SB. Secondary infection as a risk factor for dengue hemorrhagic fever/dengue shock syndrome: An historical perspective and role of antibody-dependent enhancement of infection. Archives of Virology. 2013;158(7):1445-1459
  107. 107. Sangkawibha N, Rojanasuphot S, Ahandrik S, Viriyapongse S, Jatanasen S, Salitul V, et al. Risk factors in dengue shock syndrome: A prospective epidemiologic study in Rayong, Thailand. I. The 1980 outbreak. American Journal of Epidemiology. 1984;120(5):653-669
  108. 108. Conde JN, Silva EM, Barbosa AS, Mohana-Borges R. The complement system in flavivirus infections. Frontiers in Microbiology. 2017;8:213
  109. 109. Malasit P. Complement and dengue haemorrhagic fever/shock syndrome. The Southeast Asian Journal of Tropical Medicine and Public Health. 1987;18(3):316-320
  110. 110. Rico-Hesse R. Dengue virus markers of virulence and pathogenicity. Future Virology. 2009;4(6):581
  111. 111. Diamond MS, Edgil D, Roberts TG, Lu B, Harris E. Infection of human cells by dengue virus is modulated by different cell types and viral strains. Journal of Virology. 2000;74(17):7814
  112. 112. Begum F, Das S, Mukherjee D, Ray U. Hijacking the host immune cells by dengue virus: Molecular interplay of receptors and dengue virus envelope. Microorganisms. 2019;7(9):323
  113. 113. Phoolcharoen W, Smith DR. Internalization of the dengue virus is cell cycle modulated in HepG2, but not vero cells. Journal of Medical Virology. 2004;74(3):434-441
  114. 114. Soto-Acosta R, Mosso C, Cervantes-Salazar M, Puerta-Guardo H, Medina F, Favari L, 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(2):132-147
  115. 115. King CA, Anderson R, Marshall JS. Dengue virus selectively induces human mast cell chemokine production. Journal of Virology. 2002;76(16):8408-8419
  116. 116. King AD, Nisalak A, Kalayanrooj S, Myint KS, Pattanapanyasat K, Nimmannitya S, et al. B cells are the principal circulating mononuclear cells infected by dengue virus. The Southeast Asian Journal of Tropical Medicine and Public Health. 1999;30(4):718-728
  117. 117. Salgado DM, Eltit JM, Mansfield K, Panqueba C, Castro D, Vega MR, et al. Heart and skeletal muscle are targets of dengue virus infection. The Pediatric Infectious Disease Journal. 2010;29(3):238-242
  118. 118. Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. The Journal of Infectious Diseases. 2004;189(8):1411-1418
  119. 119. Win MM, Charngkaew K, Punyadee N, Aye KS, Win N, Chaisri U, et al. Ultrastructural features of human liver specimens from patients who died of dengue hemorrhagic fever. Tropical Medicine and Infectious Disease. 2019;4(2):63
  120. 120. Aye KS, Charngkaew K, Win N, Wai KZ, Moe K, Punyadee N, et al. Pathologic highlights of dengue hemorrhagic fever in 13 autopsy cases from Myanmar. Human Pathology. 2014;45(6):1221-1233
  121. 121. Kangwanpong D, Bhamarapravati N, Lucia HL. Diagnosing dengue virus infection in archived autopsy tissues by means of the in situ PCR method: A case report. Clinical and Diagnostic Virology. 1995;3(2):165-172
  122. 122. Balsitis SJ, Coloma J, Castro G, Alava A, Flores D, McKerrow JH, et al. Tropism of dengue virus in mice and humans defined by viral nonstructural protein 3-specific immunostaining. The American Journal of Tropical Medicine and Hygiene. 2009;80(3):416-424
  123. 123. Balsitis SJ, Williams KL, Lachica R, Flores D, Kyle JL, Mehlhop E, et al. Lethal antibody enhancement of dengue disease in mice is prevented by Fc modification. PLOS Pathogens. 2010;6(2):e1000790
  124. 124. Shresta S, Kyle JL, Snider HM, Basavapatna M, Beatty PR, Harris E. Interferon-dependent immunity is essential for resistance to primary dengue virus infection in mice, whereas T- and B-cell-dependent immunity are less critical. Journal of Virology. 2004;78(6):2701-2710
  125. 125. Zellweger RM, Prestwood TR, Shresta S. Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe dengue disease. Cell Host & Microbe. 2010;7(2):128-139
  126. 126. Orozco S, Schmid MA, Parameswaran P, Lachica R, Henn MR, Beatty R, et al. Characterization of a model of lethal dengue virus 2 infection in C57BL/6 mice deficient in the alpha/beta interferon receptor. The Journal of General Virology. 2012;93(Pt 10):2152-2157
  127. 127. Shresta S, Sharar KL, Prigozhin DM, Beatty PR, Harris E. Murine model for dengue virus-induced lethal disease with increased vascular permeability. Journal of Virology. 2006;80(20):10208-10217
  128. 128. Zellweger RM, Shresta S. Mouse models to study dengue virus immunology and pathogenesis. Frontiers in Immunology. 2014;5:151
  129. 129. Tassaneetrithep B, Burgess TH, Granelli-Piperno A, Trumpfheller C, Finke J, Sun W, et al. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. The Journal of Experimental Medicine. 2003;197(7):823-829
  130. 130. Navarro-Sanchez E, Altmeyer R, Amara A, Schwartz O, Fieschi F, Virelizier JL, et al. Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Reports. 2003;4(7):723-728
  131. 131. Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, Lei HY, et al. CLEC5A is critical for dengue-virus-induced lethal disease. Nature. 2008;453(7195):672-676
  132. 132. Miller JL, de Wet BJ, Martinez-Pomares L, Radcliffe CM, Dwek RA, Rudd PM, et al. The mannose receptor mediates dengue virus infection of macrophages. PLOS Pathogens. 2008;4(2):e17
  133. 133. Boonnak K, Dambach KM, Donofrio GC, Tassaneetrithep B, Marovich MA. Cell type specificity and host genetic polymorphisms influence antibody-dependent enhancement of dengue virus infection. Journal of Virology. 2011;85(4):1671-1683
  134. 134. Wu MF, Chen ST, Yang AH, Lin WW, Lin YL, Chen NJ, et al. CLEC5A is critical for dengue virus-induced inflammasome activation in human macrophages. Blood. 2013;121(1):95-106
  135. 135. Luplertlop N, Misse D, Bray D, Deleuze V, Gonzalez JP, Leardkamolkarn V, et al. Dengue-virus-infected dendritic cells trigger vascular leakage through metalloproteinase overproduction. EMBO Reports. 2006;7(11):1176-1181
  136. 136. Avirutnan P, Matangkasombut P. Unmasking the role of mast cells in dengue. eLife. 2013;2:e00767
  137. 137. Sherif NA, Zayan AH, Elkady AH, Ghozy S, Ahmed AR, Omran ES, et al. Mast cell mediators in relation to dengue severity: A systematic review and meta-analysis. Reviews in Medical Virology. 2020;30(1):e2084
  138. 138. Rathore APS, St John AL. Immune responses to dengue virus in the skin. Open Biology. 2018;8(8):180087
  139. 139. Brown MG, Hermann LL, Issekutz AC, Marshall JS, Rowter D, Al-Afif A, et al. Dengue virus infection of mast cells triggers endothelial cell activation. Journal of Virology. 2011;85(2):1145
  140. 140. St John AL, Rathore AP, Raghavan B, Ng ML, Abraham SN. Contributions of mast cells and vasoactive products, leukotrienes and chymase, to dengue virus-induced vascular leakage. eLife. 2013;2:e00481
  141. 141. Zimmer CL, Cornillet M, Sola-Riera C, Cheung KW, Ivarsson MA, Lim MQ , et al. NK cells are activated and primed for skin-homing during acute dengue virus infection in humans. Nature Communications. 2019;10(1):3897
  142. 142. Durbin AP, Vargas MJ, Wanionek K, Hammond SN, Gordon A, Rocha C, et al. Phenotyping of peripheral blood mononuclear cells during acute dengue illness demonstrates infection and increased activation of monocytes in severe cases compared to classic dengue fever. Virology. 2008;376(2):429-435
  143. 143. Castillo JA, Naranjo JS, Rojas M, Castano D, Velilla PA. Role of monocytes in the pathogenesis of dengue. Archivum Immunologiae et Therapiae Experimentalis (Warsz). 2019;67(1):27-40
  144. 144. Schmid MA, Harris E. Monocyte recruitment to the dermis and differentiation to dendritic cells increases the targets for dengue virus replication. PLOS Pathogens. 2014;10(12):e1004541
  145. 145. Kyle JL, Beatty PR, Harris E. Dengue virus infects macrophages and dendritic cells in a mouse model of infection. The Journal of Infectious Diseases. 2007;195(12):1808-1817
  146. 146. Marianneau P, Steffan AM, Royer C, Drouet MT, Jaeck D, Kirn A, et al. Infection of primary cultures of human Kupffer cells by dengue virus: No viral progeny synthesis, but cytokine production is evident. Journal of Virology. 1999;73(6):5201-5206
  147. 147. Povoa TF, Alves AM, Oliveira CA, Nuovo GJ, Chagas VL, Paes MV. The pathology of severe dengue in multiple organs of human fatal cases: Histopathology, ultrastructure and virus replication. PLOS One. 2014;9(4):e83386
  148. 148. Moi ML, Lim CK, Takasaki T, Kurane I. Involvement of the Fc gamma receptor IIA cytoplasmic domain in antibody-dependent enhancement of dengue virus infection. The Journal of General Virology. 2010;91(Pt 1):103-111
  149. 149. Littaua R, Kurane I, Ennis FA. Human IgG Fc receptor II mediates antibody-dependent enhancement of dengue virus infection. Journal of Immunology. 1990;144(8):3183-3186
  150. 150. Kontny U, Kurane I, Ennis FA. Gamma interferon augments Fc gamma receptor-mediated dengue virus infection of human monocytic cells. Journal of Virology. 1988;62(11):3928-3933
  151. 151. Rodrigo WW, Jin X, Blackley SD, Rose RC, Schlesinger JJ. Differential enhancement of dengue virus immune complex infectivity mediated by signaling-competent and signaling-incompetent human Fcgamma RIA (CD64) or FcgammaRIIA (CD32). Journal of Virology. 2006;80(20):10128-10138
  152. 152. Homchampa P, Sarasombath S, Suvatte V, Vongskul M. Natural killer cells in dengue hemorrhagic fever/dengue shock syndrome. Asian Pacific Journal of Allergy and Immunology. 1988;6(2):95-102
  153. 153. Azeredo EL, De Oliveira-Pinto LM, Zagne SM, Cerqueira DI, Nogueira RM, Kubelka CF. NK cells, displaying early activation, cytotoxicity and adhesion molecules, are associated with mild dengue disease. Clinical and Experimental Immunology. 2006;143(2):345-356
  154. 154. Kou Z, Quinn M, Chen H, Rodrigo WW, Rose RC, Schlesinger JJ, et al. Monocytes, but not T or B cells, are the principal target cells for dengue virus (DV) infection among human peripheral blood mononuclear cells. Journal of Medical Virology. 2008;80(1):134-146
  155. 155. Lin YW, Wang KJ, Lei HY, Lin YS, Yeh TM, Liu HS, et al. Virus replication and cytokine production in dengue virus-infected human B lymphocytes. Journal of Virology. 2002;76(23):12242-12249
  156. 156. Silveira GF, Wowk PF, Cataneo AHD, Dos Santos PF, Delgobo M, Stimamiglio MA, et al. Human T lymphocytes are permissive for dengue virus replication. Journal of Virology. 2018;92(10):e02181-17
  157. 157. Kurane I, Kontny U, Janus J, Ennis FA. Dengue-2 virus infection of human mononuclear cell lines and establishment of persistent infections. Archives of Virology. 1990;110(1-2):91-101
  158. 158. Boonpucknavig S, Bhamarapravati N, Nimmannitya S, Phalavadhtana A, Siripont J. Immunofluorescent staining of the surfaces of lymphocytes in suspension from patients with dengue hemorrhagic fever. The American Journal of Pathology. 1976;85(1):37-48
  159. 159. Mentor NA, Kurane I. Dengue virus infection of human T lymphocytes. Acta Virologica. 1997;41(3):175-176
  160. 160. Mota J, Rico-Hesse R. Dengue virus tropism in humanized mice recapitulates human dengue fever. PLOS One. 2011;6(6):e20762-e
  161. 161. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805-820
  162. 162. Kawamura T, Ogawa Y, Aoki R, Shimada S. Innate and intrinsic antiviral immunity in skin. Journal of Dermatological Science. 2014;75(3):159-166
  163. 163. Thompson MR, Kaminski JJ, Kurt-Jones EA, Fitzgerald KA. Pattern recognition receptors and the innate immune response to viral infection. Viruses. 2011;3(6):920-940
  164. 164. Morrison J, Aguirre S, Fernandez-Sesma A. Innate immunity evasion by dengue virus. Viruses. 2012;4(3):397-413
  165. 165. Uno N, Ross TM. Dengue virus and the host innate immune response. Emerging Microbes & Infections. 2018;7(1):167
  166. 166. Nasirudeen AM, Wong HH, Thien P, Xu S, Lam KP, Liu DX. RIG-I, MDA5 and TLR3 synergistically play an important role in restriction of dengue virus infection. PLOS Neglected Tropical Diseases. 2011;5(1):e926
  167. 167. Green AM, Beatty PR, Hadjilaou A, Harris E. Innate immunity to dengue virus infection and subversion of antiviral responses. Journal of Molecular Biology. 2014;426(6):1148-1160
  168. 168. Tsai YT, Chang SY, Lee CN, Kao CL. Human TLR3 recognizes dengue virus and modulates viral replication in vitro. Cellular Microbiology. 2009;11(4):604-615
  169. 169. Sariol CA, Martínez MI, Rivera F, Rodríguez IV, Pantoja P, Abel K, et al. Decreased dengue replication and an increased anti-viral Humoral response with the use of combined toll-like receptor 3 and 7/8 agonists in macaques. PLOS One. 2011;6(4):e19323
  170. 170. Sun B, Sundström KB, Chew JJ, Bist P, Gan ES, Tan HC, et al. Dengue virus activates cGAS through the release of mitochondrial DNA. Scientific Reports. 2017;7(1):3594
  171. 171. Aguirre S, Fernandez-Sesma A. Collateral damage during dengue virus infection: Making sense of DNA by cGAS. Journal of Virology. 2017;91(14):e01081-e01016
  172. 172. Cai X, Chiu YH, Chen ZJ. The cGAS-cGAMP-STING pathway of cytosolic DNA sensing and signaling. Molecular Cell. 2014;54(2):289-296
  173. 173. Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nature Reviews. Molecular Cell Biology. 2019;20(1):21-37
  174. 174. Stoermer KA, Morrison TE. Complement and viral pathogenesis. Virology. 2011;411(2):362-373
  175. 175. O’Brien J, Hayder H, Zayed Y, Peng C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Frontiers in Endocrinology (Lausanne). 2018;9:402
  176. 176. Ding SW, Voinnet O. Antiviral immunity directed by small RNAs. Cell. 2007;130(3):413-426
  177. 177. Wong RR, Abd-Aziz N, Affendi S, Poh CL. Role of microRNAs in antiviral responses to dengue infection. Journal of Biomedical Science. 2020;27(1):4
  178. 178. Ang F, Wong AP, Ng MM, Chu JJ. Small interference RNA profiling reveals the essential role of human membrane trafficking genes in mediating the infectious entry of dengue virus. Virology Journal. 2010;7:24
  179. 179. Kakumani PK, Ponia SS, Rajgokul SK, Sood V, Chinnappan M, Banerjea AC, et al. Role of RNA interference (RNAi) in dengue virus replication and identification of NS4B as an RNAi suppressor. Journal of Virology. 2013;87(16):8870-8883
  180. 180. Mazeaud C, Freppel W, Chatel-Chaix L. The multiples fates of the Flavivirus RNA genome during pathogenesis. Frontiers in Genetics. 2018;9:595
  181. 181. Pijlman GP, Funk A, Kondratieva N, Leung J, Torres S, van der Aa L, et al. A highly structured, nuclease-resistant, noncoding RNA produced by flaviviruses is required for pathogenicity. Cell Host & Microbe. 2008;4(6):579-591
  182. 182. Schnettler E, Sterken MG, Leung JY, Metz SW, Geertsema C, Goldbach RW, et al. Noncoding flavivirus RNA displays RNA interference suppressor activity in insect and mammalian cells. Journal of Virology. 2012;86(24):13486-13500
  183. 183. Moon SL, Anderson JR, Kumagai Y, Wilusz CJ, Akira S, Khromykh AA, et al. A noncoding RNA produced by arthropod-borne flaviviruses inhibits the cellular exoribonuclease XRN1 and alters host mRNA stability. RNA. 2012;18(11):2029-2040
  184. 184. Urcuqui-Inchima S, Cabrera J, Haenni AL. Interplay between dengue virus and toll-like receptors, RIG-I/MDA5 and microRNAs: Implications for pathogenesis. Antiviral Research. 2017;147:47-57
  185. 185. Jiang L, Sun Q . The expression profile of human peripheral blood mononuclear cell miRNA is altered by antibody-dependent enhancement of infection with dengue virus serotype 3. Virology Journal. 2018;15(1):50
  186. 186. Zhu X, He Z, Hu Y, Wen W, Lin C, Yu J, et al. MicroRNA-30e* suppresses dengue virus replication by promoting NF-κB–dependent IFN production. PLOS Neglected Tropical Diseases. 2014;8(8):e3088
  187. 187. Castillo JA, Castrillón JC, Diosa-Toro M, Betancur JG, St Laurent G 3rd, Smit JM, et al. Complex interaction between dengue virus replication and expression of miRNA-133a. BMC Infectious Diseases. 2016;16:29
  188. 188. Diosa-Toro M, Echavarria-Consuegra L, Flipse J, Fernandez GJ, Kluiver J, van den Berg A, et al. MicroRNA profiling of human primary macrophages exposed to dengue virus identifies miRNA-3614-5p as antiviral and regulator of ADAR1 expression. PLOS Neglected Tropical Diseases. 2017;11(10):e0005981
  189. 189. Ouyang X, Jiang X, Gu D, Zhang Y, Kong SK, Jiang C, et al. Dysregulated serum MiRNA profile and promising biomarkers in dengue-infected patients. International Journal of Medical Sciences. 2016;13(3):195-205
  190. 190. Shahen M, Guo Z, Shar AH, Ebaid R, Tao Q , Zhang W, et al. Dengue virus causes changes of microRNA-genes regulatory network revealing potential targets for antiviral drugs. BMC Systems Biology. 2018;12(1):2
  191. 191. Feng X, Zhou S, Wang J, Hu W. microRNA profiles and functions in mosquitoes. PLOS Neglected Tropical Diseases. 2018;12(5):e0006463-e
  192. 192. Su J, Wang G, Li C, Xing D, Yan T, Zhu X, et al. Screening for differentially expressed miRNAs in Aedes albopictus (Diptera: Culicidae) exposed to DENV-2 and their effect on replication of DENV-2 in C6/36 cells. Parasites & Vectors. 2019;12(1):44
  193. 193. Su J, Li C, Zhang Y, Yan T, Zhu X, Zhao M, et al. Identification of microRNAs expressed in the midgut of Aedes albopictus during dengue infection. Parasites & Vectors. 2017;10(1):63
  194. 194. Lee W-S, Webster JA, Madzokere ET, Stephenson EB, Herrero LJ. Mosquito antiviral defense mechanisms: A delicate balance between innate immunity and persistent viral infection. Parasites & Vectors. 2019;12(1):165
  195. 195. Blair CD. Mosquito RNAi is the major innate immune pathway controlling arbovirus infection and transmission. Future Microbiology. 2011;6(3):265-277
  196. 196. Shahid I. RNA association, RNA interference, and microRNA pathways in dengue fever virus-host interaction. Current Topics in Tropical Emerging Diseases and Travel Medicine. 2018;6:93-114
  197. 197. Shresta S. Role of complement in dengue virus infection: Protection or pathogenesis? mBio. 2012;3(1):e00003-12
  198. 198. Cabezas S, Bracho G, Aloia AL, Adamson PJ, Bonder CS, Smith JR, et al. Dengue virus induces increased activity of the complement alternative pathway in infected cells. Journal of Virology. 2018;92(14):e00633-e00618
  199. 199. Carr JM, Cabezas-Falcon S, Dubowsky JG, Hulme-Jones J, Gordon DL. Dengue virus and the complement alternative pathway. FEBS Letters. 2020;594:2543-2555
  200. 200. Bokisch VA, Top FH Jr, Russell PK, Dixon FJ, Muller-Eberhard HJ. The potential pathogenic role of complement in dengue hemorrhagic shock syndrome. The New England Journal of Medicine. 1973;289(19):996-1000
  201. 201. Koyama S, Ishii KJ, Coban C, Akira S. Innate immune response to viral infection. Cytokine. 2008;43(3):336-341
  202. 202. Takeuchi O, Akira S. Innate immunity to virus infection. Immunological Reviews. 2009;227(1):75-86
  203. 203. Munoz-Jordan JL, Sanchez-Burgos GG, Laurent-Rolle M, Garcia-Sastre A. Inhibition of interferon signaling by dengue virus. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(24):14333-14338
  204. 204. Diamond MS, Roberts TG, Edgil D, Lu B, Ernst J, Harris E. Modulation of dengue virus infection in human cells by alpha, beta, and gamma interferons. Journal of Virology. 2000;74(11):4957-4966
  205. 205. Perry AK, Chen G, Zheng D, Tang H, Cheng G. The host type I interferon response to viral and bacterial infections. Cell Research. 2005;15(6):407-422
  206. 206. Baum A, García-Sastre A. Induction of type I interferon by RNA viruses: Cellular receptors and their substrates. Amino Acids. 2010;38(5):1283-1299
  207. 207. Schoggins JW, Rice CM. Interferon-stimulated genes and their antiviral effector functions. Current Opinion in Virology. 2011;1(6):519-525
  208. 208. Huang X, Yue Y, Li D, Zhao Y, Qiu L, Chen J, et al. Antibody-dependent enhancement of dengue virus infection inhibits RLR-mediated type-I IFN-independent signalling through upregulation of cellular autophagy. Scientific Reports. 2016;6:22303
  209. 209. Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472(7344):481-485
  210. 210. Liu SY, Sanchez DJ, Aliyari R, Lu S, Cheng G. Systematic identification of type I and type II interferon-induced antiviral factors. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(11):4239-4244
  211. 211. Pulit-Penaloza JA, Scherbik SV, Brinton MA. Type 1 IFN-independent activation of a subset of interferon stimulated genes in West Nile virus Eg101-infected mouse cells. Virology. 2012;425(2):82-94
  212. 212. Sariol CA, Muñoz-Jordán JL, Abel K, Rosado LC, Pantoja P, Giavedoni L, et al. Transcriptional activation of interferon-stimulated genes but not of cytokine genes after primary infection of rhesus macaques with dengue virus type 1. Clinical and Vaccine Immunology. 2007;14(6):756
  213. 213. Wu X, Dao Thi VL, Huang Y, Billerbeck E, Saha D, Hoffmann H-H, et al. Intrinsic immunity shapes viral resistance of stem cells. Cell. 2018;172(3):423-438.e25
  214. 214. Dai J, Pan W, Wang P. ISG15 facilitates cellular antiviral response to dengue and west nile virus infection in vitro. Virology Journal. 2011;8(1):468
  215. 215. Jiang D, Weidner JM, Qing M, Pan X-B, Guo H, Xu C, et al. Identification of five interferon-induced cellular proteins that inhibit West Nile virus and dengue virus infections. Journal of Virology. 2010;84(16):8332
  216. 216. Wang K, Zou C, Wang X, Huang C, Feng T, Pan W, et al. Interferon-stimulated TRIM69 interrupts dengue virus replication by ubiquitinating viral nonstructural protein 3. PLOS Pathogens. 2018;14(8):e1007287-e
  217. 217. Hertzog PJ, O’Neill LA, Hamilton JA. The interferon in TLR signaling: More than just antiviral. Trends in Immunology. 2003;24(10):534-539
  218. 218. Chen H-W, King K, Tu J, Sanchez M, Luster AD, Shresta S. The roles of IRF-3 and IRF-7 in innate antiviral immunity against dengue virus. Journal of Immunology (Baltimore, Md. : 1950). 2013;191(8):4194-4201
  219. 219. Carlin AF, Plummer EM, Vizcarra EA, Sheets N, Joo Y, Tang W, 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(6):1600-1612
  220. 220. 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(3):e00553-15
  221. 221. Modhiran N, Kalayanarooj S, Ubol S. Subversion of innate defenses by the interplay between DENV and pre-existing enhancing antibodies: TLRs signaling collapse. PLOS Neglected Tropical Diseases. 2010;4(12):e924
  222. 222. Munoz-Jordan JL. Subversion of interferon by dengue virus. Current Topics in Microbiology and Immunology. 2010;338:35-44
  223. 223. Kurane I, Innis BL, Nimmannitya S, Nisalak A, Meager A, Ennis FA. High levels of interferon alpha in the sera of children with dengue virus infection. The American Journal of Tropical Medicine and Hygiene. 1993;48(2):222-229
  224. 224. Chakravarti A, Kumaria R. Circulating levels of tumour necrosis factor-alpha & interferon-gamma in patients with dengue & dengue haemorrhagic fever during an outbreak. The Indian Journal of Medical Research. 2006;123(1):25-30
  225. 225. Nguyen TH, Lei HY, Nguyen TL, Lin YS, Huang KJ, Le BL, et al. Dengue hemorrhagic fever in infants: A study of clinical and cytokine profiles. The Journal of Infectious Diseases. 2004;189(2):221-232
  226. 226. De La Cruz Hernandez SI, Puerta-Guardo H, Flores-Aguilar H, Gonzalez-Mateos S, Lopez-Martinez I, Ortiz-Navarrete V, et al. A strong interferon response correlates with a milder dengue clinical condition. Journal of Clinical Virology. 2014;60(3):196-199
  227. 227. Fink J, Gu F, Ling L, Tolfvenstam T, Olfat F, Chin KC, et al. Host gene expression profiling of dengue virus infection in cell lines and patients. PLOS Neglected Tropical Diseases. 2007;1(2):e86
  228. 228. Simmons CP, Popper S, Dolocek C, Chau TN, Griffiths M, Dung NT, et al. Patterns of host genome-wide gene transcript abundance in the peripheral blood of patients with acute dengue hemorrhagic fever. The Journal of Infectious Diseases. 2007;195(8):1097-1107
  229. 229. Nascimento EJ, Braga-Neto U, Calzavara-Silva CE, Gomes AL, Abath FG, Brito CA, et al. Gene expression profiling during early acute febrile stage of dengue infection can predict the disease outcome. PLOS One. 2009;4(11):e7892
  230. 230. Ashour J, Laurent-Rolle M, Shi PY, Garcia-Sastre A. NS5 of dengue virus mediates STAT2 binding and degradation. Journal of Virology. 2009;83(11):5408-5418
  231. 231. Rodriguez-Madoz JR, Belicha-Villanueva A, Bernal-Rubio D, Ashour J, Ayllon J, Fernandez-Sesma A. Inhibition of the type I interferon response in human dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex. Journal of Virology. 2010;84(19):9760-9774
  232. 232. Muñoz-Jordán JL, Laurent-Rolle M, Ashour J, Martínez-Sobrido L, Ashok M, Lipkin WI, et al. Inhibition of alpha/beta interferon signaling by the NS4B protein of flaviviruses. Journal of Virology. 2005;79(13):8004
  233. 233. Mazzon M, Jones M, Davidson A, Chain B, Jacobs M. Dengue virus NS5 inhibits interferon-alpha signaling by blocking signal transducer and activator of transcription 2 phosphorylation. The Journal of Infectious Diseases. 2009;200(8):1261-1270
  234. 234. Shresta S, Sharar KL, Prigozhin DM, Snider HM, Beatty PR, Harris E. Critical roles for both STAT1-dependent and STAT1-independent pathways in the control of primary dengue virus infection in mice. Journal of Immunology. 2005;175(6):3946-3954
  235. 235. Aguirre S, Luthra P, Sanchez-Aparicio MT, Maestre AM, Patel J, Lamothe F, et al. Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection. Nature Microbiology. 2017;2:17037
  236. 236. Aguirre S, Maestre AM, Pagni S, Patel JR, Savage T, Gutman D, et al. DENV inhibits type I IFN production in infected cells by cleaving human STING. PLOS Pathogens. 2012;8(10):e1002934
  237. 237. Stabell AC, Meyerson NR, Gullberg RC, Gilchrist AR, Webb KJ, Old WM, et al. Dengue viruses cleave STING in humans but not in nonhuman primates, their presumed natural reservoir. eLife. 2018;7:e31919
  238. 238. Diosa-Toro M, Prasanth KR, Bradrick SS, Garcia Blanco MA. Role of RNA-binding proteins during the late stages of flavivirus replication cycle. Virology Journal. 2020;17(1):60
  239. 239. Alvarez DE, De Lella Ezcurra AL, Fucito S, Gamarnik AV. Role of RNA structures present at the 3’UTR of dengue virus on translation, RNA synthesis, and viral replication. Virology. 2005;339(2):200-212
  240. 240. Manokaran G, Finol E, Wang C, Gunaratne J, Bahl J, Ong EZ, et al. Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness. Science. 2015;350(6257):217-221
  241. 241. Pompon J, Manuel M, Ng GK, Wong B, Shan C, Manokaran G, et al. Dengue subgenomic flaviviral RNA disrupts immunity in mosquito salivary glands to increase virus transmission. PLOS Pathogens. 2017;13(7):e1006535
  242. 242. Pena J, Harris E. Early dengue virus protein synthesis induces extensive rearrangement of the endoplasmic reticulum independent of the UPR and SREBP-2 pathway. PLOS One. 2012;7(6):e38202
  243. 243. Umareddy I, Pluquet O, Wang QY, Vasudevan SG, Chevet E, Gu F. Dengue virus serotype infection specifies the activation of the unfolded protein response. Virology Journal. 2007;4:91
  244. 244. Perera N, Miller JL, Zitzmann N. The role of the unfolded protein response in dengue virus pathogenesis. Cellular Microbiology. 2017;19(5):e12734
  245. 245. Lee YR, Kuo SH, Lin CY, Fu PJ, Lin YS, Yeh TM, et al. Dengue virus-induced ER stress is required for autophagy activation, viral replication, and pathogenesis both in vitro and in vivo. Scientific Reports. 2018;8(1):489
  246. 246. Ke PY. The multifaceted roles of autophagy in Flavivirus-host interactions. International Journal of Molecular Sciences. 2018;19(12):3940
  247. 247. Heaton NS, Randall G. Dengue virus-induced autophagy regulates lipid metabolism. Cell Host & Microbe. 2010;8(5):422-432
  248. 248. Mizushima N. Autophagy: Process and function. Genes & Development. 2007;21(22):2861-2873
  249. 249. Dong X, Levine B. Autophagy and viruses: Adversaries or allies? Journal of Innate Immunity. 2013;5(5):480-493
  250. 250. Kuballa P, Nolte WM, Castoreno AB, Xavier RJ. Autophagy and the immune system. Annual Review of Immunology. 2012;30:611-646
  251. 251. Mateo R, Nagamine CM, Spagnolo J, Méndez E, Rahe M, Gale M, et al. Inhibition of cellular autophagy deranges dengue virion maturation. Journal of Virology. 2013;87(3):1312
  252. 252. Heaton NS, Randall G. Dengue virus and autophagy. Viruses. 2011;3(8):1332-1341
  253. 253. Panyasrivanit M, Khakpoor A, Wikan N, Smith DR. Linking dengue virus entry and translation/replication through amphisomes. Autophagy. 2009;5(3):434-435
  254. 254. Metz P, Chiramel A, Chatel-Chaix L, Alvisi G, Bankhead P, Mora-Rodríguez R, et al. Dengue virus inhibition of autophagic flux and dependency of viral replication on proteasomal degradation of the autophagy receptor p62. Journal of Virology. 2015;89(15):8026
  255. 255. Acharya B, Gyeltshen S, Chaijaroenkul W, Na-Bangchang K. Significance of autophagy in dengue virus infection: A brief review. The American Journal of Tropical Medicine and Hygiene. 2019;100(4):783-790
  256. 256. Heaton NS, Perera R, Berger KL, Khadka S, Lacount DJ, Kuhn RJ, 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 of the United States of America. 2010;107(40):17345-17350
  257. 257. Samsa MM, Mondotte JA, Iglesias NG, Assuncao-Miranda I, Barbosa-Lima G, Da Poian AT, et al. Dengue virus capsid protein usurps lipid droplets for viral particle formation. PLOS Pathogens. 2009;5(10):e1000632
  258. 258. Melo CFOR, Delafiori J, Dabaja MZ, de Oliveira DN, Guerreiro TM, Colombo TE, et al. The role of lipids in the inception, maintenance and complications of dengue virus infection. Scientific Reports. 2018;8(1):11826
  259. 259. Perera R, Riley C, Isaac G, Hopf-Jannasch AS, Moore RJ, Weitz KW, et al. Dengue virus infection perturbs lipid homeostasis in infected mosquito cells. PLOS Pathogens. 2012;8(3):e1002584-e
  260. 260. Leier HC, Messer WB, Tafesse FG. Lipids and pathogenic flaviviruses: An intimate union. PLOS Pathogens. 2018;14(5):e1006952
  261. 261. Randall G. Lipid droplet metabolism during dengue virus infection. Trends in Microbiology. 2018;26(8):640-642
  262. 262. Osuna-Ramos JF, Reyes-Ruiz JM, Del Angel RM. The role of host cholesterol during Flavivirus infection. Frontiers in Cellular and Infection Microbiology. 2018;8:388
  263. 263. Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, Angel-Ambrocio AH, del Angel RM. DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target. PLOS Pathogens. 2017;13(4):e1006257
  264. 264. Duran A, Carrero R, Parra B, Gonzalez A, Delgado L, Mosquera J, et al. Association of lipid profile alterations with severe forms of dengue in humans. Archives of Virology. 2015;160(7):1687-1692
  265. 265. Lima WG, Souza NA, Fernandes SOA, Cardoso VN, Godoi IP. Serum lipid profile as a predictor of dengue severity: A systematic review and meta-analysis. Reviews in Medical Virology. 2019;29(5):e2056
  266. 266. van Gorp EC, Suharti C, Mairuhu AT, Dolmans WM, van Der Ven J, Demacker PN, et al. Changes in the plasma lipid profile as a potential predictor of clinical outcome in dengue hemorrhagic fever. Clinical Infectious Diseases. 2002;34(8):1150-1153
  267. 267. Biswas HH, Gordon A, Nuñez A, Perez MA, Balmaseda A, Harris E. Lower low-density lipoprotein cholesterol levels are associated with severe dengue outcome. PLOS Neglected Tropical Diseases. 2015;9(9):e0003904-e
  268. 268. Conde JN, da Silva EM, Allonso D, Coelho DR, Andrade IDS, de Medeiros LN, et al. Inhibition of the membrane attack complex by dengue virus NS1 through interaction with vitronectin and terminal complement proteins. Journal of Virology. 2016;90(21):9570-9581
  269. 269. Avirutnan P, Hauhart RE, Somnuke P, Blom AM, Diamond MS, Atkinson JP. Binding of Flavivirus nonstructural protein NS1 to C4b binding protein modulates complement activation. The Journal of Immunology. 2011;187(1):424
  270. 270. Chung KM, Liszewski MK, Nybakken G, Davis AE, Townsend RR, Fremont DH, et al. West Nile virus nonstructural protein NS1 inhibits complement activation by binding the regulatory protein factor H. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(50):19111-19116
  271. 271. Schlesinger JJ. Flavivirus nonstructural protein NS1: Complementary surprises. Proceedings of the National Academy of Sciences. 2006;103(50):18879
  272. 272. Thiemmeca S, Tamdet C, Punyadee N, Prommool T, Songjaeng A, Noisakran S, et al. Secreted NS1 protects dengue virus from mannose-binding Lectin–mediated neutralization. The Journal of Immunology. 2016;197(10):4053

Written By

Henry Puerta-Guardo, Scott B. Biering, Eva Harris, Norma Pavia-Ruz, Gonzalo Vázquez-Prokopec, Guadalupe Ayora-Talavera and Pablo Manrique-Saide

Submitted: 11 May 2020 Reviewed: 09 June 2020 Published: 12 September 2020